VolumeVolume 14 1 - -Number Number 12 1 MayDecember - September 2010 1997

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Scope

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

Editorial correspondance

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

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

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

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

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

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

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Editor

Jean-Loup Huret (Poitiers, France) Editorial Board

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

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Volume 14, Number 12, December 2010

Table of contents

Gene Section

CKS2 (CDC28 protein kinase regulatory subunit 2) 1104 Yongyou Zhang CRTC2 (CREB regulated transcription coactivator 2) 1108 Kristy A Brown, Nirukshi Samarageewa IL22RA1 (interleukin 22 receptor, alpha 1) 1110 Pascal Gelebart, Raymond Lai MAPK7 (mitogen-activated protein kinase 7) 1114 Francisco de Asís Iñesta-Vaquera, Ana Cuenda SLC16A1 (solute carrier family 16, member 1 (monocarboxylic acid transporter 1)) 1118 Céline Pinheiro, Fátima Baltazar STOML2 (stomatin (EPB72)-like 2) 1121 Wenfeng Cao, Liyong Zhang, Fang Ding, Zhumei Cui, Zhihua Liu AMOT (angiomotin) 1124 Roshan Mandrawalia, Ranjan Tamuli BRCA2 (breast cancer 2, early onset) 1127 Frédéric Guénard, Francine Durocher FST (follistatin) 1135 Michael Grusch GATA6 (GATA binding protein 6) 1139 Rosalyn M Adam, Joshua Mauney HIPK2 (homeodomain interacting protein kinase 2) 1144 Dirk Sombroek, Thomas G Hofmann RAD9A (RAD9 homolog A (S. pombe)) 1148 Vivian Chan SCAF1 (SR-related CTD-associated factor 1) 1152 Christos Kontos, Andreas Scorilas SIRT1 (sirtuin (silent mating type information regulation 2 homolog) 1 (S. cerevisiae)) 1155 Ruo-Chia Tseng, Yi-Ching Wang SLC16A3 (solute carrier family 16, member 3 (monocarboxylic acid transporter 4)) 1160 Céline Pinheiro, Fátima Baltazar SPAM1 (sperm adhesion molecule 1 (PH-20 hyaluronidase, zona pellucida binding)) 1163 Asli Sade, Sreeparna Banerjee TMPRSS2 (transmembrane protease, serine 2) 1166 Youngwoo Park TMSB10 ( beta 10) 1169 Xueshan Qiu

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) Atlast(11;14)(q 13;q32)of Genetics in multiple myeloma and Cytogenetics Huret JL, Laï JL in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

TYMP (thymidine phosphorylase) 1173 Irene V Bijnsdorp, Godefridus J Peters

Leukaemia Section der(6)t(1;6)(q21-23;p21) 1178 Adriana Zamecnikova ins(9;4)(q33;q12q25) 1180 Jean-Loup Huret

Solid Tumour Section t(19;22)(q13;q12) in myoepithelial carcinoma 1182 Jean-Loup Huret

Deep Insight Section

Glutathione S-Transferase pi (GSTP1) 1184 Isabelle Meiers The roles of SRA1 in breast cancer 1190 Yi Yan, Charlton Cooper, Etienne Leygue

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

CKS2 (CDC28 protein kinase regulatory subunit 2) Yongyou Zhang Case Western Reserve University, WRB-3101, 2103 Cornell Rd, Cleveland, OH 44106, USA (YZ)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/CKS2ID40093ch9q22.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI CKS2ID40093ch9q22.txt

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 Description The open reading frame encodes a 79 amino acid Other names: CKSHS2 protein, with an estimated molecular weight of HGNC (Hugo): CKS2 approximately 9860 Da. Location: 9q22.2 Expression Note: There is no evidence that CKS2 gene has Basic level expression in all mammalian cells and different transcript variant. aberrant expression in cancer cells. DNA/RNA Localisation Cytoplasm and nucleus. Function CKS2 protein binds to the catalytic subunit of the

Genomic organization of the CKS2 gene. cyclin-dependent kinases and is essential for their biological function of cell cycle control. Especially, Description CKS2 is required for the first metaphase/anaphase Three exons, spans approximately 5.5 kb of transition of mammalian meiosis. The mice ablated genomic DNA in the centromere-to-telomere of Cks2 are viable but sterile in both sexes. Sterility orientation. The translation initiation codon ATG is is due to failure of both male and female germ cells located in exon 1, and the stop codon in exon 3. to progress from the first meiotic metaphase to Transcription anaphase. In cancer cells, CKS2 may protect the cells from apoptosis. mRNA is 627 bp. Homology Pseudogene The CKS2 protein is evolutionary conserved. 1 processed, non-expressed, pseudogene in human Mammalian cells express two well-conserved CKS genome. members, like the human CKS2 and CKS1B proteins. CKS2 and CKS1B may have redundant Protein function in some context and have different Note functions in other context. The CKS2 protein is highly conserved cross species. The Cks2 protein can form a special homohexamer structure. Six kinase subunits can bind the assembled hexamer, and therefore this Cks2 Mutations hexamer may participate in cell cycle control by Note acting as the hub for Cdk multimerization in vivo. Mutation of glutamine for glutamate 63 (E63Q),

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1104

CKS2 (CDC28 protein kinase regulatory subunit 2) Zhang Y

disrupted the essential biological function of the (Wiese et al., 2007). CKS2 was also showed to be protein and significantly reduced its ability to bind higher in metastasis compared with primary to cyclin-dependent kinases, but preserves protein colon cancer (Lin et al., 2007). structure and . Oncogenesis Amplification and overexpression of CKS2 were Implicated in associated with liver metastasis and poor prognosis Various cancers in colon cancer. CKS2 is required for germ cell to go past the first meiotic metaphase and enter Note anaphase. In cancer cell, overexpression of CKS2 Emerging evidence showed that the expression of can accelerate the cell cycles and promote the cell CKS2 is elevated in multiple cancers, including proliferation. Recently research showed that CKS2 , breast cancer, gastric cancer, may also be involved in apoptosis and metabolism colorectal cancer, uterine cervical cancer, bladder since it can protect mitochondrial genome integrity cancer, nasopharyngeal carcinoma, melanoma, via interaction with mitochondrial single-stranded lymphoma, lung cancer, esophageal squamous cell DNA-binding protein. Study also showed CKS2 as carcinoma et al. The expression of CKS2 is a transcriptional target downregulated by the tumor correlated with poor survival rate of the patients of suppressor p53. CKS2 expression was found to be some cancers. repressed by p53 both at the mRNA and the protein Prognosis levels, which may provide a mechanism that Overexpression of CKS2 has been reported to be explain why CKS2 is upregulated in many types of associated with high aggressiveness and a poor cancer. All of these suggest that CKS2 alterations prognosis in multiple cancers, including breast may have a significant biological role in the cancer, prostate cancer, colon cancer, hepatocellular tumorigenesis in different tissue. The novel carcinoma and meningiomas et al. therapeutic strategy for cancer though may be developed via inhibiting the CKS2 activity. Hepatocellular carcinoma (HCC) Therefore, disruption of CKS2-Cyclin Complex Note assembly or down-regulation of CKS2 expression Expressions of CKS2 were significantly higher in may be used for cancer therapy. HCC compared with the adjacent noncancerous Esophageal squamous cell tissues (including chronic hepatitis and cirrhosis) and normal liver tissues. Overexpression of CKS2 carcinoma in HCC were closely associated with poor Note differentiation features (Shen et al., 2010). profiling of lymph node metastasis Gastric cancer by oligomicroarray analysis and Real-time RT-PCR confirmed that CKS2 is unregulated in laser Note microdissection of esophageal squamous cell CKS2 was showed to be significantly unregulated carcinoma compared with adjacent normal tissue in gastric cancers. The high level of CKS2 was (Uchikado et al., 2006). highly correlated with tumor differentiation and pathological grade of the tumor size, lymph node, Uterine cervical cancer and metastasis stage (Kang et al., 2009). Note Prostate cancer CKS2 was showed significantly higher in node positive tumor compared with negative one. The Note CKS2 expression is correlated with metastatic CKS2 were significantly unregulated in prostate phenotypes and progression free survival. (Lyng et tumors of human and animal models, as well as al., 2006). prostatic cancer cell lines. Forced expression of CKS2 in benign prostate tumor epithelial cells Bladder cancer promoted cell population growth. Inhibition of Note CKS2 expression can induce programmed cell Large-scale gene expression profiling and Real- death and inhibit the tumorigenesis. (Lan et al., Time RT-PCR confirmed that a the CKS2 2008). Over expression of CKS2 may linked with expression is elevated in invasive bladder cancer androgen-independent prostate cancer progression compared with superficial cancer (Kawakami et al., (Stanbrough et al., 2006). 2006). Colon cancer Glioblastoma Note Note CKS2 was reported significantly overexpressed in CKS2 was significantly up-regulated in primary microdissected invasive colon tumor cells glioblastomas compared with the non-neoplastic compared with adjacent normal epithelial cells brain tissues (Scrideli et al., 2008).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1105

CKS2 (CDC28 protein kinase regulatory subunit 2) Zhang Y

Meningioma of RASSF1A modulated genes in nasopharyngeal carcinoma. Oncogene. 2006 Jan 12;25(2):310-6 Note Kawakami K, Enokida H, Tachiwada T, Gotanda T, This microarray-based expression profiling study Tsuneyoshi K, Kubo H, Nishiyama K, Takiguchi M, showed CKS2 is unregulated in atypical and Nakagawa M, Seki N. Identification of differentially anaplastic meningiomas compared with benign expressed genes in human bladder cancer through meningiomas (Fevre-Montange et al., 2009). genome-wide gene expression profiling. Oncol Rep. 2006 Sep;16(3):521-31 References Lyng H, Brøvig RS, Svendsrud DH, Holm R, Kaalhus O, Knutstad K, Oksefjell H, Sundfør K, Kristensen GB, Stokke Parge HE, Arvai AS, Murtari DJ, Reed SI, Tainer JA. T. Gene expressions and copy numbers associated with Human CksHs2 atomic structure: a role for its hexameric metastatic phenotypes of uterine cervical cancer. BMC assembly in cell cycle control. Science. 1993 Oct Genomics. 2006 Oct 20;7:268 15;262(5132):387-95 Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA, Demetrick DJ, Zhang H, Beach DH. Chromosomal Penning TM, Febbo PG, Balk SP. Increased expression of mapping of the human genes CKS1 to 8q21 and CKS2 to genes converting adrenal androgens to in 9q22. Cytogenet Cell Genet. 1996;73(3):250-4 androgen-independent prostate cancer. Cancer Res. 2006 Mar 1;66(5):2815-25 Pines J. Cell cycle: reaching for a role for the Cks proteins. Curr Biol. 1996 Nov 1;6(11):1399-402 Uchikado Y, Inoue H, Haraguchi N, Mimori K, Natsugoe S, Okumura H, Aikou T, Mori M. Gene expression profiling of Watson MH, Bourne Y, Arvai AS, Hickey MJ, Santiago A, lymph node metastasis by oligomicroarray analysis using Bernstein SL, Tainer JA, Reed SI. A mutation in the human laser microdissection in esophageal squamous cell cyclin-dependent kinase interacting protein, CksHs2, carcinoma. Int J Oncol. 2006 Dec;29(6):1337-47 interferes with cyclin-dependent kinase binding and biological function, but preserves protein structure and Wong YF, Cheung TH, Tsao GS, Lo KW, Yim SF, Wang assembly. J Mol Biol. 1996 Sep 6;261(5):646-57 VW, Heung MM, Chan SC, Chan LK, Ho TW, Wong KW, Li C, Guo Y, Chung TK, Smith DI. Genome-wide gene Egan EA, Solomon MJ. Cyclin-stimulated binding of Cks expression profiling of cervical cancer in Hong Kong proteins to cyclin-dependent kinases. Mol Cell Biol. 1998 women by oligonucleotide microarray. Int J Cancer. 2006 Jul;18(7):3659-67 May 15;118(10):2461-9 Urbanowicz-Kachnowicz I, Baghdassarian N, Nakache C, Lin CY, Ström A, Li Kong S, Kietz S, Thomsen JS, Tee JB, Gracia D, Mekki Y, Bryon PA, Ffrench M. ckshs expression Vega VB, Miller LD, Smeds J, Bergh J, Gustafsson JA, Liu is linked to cell proliferation in normal and malignant ET. Inhibitory effects of receptor beta on specific human lymphoid cells. Int J Cancer. 1999 Jul 2;82(1):98- -responsive gene expression and association with 104 disease outcome in primary breast cancer. Breast Cancer Res. 2007;9(2):R25 Seeliger MA, Schymkowitz JW, Rousseau F, Wilkinson HR, Itzhaki LS. Folding and association of the human cell Lin HM, Chatterjee A, Lin YH, Anjomshoaa A, Fukuzawa cycle regulatory proteins ckshs1 and ckshs2. Biochemistry. R, McCall JL, Reeve AE. Genome wide expression 2002 Jan 29;41(4):1202-10 profiling identifies genes associated with colorectal liver metastasis. Oncol Rep. 2007 Jun;17(6):1541-9 Donovan PJ, Reed SI. Germline exclusion of Cks1 in the mouse reveals a metaphase I role for Cks proteins in male Rother K, Dengl M, Lorenz J, Tschöp K, Kirschner R, and female meiosis. Cell Cycle. 2003 Jul-Aug;2(4):275-6 Mössner J, Engeland K. Gene expression of cyclin- dependent kinase subunit Cks2 is repressed by the tumor Lu X, Guo J, Hsieh TC. PC-SPES inhibits cell proliferation suppressor p53 but not by the related proteins p63 or p73. by modulating p21, cyclins D, E and B and multiple cell FEBS Lett. 2007 Mar 20;581(6):1166-72 cycle-related genes in prostate cancer cells. Cell Cycle. 2003 Jan-Feb;2(1):59-63 Wiese AH, Auer J, Lassmann S, Nährig J, Rosenberg R, Höfler H, Rüger R, Werner M. Identification of gene Seeliger MA, Breward SE, Itzhaki LS. Weak cooperativity signatures for invasive colorectal tumor cells. Cancer in the core causes a switch in folding mechanism between Detect Prev. 2007;31(4):282-95 two proteins of the cks family. J Mol Biol. 2003 Jan 3;325(1):189-99 Haaber J, Abildgaard N, Knudsen LM, Dahl IM, Lodahl M, Thomassen M, Kerndrup GB, Rasmussen T. Myeloma cell Spruck CH, de Miguel MP, Smith AP, Ryan A, Stein P, expression of 10 candidate genes for osteolytic bone Schultz RM, Lincoln AJ, Donovan PJ, Reed SI. disease. Only overexpression of DKK1 correlates with Requirement of Cks2 for the first metaphase/anaphase clinical bone involvement at diagnosis. Br J Haematol. transition of mammalian meiosis. Science. 2003 Apr 2008 Jan;140(1):25-35 25;300(5619):647-50 Lan Y, Zhang Y, Wang J, Lin C, Ittmann MM, Wang F. Li M, Lin YM, Hasegawa S, Shimokawa T, Murata K, Aberrant expression of Cks1 and Cks2 contributes to Kameyama M, Ishikawa O, Katagiri T, Tsunoda T, prostate tumorigenesis by promoting proliferation and Nakamura Y, Furukawa Y. Genes associated with liver inhibiting programmed cell death. Int J Cancer. 2008 Aug metastasis of colon cancer, identified by genome-wide 1;123(3):543-51 cDNA microarray. Int J Oncol. 2004 Feb;24(2):305-12 Martinsson-Ahlzén HS, Liberal V, Grünenfelder B, Chaves de Wit NJ, Rijntjes J, Diepstra JH, van Kuppevelt TH, SR, Spruck CH, Reed SI. Cyclin-dependent kinase- Weidle UH, Ruiter DJ, van Muijen GN. Analysis of associated proteins Cks1 and Cks2 are essential during differential gene expression in human melanocytic tumour early embryogenesis and for cell cycle progression in lesions by custom made oligonucleotide arrays. Br J somatic cells. Mol Cell Biol. 2008 Sep;28(18):5698-709 Cancer. 2005 Jun 20;92(12):2249-61 Scrideli CA, Carlotti CG Jr, Okamoto OK, Andrade VS, Chow LS, Lam CW, Chan SY, Tsao SW, To KF, Tong SF, Cortez MA, Motta FJ, Lucio-Eterovic AK, Neder L, Hung WK, Dammann R, Huang DP, Lo KW. Identification Rosemberg S, Oba-Shinjo SM, Marie SK, Tone LG. Gene

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1106

CKS2 (CDC28 protein kinase regulatory subunit 2) Zhang Y

expression profile analysis of primary glioblastomas and Miller WR. Clinical, pathological, proliferative and non-neoplastic brain tissue: identification of potential target molecular responses associated with neoadjuvant genes by oligonucleotide microarray and real-time aromatase inhibitor treatment in breast cancer. J Steroid quantitative PCR. J Neurooncol. 2008 Jul;88(3):281-91 Biochem Mol Biol. 2010 Feb 28;118(4-5):273-6 Fèvre-Montange M, Champier J, Durand A, Wierinckx A, Radulovic M, Crane E, Crawford M, Godovac- Honnorat J, Guyotat J, Jouvet A. Microarray gene Zimmermann J, Yu VP. CKS proteins protect mitochondrial expression profiling in meningiomas: differential genome integrity by interacting with mitochondrial single- expression according to grade or histopathological stranded DNA-binding protein. Mol Cell Proteomics. 2010 subtype. Int J Oncol. 2009 Dec;35(6):1395-407 Jan;9(1):145-52 Kang MA, Kim JT, Kim JH, Kim SY, Kim YH, Yeom YI, Lee Shen DY, Fang ZX, You P, Liu PG, Wang F, Huang CL, Y, Lee HG. Upregulation of the cycline kinase subunit Yao XB, Chen ZX, Zhang ZY. Clinical significance and CKS2 increases cell proliferation rate in gastric cancer. J expression of cyclin kinase subunits 1 and 2 in Cancer Res Clin Oncol. 2009 Jun;135(6):761-9 hepatocellular carcinoma. Liver Int. 2010 Jan;30(1):119-25

Wang F, Kuang Y, Salem N, Anderson PW, Lee Z. Cross- This article should be referenced as such: species hybridization of woodchuck hepatitis viral infection- induced woodchuck hepatocellular carcinoma using Zhang Y. CKS2 (CDC28 protein kinase regulatory subunit human, rat and mouse oligonucleotide microarrays. J 2). Atlas Genet Cytogenet Oncol Haematol. 2010; Gastroenterol Hepatol. 2009 Apr;24(4):605-17 14(12):1104-1107.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1107

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

CRTC2 (CREB regulated transcription coactivator 2) Kristy A Brown, Nirukshi Samarageewa Prince Henry's Institute, Clayton, Victoria, 3168, Australia (KAB, NS)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/CRTC2ID50581ch1q21.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI CRTC2ID50581ch1q21.txt

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

activated protein kinase (AMPK) and the salt- Identity inducible kinases (SIKs). Dephosphorylated Other names: TORC2, RP11-422P24.6 CRTC2 readily translocates to the nucleus. CRTC2 HGNC (Hugo): CRTC2 contains a nuclear localisation sequence (NLS) at amino acids 56-144 as well as two nuclear export Location: 1q21.3 sequences (NES1 and NES2) within the region of DNA/RNA amino acids 145-320. Function Description Transcriptional coactivator for CREB (cAMP- 10,893 bases; on minus strand. responsive element binding protein).The highly Includes 14 exons. conserved N-terminal coiled-coil domain of the Transcription CRTC2 interacts with the bZip domain of CREB which activates both consensus and variant cAMP Transcript measures 2598 bp with a 2082 bp coding response element (CRE) sites, leading to activation sequence. of CREB target gene expression. CRTC2 responds to stimulation by cAMP, calcium, fasting Protein , G protein-coupled receptors, and Description AMPK/SIKs. 693 amino acids; 73,302 Da. Implicated in Expression Peutz-Jeghers syndrome Particularly abundant in B and T lymphocytes. Note Higher levels were also seen in muscle, lung, spleen, and breast. Lower expressions found Peutz-Jeghers syndrome (PJS) is an autosomal- in brain, colon, , , prostate, small dominant genetic disorder that is characterised by intestine and , with significantly lowest an increased risk of developing malignant tumours. expression in liver and . Most of the identified mutations in the LKB1 gene are localised to the catalytic kinase domain so that it Localisation is thought that PJS results from loss of LKB1 Phosphorylation of CRTC2 triggers the kinase activity. The silencing of LKB1, leads to the phosphorylation-dependent binding to 14-3-3 decreased activity of AMPK and SIK and leads to proteins, and hence sequestration of CRTC2 in the the increased nuclear translocation and activity of cytosol thereby preventing its nuclear translocation CRTC2. and the activation of CREB. Proteins known to Disease phosphorylate CRTC2 at Ser171 include AMP- Gastrointestinal polyps and cancers including

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1108

CRTC2 (CREB regulated transcription coactivator 2) Brown KA, Samarageewa N

esophagus, stomach, small intestine, colon, Sofi M, Young MJ, Papamakarios T, Simpson ER, Clyne pancreas, lung, testes, breast, uterus, ovary and CD. Role of CRE-binding protein (CREB) in aromatase expression in breast adipose. Breast Cancer Res Treat. cervix. 2003 Jun;79(3):399-407 Oestrogen-receptor (ER) positive Screaton RA, Conkright MD, Katoh Y, Best JL, Canettieri breast cancer G, Jeffries S, Guzman E, Niessen S, Yates JR 3rd, Takemori H, Okamoto M, Montminy M. The CREB Note coactivator TORC2 functions as a calcium- and cAMP- The increased prevalence of oestrogen-dependent, sensitive coincidence detector. Cell. 2004 Oct 1;119(1):61- postmenopausal breast cancers is correlated with 74 elevated local levels of oestrogens as a result of an Alessi DR, Sakamoto K, Bayascas JR. LKB1-dependent increase in cytochrome P450 aromatase expression signaling pathways. Annu Rev Biochem. 2006;75:137-63 within the adipose stromal (hAS) cells surrounding Katoh Y, Takemori H, Lin XZ, Tamura M, Muraoka M, the breast tumour - aromatase is the enzyme Satoh T, Tsuchiya Y, Min L, Doi J, Miyauchi A, Witters LA, responsible for the conversion of androgens to Nakamura H, Okamoto M. Silencing the constitutive active transcription factor CREB by the LKB1-SIK signaling oestrogens. This is governed by promoter switching cascade. FEBS J. 2006 Jun;273(12):2730-48 from the distal promoter I.4 to the proximal Shaw RJ. Glucose metabolism and cancer. Curr Opin Cell promoter PII on the CYP19A1 gene, that encodes Biol. 2006 Dec;18(6):598-608 aromatase, in response to factors derived from the tumour such as E2 (PGE2). Wu Z, Huang X, Feng Y, Handschin C, Feng Y, Gullicksen PS, Bare O, Labow M, Spiegelman B, Stevenson SC. Interestingly, the LKB1/ AMPK pathway has been Transducer of regulated CREB-binding proteins (TORCs) shown to inhibit aromatase expression via the induce PGC-1alpha transcription and mitochondrial cytoplasmic sequestration of CRTC2. However, biogenesis in muscle cells. Proc Natl Acad Sci U S A. 2006 PGE2 inhibits LKB1/AMPK signaling, leading to Sep 26;103(39):14379-84 the nuclear translocation of CRTC2 and its Brown KA, McInnes KJ, Hunger NI, Oakhill JS, Steinberg enhanced binding and activation of aromatase GR, Simpson ER. Subcellular localization of cyclic AMP- responsive element binding protein-regulated transcription promoter PII in hAS cells. Furthermore, the coactivator 2 provides a link between obesity and breast adipokine , produced at higher levels in cancer in postmenopausal women. Cancer Res. 2009 Jul obesity, has been shown to cause an increase in 1;69(13):5392-9 CRTC2 nuclear translocation and consequently, in Brown KA, Simpson ER. Obesity and breast cancer: aromatase expression. progress to understanding the relationship. Cancer Res. 2010 Jan 1;70(1):4-7 References This article should be referenced as such: Conkright MD, Canettieri G, Screaton R, Guzman E, Brown KA, Samarageewa N. CRTC2 (CREB regulated Miraglia L, Hogenesch JB, Montminy M. TORCs: transcription coactivator 2). Atlas Genet Cytogenet Oncol transducers of regulated CREB activity. Mol Cell. 2003 Haematol. 2010; 14(12):1108-1109. Aug;12(2):413-23

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1109

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

IL22RA1 (interleukin 22 receptor, alpha 1) Pascal Gelebart, Raymond Lai Department of Laboratory Medicine and Pathology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada (PG, RL)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/IL22RA1ID44568ch1p36.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI IL22RA1ID44568ch1p36.txt

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

nucleotides, encoding a protein of 594 amino acid Identity residues. Other names: CRF2-9, IL22R, IL22R1 Pseudogene HGNC (Hugo): IL22RA1 None. Location: 1p36.11 Protein DNA/RNA Description Description IL22RA1 is composed of 574 amino acid residues, The gene spans a region of 23.3 kb including seven and the predicted molecular weight of the immature exons. protein is 63 kDa. IL22RA1 protein is composed of six putative domains, including the signal Transcription (residue 1 to 15), the extracellular domain (residue One only transcript form containing 7 exons has 16 to 228), the transmembrane domain (residue 229 been described. to 249), the cytoplasmic domain (residue 250 to The last exon is partially untranslated. The 574), and two fibronectin type-III domains (residue transcript length is 1725 18-115 and 141-221).

Representation of the IL22RA1 gene organization. IL22RA1 gene and RNA.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1110

IL22RA1 (interleukin 22 receptor, alpha 1) Gelebart P, Lai R

IL22RA1 protein organization and localization. IL22RA1 protein domains.

Localization of IL22RA1 by immunufluorescence confocal microscopy in ALK+ALCL cells.

IL22RA1 is not detectable in normal immune cells, including monocytes, B-cells, T-cells, natural killer cells, macrophages and dendritic cells, cell types that are normally found in the bone marrow, peripheral blood, and spleen.

Crystal structure of IL22RA1 with IL22 at 1.9 A resolution. Adapted from PDB (access number: 3DLQ). FACS analysis of IL22RA1 expression in peripheral Expression mononuclear cells from healthy donor. IL22RA1 expression is relatively restricted, being found at the highest level in the pancreas, small Localisation intestine, colon, kidney, and liver. Importantly, IL22RA1 is localized at the plasma membrane.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1111

IL22RA1 (interleukin 22 receptor, alpha 1) Gelebart P, Lai R

IL22RA1 signaling.

Function Disease IL22RA1 is one of the subunits of the IL20, IL22 Anaplastic lymphoma kinase (ALK)-positive and IL24 receptor complex. Cytokine binding to anaplastic large-cell lymphoma (ALCL), or IL22RA1 results in its aggregation, which activates ALK+ALCL, is a specific type of non-Hodgkin the associated JAK via its autophosphorylation. lymphoma characterized by the T/null-cell This in turn leads to the phosphorylation and immunophenotype, consistent expression of CD30 activation of STAT proteins. Subsequently, and reciprocal chromosomal translocations phosphorylated STAT proteins dimerize and involving the ALK gene. In most cases, the translocate to the nucleus to modulate the chromosomal translocation is that of the transcription of various target genes. t(2;5)(p23;q35), which leads to the juxtaposition of the nucleophosmin () gene at 5q35 with the Mutations ALK gene at 2p23. Mounting evidence suggests that the resulted oncogenic fusion protein, NPM- Site-directed mutagenesis experiments have ALK, plays crucial roles in the pathogenesis of revealed critical amino acid residues involved in its these tumors. binding to IL22. Specifically, mutation of residue Prognosis 58 from K to A reduces the binding of IL22. Patients with ALK+ALCL are typically treated with Mutation of the residue 60 from Y to A or R results combination chemotherapy containing doxorubicin. in a complete loss of response to IL22. ALK+ALCL represents one of the most common Natural IL22RA1 variants have been reported, pediatric lymphoid malignancies. The prognosis of including those carrying mutations at the residue pediatric ALK+ALCL patients is significant better 130 (S to P), 205 (V to I), 209 (A to S), 222 (L to than that of adult patients. P), 407 (M to V) and 518 (R to G). Cytogenetics Implicated in t(2;5)(p23;q35) in most ALK+ALCL patients; other translocation variants have been described. ALK-positive anaplastic large cell Hybrid/Mutated gene lymphoma (ALK+ALCL) NPM-ALK

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1112

IL22RA1 (interleukin 22 receptor, alpha 1) Gelebart P, Lai R

Representation of the NPM-ALK oncoprotein organization and sequence.

Abnormal protein heterodimeric receptors, IL-22R1/IL-20R2 and IL-20R1/IL- 20R2. J Biol Chem. 2002 Mar 1;277(9):7341-7 NPM-ALK Dumoutier L, Lejeune D, Hor S, Fickenscher H, Renauld JC. Cloning of a new type II cytokine receptor activating signal transducer and activator of transcription (STAT)1, STAT2 and STAT3. Biochem J. 2003 Mar 1;370(Pt 2):391- 6 Structure of the oncogenic fusion protein NPM-ALK. Amin HM, Lai R. Pathobiology of ALK+ anaplastic large- Oncogenesis cell lymphoma. Blood. 2007 Oct 1;110(7):2259-67 Aberrant expression of IL22RA1 in ALK+ALCL Bard JD, Gelebart P, Anand M, Amin HM, Lai R. Aberrant lymphoma cells allows these cells to be responsive expression of IL-22 receptor 1 and autocrine IL-22 to IL-22 stimulation, which further stimulate stimulation contribute to tumorigenicity in ALK+ anaplastic large cell lymphoma. Leukemia. 2008 Aug;22(8):1595-603 STAT3 signaling and the growth of these cells. Blocking the IL-22 signaling pathway using a Bleicher L, de Moura PR, Watanabe L, Colau D, Dumoutier L, Renauld JC, Polikarpov I. Crystal structure of neutralizing antibody has been shown to the IL-22/IL-22R1 complex and its implications for the IL- significantly decrease the growth of ALK+ALCL 22 signaling mechanism. FEBS Lett. 2008 Sep cells in-vitro. The aberrant expression of IL22RA1 3;582(20):2985-92 in ALK+ALCL is dependent on the expression of de Oliveira Neto M, Ferreira JR Jr, Colau D, Fischer H, NPM-ALK, since siRNA to downregulate NPM- Nascimento AS, Craievich AF, Dumoutier L, Renauld JC, ALK dramatically shut down IL22RA1 expression. Polikarpov I. Interleukin-22 forms dimers that are recognized by two interleukin-22R1 receptor chains. References Biophys J. 2008 Mar 1;94(5):1754-65 Dumoutier L, de Meester C, Tavernier J, Renauld JC. New Kotenko SV, Izotova LS, Mirochnitchenko OV, Esterova E, activation modus of STAT3: a tyrosine-less region of the Dickensheets H, Donnelly RP, Pestka S. Identification of interleukin-22 receptor recruits STAT3 by interacting with the functional interleukin-22 (IL-22) receptor complex: the its coiled-coil domain. J Biol Chem. 2009 Sep IL-10R2 chain (IL-10Rbeta ) is a common chain of both the 25;284(39):26377-84 IL-10 and IL-22 (IL-10-related T cell-derived inducible factor, IL-TIF) receptor complexes. J Biol Chem. 2001 Jan Endam LM, Bossé Y, Filali-Mouhim A, Cormier C, Boisvert 26;276(4):2725-32 P, Boulet LP, Hudson TJ, Desrosiers M. Polymorphisms in the interleukin-22 receptor alpha-1 gene are associated Lécart S, Morel F, Noraz N, Pène J, Garcia M, Boniface K, with severe chronic rhinosinusitis. Otolaryngol Head Neck Lecron JC, Yssel H. IL-22, in contrast to IL-10, does not Surg. 2009 May;140(5):741-7 induce Ig production, due to absence of a functional IL-22 receptor on activated human B cells. Int Immunol. 2002 This article should be referenced as such: Nov;14(11):1351-6 Gelebart P, Lai R. IL22RA1 (interleukin 22 receptor, alpha Wang M, Tan Z, Zhang R, Kotenko SV, Liang P. 1). Atlas Genet Cytogenet Oncol Haematol. 2010; Interleukin 24 (MDA-7/MOB-5) signals through two 14(12):1110-1113.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1113

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

MAPK7 (mitogen-activated protein kinase 7) Francisco de Asís Iñesta-Vaquera, Ana Cuenda Centro Nacional de Biotecnologia-CSIC, Department of Immunology and Oncology, Madrid, Spain (FdAIV, AC)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/MAPK7ID41294ch17p11.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI MAPK7ID41294ch17p11.txt

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 (MAPK7a, b and c) have been reported. Mouse splice variants are generated by alternative splicing Other names: BMK1, ERK4, ERK5, PRKM7 across introns 1 and/or 2 (Yan et al., 2001). HGNC (Hugo): MAPK7 Pseudogene Location: 17p11.2 No human or mouse pseudogene known. DNA/RNA Protein Description Note The MAPK7 entire gene spans 5,82 kb on the short ERK5, also known as MAPK7 or "Big MAP- arm of 17. It contains 6 exons. Kinase 1" (BMK1) belongs to the Mitogen Transcription Activated Protein Kinase (MAPK) family, and therefore to the CGMC kinases in the human The human MAPK7 gene encodes an 816 amino- kinome (Manning et al., 2002). ERK5, at 98 kDa, is acids protein of about 98 kDa. MAPK7 mRNA is twice the size of other MAPKs and hence the 2445 bp. There are 11 transcripts, seven of which largest kinase within its group. are protein coding. In mice, three splice variants

MAPK7 genomic context (Chromosome 17; location 17p11.2).

Genomic organization of MAPK7 gene on chromosome 17p11.2. The boxes indicate coding regions (exons 1-6) of the gene.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1114

MAPK7 (mitogen-activated protein kinase 7) Iñesta-Vaquera FdA, Cuenda A

Schematic representation of the human ERK5 (MAPK7) protein domains. NES1 and NES2, bipartite nuclear exportation signal; PB1-BD, PB1 (Phox and Bem domain 1) binding domain; Kinase Domain, catalytic kinase domain; TEY, sequence motif containing ERK5 regulatory phosphorylation residues; PR-1 and PR-2, proline rich domains; Transcriptional trans-activation, transcriptional activity domain.

It possesses a catalytic N-terminal domain, which (Regan et al., 2002; Sohn et al., 2002; Yan et al., share 50% homology with ERK1 (MAPK3) and 2003; Hayashi et al., 2004; Wang et al., 2005). ERK2 (MAPK1) and a unique C-terminal tail of ERK5 also regulates cell survival in a variety of about 400 amino-acids long. In vivo, ERK5 is tissues. At nervous system, ERK5 acts as a activated to the same extent by environmental neuroprotector from neurotrophic factor withdrawal stresses, such as oxidative and osmotic shock, and and toxic insults (Cavanaugh, 2004). Also, ERK5 is by growth factors. In addition, ERK5 may be required to mediate the survival response of activated by the cytokine Interleukin-6 in B cells. neurons to (Finegan et al., Description 2009). In the immune system, the ERK5 pathway regulates apoptosis of developing thymocytes Human ERK5 (MAPK7) is a Ser/Thr protein kinase (Sohn et al., 2008) and protects B cells from of 816 amino-acids with a predicted mass of 98 proapoptotic stimuli (Carvajal-Vergara et al., 2005). kDa. The ERK5 N-terminus domain resembles the ERK5 is also required for cell cycle progression. It typical MAPK catalytic domain and includes the regulates cyclin D1 expression (Mulloy et al., 2003) MAPK-conserved TXY activation sequence 218 220 and is necessary for EGF-induced cell proliferation (T EY ) in the activation loop. The activation of and progression through the cell cycle (Kato et al., ERK5 occurs via interaction with and dual 1998). Moreover, it has been suggested that the phosphorylation in its TEY motif by MKK5 (Mody ERK5-NFKappaB pathway may be required for a et al., 2003). MKK5 mediated ERK5 activation timely mitotic entry (Cude et al., 2007). leads to ERK5 autophosphorylation in its unique C- Additionally, ERK5, along with other MAPK terminal domain (Morimoto et al., 2007). pathways can play an indirect role in cytoskeleton Expression rearrangement (Barros and Marshall, 2005), in promoting SRC-induced podosome formation ERK5 (MAPK7) mRNA is widely expressed (Schramp et al., 2008), and in cell attachment to the throughout all tissues. extracellular matrix and in endothelial cell Localisation migration (Spiering et al., 2009; Sawhney et al., Both in tissues and in cultured cells, ERK5 2009). (MAPK7) localizes to the cytoplasm of cells and/or ERK5 (MAPK7) is a protein with kinase activity to the nucleus. As shown in the above diagram, (in its N-terminal region) and also transcriptional ERK5 molecule contains a bipartite nuclear activation activity (in the C-terminal half). exportation signal. In resting cells, the N- and C- Downstream targets of ERK5 include the terminal halves of ERK5 interact producing a transcription factors MEF2A, MEF2C and MEF2D, nuclear export signal (NES) that retains ERK5 in SAP1a, c-Myc and CREB. For example, ERK5 the cytoplasm of the cells. Upon stimulation, the phosphorylates SAP1, which enhances its interaction between the N- and the C-terminal transcriptional activity promoting c-FOS expression halves is disrupted, and therefore ERK5 enters the (Terasawa et al., 2003), and activates the serum- nucleus (Kondoh et al., 2006). and glucocorticoid-inducible kinase1 (SGK1) by phosphorylating Ser78 in response to growth Function factors (Hayashi et al., 2001). In cardiac tissue, Genetic studies have shown that ERK5 (MAPK7) is ERK5 may couple cells electrically and essential for cardiovascular development and metabolically by phosphorylating the gap-junction neuronal differentiation. ERK5 knock-out mice die protein Cx43 at a key residue for gap junction at midgestation due to developmental failures in communication (Cameron et al., 2003). Also, structures as , heart and vascular system phosphorylated ERK5 regulates gene expression

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1115

MAPK7 (mitogen-activated protein kinase 7) Iñesta-Vaquera FdA, Cuenda A

through its C-terminal transcriptional activation Yan C, Luo H, Lee JD, Abe J, Berk BC. Molecular cloning domain (Morimoto et al., 2007). of mouse ERK5/BMK1 splice variants and characterization of ERK5 functional domains. J Biol Chem. 2001 Apr Homology 6;276(14):10870-8 ERK5 (MAPK7) N-terminal half shares a 50% Esparís-Ogando A, Díaz-Rodríguez E, Montero JC, Yuste sequence identity with ERK1/2. The homology of L, Crespo P, Pandiella A. Erk5 participates in neuregulin signal transduction and is constitutively active in breast the C-terminal part of ERK5 with other protein has cancer cells overexpressing ErbB2. Mol Cell Biol. 2002 not been reported. ERK5 possesses ortholog in the Jan;22(1):270-85 majority of mammals (sharing 80-98% homology). Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam In C. elegans, the SMA-5 protein is a 60% similar S. The protein kinase complement of the . to human ERK5 (Watanabe et al., 2005). In Science. 2002 Dec 6;298(5600):1912-34 Saccharomyces cerevisiae, Slt2p (Mpk1p) is an Regan CP, Li W, Boucher DM, Spatz S, Su MS, Kuida K. ERK5 ortholog (Truman et al., 2006). Erk5 null mice display multiple extraembryonic vascular and embryonic cardiovascular defects. Proc Natl Acad Sci Mutations U S A. 2002 Jul 9;99(14):9248-53 Sohn SJ, Sarvis BK, Cado D, Winoto A. ERK5 MAPK Note regulates embryonic angiogenesis and acts as a hypoxia- Not identified. sensitive repressor of vascular endothelial growth factor expression. J Biol Chem. 2002 Nov 8;277(45):43344-51 Implicated in Cameron SJ, Malik S, Akaike M, Lerner-Marmarosh N, Yan C, Lee JD, Abe J, Yang J. Regulation of epidermal Breast cancer growth factor-induced connexin 43 gap junction communication by big mitogen-activated protein Note kinase1/ERK5 but not ERK1/2 kinase activation. J Biol ERK5 (MAPK7) expression and activity is Chem. 2003 May 16;278(20):18682-8 increased in breast cancer tumours. ERK5 Mody N, Campbell DG, Morrice N, Peggie M, Cohen P. An overexpression has been established as an analysis of the phosphorylation and activation of independent predictor of disease-free survival in extracellular-signal-regulated protein kinase 5 (ERK5) by mitogen-activated protein kinase kinase 5 (MKK5) in vitro. breast cancer (Montero et al., 2009). In cell models, Biochem J. 2003 Jun 1;372(Pt 2):567-75 ERK5 has been linked to the regulation of breast cancer cells proliferation (Esparís-Ogando et al., Mulloy R, Salinas S, Philips A, Hipskind RA. Activation of cyclin D1 expression by the ERK5 cascade. Oncogene. 2002). 2003 Aug 21;22(35):5387-98 Prostatic cancer Terasawa K, Okazaki K, Nishida E. Regulation of c-Fos Note and Fra-1 by the MEK5-ERK5 pathway. Genes Cells. 2003 Mar;8(3):263-73 ERK5 (MAPK7) immunoreactivity is significantly up-regulated in high-grade prostate cancer. Yan L, Carr J, Ashby PR, Murry-Tait V, Thompson C, Arthur JS. Knockout of ERK5 causes multiple defects in Increased ERK5 cytoplasmic signals correlated placental and embryonic development. BMC Dev Biol. with metastases and locally advanced disease at 2003 Dec 16;3:11 diagnosis. Strong nuclear ERK5 localization in Cavanaugh JE. Role of extracellular signal regulated prostatic tumours correlates with poor disease- kinase 5 in neuronal survival. Eur J Biochem. 2004 specific survival (McCracken et al., 2008). Jun;271(11):2056-9 Hepatic carcinoma Hayashi M, Kim SW, Imanaka-Yoshida K, Yoshida T, Abel ED, Eliceiri B, Yang Y, Ulevitch RJ, Lee JD. Targeted Note deletion of BMK1/ERK5 in adult mice perturbs vascular An increase in ERK5 (MAPK7) copy number was integrity and leads to endothelial failure. J Clin Invest. 2004 detected in primary HCC tumours. It has been Apr;113(8):1138-48 suggested that MAPK7 is likely the target of 17p11 Barros JC, Marshall CJ. Activation of either ERK1/2 or amplification and that the ERK5 protein promotes ERK5 MAP kinase pathways can lead to disruption of the the growth of hepatic carcinoma cells by regulating cytoskeleton. J Cell Sci. 2005 Apr 15;118(Pt 8):1663- 71 mitotic entry (Zen et al., 2009). Carvajal-Vergara X, Tabera S, Montero JC, Esparís- Ogando A, López-Pérez R, Mateo G, Gutiérrez N, Parmo- References Cabañas M, Teixidó J, San Miguel JF, Pandiella A. Kato Y, Tapping RI, Huang S, Watson MH, Ulevitch RJ, Multifunctional role of Erk5 in multiple myeloma. Blood. Lee JD. Bmk1/Erk5 is required for cell proliferation induced 2005 Jun 1;105(11):4492-9 by . Nature. 1998 Oct Wang X, Merritt AJ, Seyfried J, Guo C, Papadakis ES, 15;395(6703):713-6 Finegan KG, Kayahara M, Dixon J, Boot-Handford RP, Hayashi M, Tapping RI, Chao TH, Lo JF, King CC, Yang Cartwright EJ, Mayer U, Tournier C. Targeted deletion of Y, Lee JD. BMK1 mediates growth factor-induced cell mek5 causes early embryonic death and defects in the proliferation through direct cellular activation of serum and extracellular signal-regulated kinase 5/myocyte enhancer glucocorticoid-inducible kinase. J Biol Chem. 2001 Mar factor 2 cell survival pathway. Mol Cell Biol. 2005 23;276(12):8631-4 Jan;25(1):336-45

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1116

MAPK7 (mitogen-activated protein kinase 7) Iñesta-Vaquera FdA, Cuenda A

Watanabe N, Nagamatsu Y, Gengyo-Ando K, Mitani S, the MEK5-ERK5 pathway in thymocyte apoptosis. EMBO Ohshima Y. Control of body size by SMA-5, a homolog of J. 2008 Jul 9;27(13):1896-906 MAP kinase BMK1/ERK5, in C. elegans. Development. 2005 Jul;132(14):3175-84 Finegan KG, Wang X, Lee EJ, Robinson AC, Tournier C. Regulation of neuronal survival by the extracellular signal- Kondoh K, Terasawa K, Morimoto H, Nishida E. regulated protein kinase 5. Cell Death Differ. 2009 Regulation of nuclear translocation of extracellular signal- May;16(5):674-83 regulated kinase 5 by active nuclear import and export mechanisms. Mol Cell Biol. 2006 Mar;26(5):1679-90 Montero JC, Ocaña A, Abad M, Ortiz-Ruiz MJ, Pandiella A, Esparís-Ogando A. Expression of Erk5 in early stage Truman AW, Millson SH, Nuttall JM, King V, Mollapour M, breast cancer and association with disease free survival Prodromou C, Pearl LH, Piper PW. Expressed in the yeast identifies this kinase as a potential therapeutic target. Saccharomyces cerevisiae, human ERK5 is a client of the PLoS One. 2009;4(5):e5565 Hsp90 chaperone that complements loss of the Slt2p (Mpk1p) cell integrity stress-activated protein kinase. Sawhney RS, Liu W, Brattain MG. A novel role of ERK5 in Eukaryot Cell. 2006 Nov;5(11):1914-24 integrin-mediated cell adhesion and motility in cancer cells via Fak signaling. J Cell Physiol. 2009 Apr;219(1):152-61 Cude K, Wang Y, Choi HJ, Hsuan SL, Zhang H, Wang CY, Xia Z. Regulation of the G2-M cell cycle progression by the Spiering D, Schmolke M, Ohnesorge N, Schmidt M, ERK5-NFkappaB signaling pathway. J Cell Biol. 2007 Apr Goebeler M, Wegener J, Wixler V, Ludwig S. MEK5/ERK5 23;177(2):253-64 signaling modulates endothelial cell migration and focal contact turnover. J Biol Chem. 2009 Sep Morimoto H, Kondoh K, Nishimoto S, Terasawa K, Nishida 11;284(37):24972-80 E. Activation of a C-terminal transcriptional activation domain of ERK5 by autophosphorylation. J Biol Chem. Zen K, Yasui K, Nakajima T, Zen Y, Zen K, Gen Y, 2007 Dec 7;282(49):35449-56 Mitsuyoshi H, Minami M, Mitsufuji S, Tanaka S, Itoh Y, Nakanuma Y, Taniwaki M, Arii S, Okanoue T, Yoshikawa McCracken SR, Ramsay A, Heer R, Mathers ME, Jenkins T. ERK5 is a target for gene amplification at 17p11 and BL, Edwards J, Robson CN, Marquez R, Cohen P, Leung promotes cell growth in hepatocellular carcinoma by HY. Aberrant expression of extracellular signal-regulated regulating mitotic entry. Genes Cancer. kinase 5 in human prostate cancer. Oncogene. 2008 May 2009 Feb;48(2):109-20 8;27(21):2978-88 This article should be referenced as such: Schramp M, Ying O, Kim TY, Martin GS. ERK5 promotes Src-induced podosome formation by limiting Rho Iñesta-Vaquera FdA, Cuenda A. MAPK7 (mitogen- activation. J Cell Biol. 2008 Jun 30;181(7):1195-210 activated protein kinase 7). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12):1114-1117. Sohn SJ, Lewis GM, Winoto A. Non-redundant function of

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1117

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

SLC16A1 (solute carrier family 16, member 1 (monocarboxylic acid transporter 1)) Céline Pinheiro, Fátima Baltazar Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal (CP, FB)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SLC16A1ID44046ch1p13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SLC16A1ID44046ch1p13.txt

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 Pseudogene 1 related pseudogene identified - AKR7 family Other names: FLJ36745, HHF7, MCT, MCT1, pseudogene (AFARP1), non-coding RNA. MGC44475 HGNC (Hugo): SLC16A1 Protein Location: 1p13.2 Description DNA/RNA 500 amino acids; 53958 Da; 12 transmembrane domains, intracellular N- and C-terminal and a Note large intracellular loop between transmembrane Human SLC16A1 was firstly cloned in 1994, by domains 6 and 7. Garcia and colleagues. Structural gene organization as well as isolation and characterization of Expression SLC16A1 promoter was achieved in 2002, by Cuff Ubiquitous. and Shirazi-Beechey. Localisation Description Plasma membrane; also described in rat 44507 bp lenght, containing 5 exons. Various SNPs mitochondrial and peroxisomal membranes. have been described in SLC16A1 gene. Transcription Function 6 transcripts have been described for this gene (4 Catalyses the proton-linked transport of with protein product, 2 with no protein product): metabolically important monocarboxylates such as SLC16A1-001 (5 exons; 3910 bps transcript length; lactate, pyruvate, branched-chain oxo acids derived 500 residues translation length); SLC16A1-002 (5 from leucine, valine and isoleucine, and ketone exons; 2101 bps transcript length; 456 residues bodies (acetoacetate, beta-hydroxybutyrate and translation length); SLC16A1-003 (4 exons; 865 acetate). bps transcript length; 215 residues translation Homology length); SLC16A1-004 (2 exons; 452 bps transcript length; no translation product); SLC16A1-005 (4 Belongs to the major facilitator superfamily (MFS). exons; 1099 bps transcript length; 296 residues Monocarboxylate porter (TC 2.A.1.13) family. translation length); SLC16A1-006 (2 exons; 430 SLC16A1 gene is conserved in chimpanzee, dog, bps transcript length; no translation product). cow, mouse, rat, chicken, and zebrafish.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1118

SLC16A1 (solute carrier family 16, member 1 Pinheiro C, Baltazar F (monocarboxylic acid transporter 1))

Protein diagram drawn following UniProtKB/Swiss-Prot database prediction, using TMRPres2D software.

Lauren's intestinal type, TNM staging and lymph- Implicated in node metastasis, in gastric cancer. Various cancers Colorectal carcinoma Note Note MCT1/SLC16A1 has been described to be MCT1/SLC16A1 has been described to be upregulated in a variety of tumours. downregulated in colorectal carcinoma (Lamber et Disease al., 2002). High grade glial neoplasms (Mathupala et al., 2004; Erythrocyte lactate transporter defect Fang et al., 2006), colorectal (Koukourakis et al., Note 2006; Pinheiro et al., 2008), lung (Koukourakis et al., 2007), cervical (Pinheiro et al., 2008), and Merezhinskaya et al. (2000) identified two breast carcinomas (Pinheiro et al., in Press). heterozygous transitions in the SLC16A1 gene, in patients with erythrocyte lactate transporter defect: Breast cancer 610A-G transition (resulting in a lys204-to-glu Prognosis (K204E) substitution in a highly conserved residue) In breast cancer, MCT1/SLC16A1 was found to be and 1414G-A transition (resulting in a gly472-to- associated with poor prognostic variables such as arg (G472R) substitution halfway along the basal-like subtype and high grade tumours cytoplasmic C-terminal chain). These substitutions (Pinheiro et al., in Press). are not conserved, but were not identified in 90 healthy control individuals. Erythrocyte lactate Oncogenesis clearance in patients with these mutations was 40 to SLC16A1 is expressed in normal breast tissue, but 50% that of normal control values. is silenced in breast cancer due to gene methylation (Asada et al., 2003). Hyperinsulinemic hypoglycemia familial 7 Gastric cancer Note Note Otonkoski et al. (2007) identified two heterozygotic The prognostic value of CD147 (a alterations in the SLC16A1, in affected members of MCT1/SLC16A1 and MCT4/SLC16A3 chaperone a Finnish family segregating autosomal dominant required for plasma membrane expression and exercise-induced hyperinsulinemic hypoglycemia. activity) was associated with MCT1/SLC16A1 co- First, a 163G-A transition in exon 1 located within expression in gastric cancer cells (Pinheiro et al., a binding site for nuclear matrix protein-1 and 2009). predicted to disrupt the binding sites of 2 potential Prognosis transcriptional repressors, and, secondly, a 25-bp Co-expression of MCT1/SLC16A1 with CD147 insertion at nucleotide -24 introducing additional was associated with advanced gastric carcinoma, binding sites for the ubiquitous transcription factors

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1119

SLC16A1 (solute carrier family 16, member 1 Pinheiro C, Baltazar F (monocarboxylic acid transporter 1))

SP1, USF and MZF1. The first variation leads to a ribonucleic acid inhibits glycolysis and induces cell death in 3-fold increase in transcription while the second malignant glioma: an in vitro study. Neurosurgery. 2004 Dec;55(6):1410-9; discussion 1419 variation leads to a 10-fold increase in transcription. These mutations were not found in 92 Finnish and Fang J, Quinones QJ, Holman TL, Morowitz MJ, Wang Q, Zhao H, Sivo F, Maris JM, Wahl ML. The H+-linked German controls. monocarboxylate transporter (MCT1/SLC16A1): a potential therapeutic target for high-risk neuroblastoma. Mol References Pharmacol. 2006 Dec;70(6):2108-15 Pinheiro C, Albergaria A, Paredes J, Sousa B, Dufloth R, Koukourakis MI, Giatromanolaki A, Harris AL, Sivridis E. Vieira D, Schmitt F, Baltazar F.. Monocarboxylate Comparison of metabolic pathways between cancer cells transporter 1 is upregulated in basal-like breast carcinoma. and stromal cells in colorectal carcinomas: a metabolic Histopathology In press survival role for tumor-associated stroma. Cancer Res. 2006 Jan 15;66(2):632-7 Garcia CK, Li X, Luna J, Francke U. cDNA cloning of the human monocarboxylate transporter 1 and chromosomal Koukourakis MI, Giatromanolaki A, Bougioukas G, Sivridis localization of the SLC16A1 locus to 1p13.2-p12. E. Lung cancer: a comparative study of metabolism related Genomics. 1994 Sep 15;23(2):500-3 protein expression in cancer cells and tumor associated stroma. Cancer Biol Ther. 2007 Sep;6(9):1476-9 Brooks GA, Brown MA, Butz CE, Sicurello JP, Dubouchaud H. Cardiac and skeletal muscle mitochondria Otonkoski T, Jiao H, Kaminen-Ahola N, Tapia-Paez I, have a monocarboxylate transporter MCT1. J Appl Physiol. Ullah MS, Parton LE, Schuit F, Quintens R, Sipilä I, 1999 Nov;87(5):1713-8 Mayatepek E, Meissner T, Halestrap AP, Rutter GA, Kere J. Physical exercise-induced hypoglycemia caused by Merezhinskaya N, Fishbein WN, Davis JI, Foellmer JW. failed silencing of monocarboxylate transporter 1 in Mutations in MCT1 cDNA in patients with symptomatic pancreatic beta cells. Am J Hum Genet. 2007 deficiency in lactate transport. Muscle Nerve. 2000 Sep;81(3):467-74 Jan;23(1):90-7 Pinheiro C, Longatto-Filho A, Ferreira L, Pereira SM, Cuff MA, Shirazi-Beechey SP. The human Etlinger D, Moreira MA, Jubé LF, Queiroz GS, Schmitt F, monocarboxylate transporter, MCT1: genomic organization Baltazar F. Increasing expression of monocarboxylate and promoter analysis. Biochem Biophys Res Commun. transporters 1 and 4 along progression to invasive cervical 2002 Apr 12;292(4):1048-56 carcinoma. Int J Gynecol Pathol. 2008 Oct;27(4):568-74 Lambert DW, Wood IS, Ellis A, Shirazi-Beechey SP. Pinheiro C, Longatto-Filho A, Scapulatempo C, Ferreira L, Molecular changes in the expression of human colonic Martins S, Pellerin L, Rodrigues M, Alves VA, Schmitt F, nutrient transporters during the transition from normality to Baltazar F. Increased expression of monocarboxylate malignancy. Br J Cancer. 2002 Apr 22;86(8):1262-9 transporters 1, 2, and 4 in colorectal carcinomas. Virchows Arch. 2008 Feb;452(2):139-46 Asada K, Miyamoto K, Fukutomi T, Tsuda H, Yagi Y, Wakazono K, Oishi S, Fukui H, Sugimura T, Ushijima T. Pinheiro C, Longatto-Filho A, Simões K, Jacob CE, Reduced expression of GNA11 and silencing of MCT1 in Bresciani CJ, Zilberstein B, Cecconello I, Alves VA, human breast cancers. Oncology. 2003;64(4):380-8 Schmitt F, Baltazar F. The prognostic value of CD147/EMMPRIN is associated with monocarboxylate McClelland GB, Khanna S, González GF, Butz CE, Brooks transporter 1 co-expression in gastric cancer. Eur J GA. Peroxisomal membrane monocarboxylate Cancer. 2009 Sep;45(13):2418-24 transporters: evidence for a redox shuttle system? Biochem Biophys Res Commun. 2003 Apr 25;304(1):130-5 This article should be referenced as such: Halestrap AP, Meredith D. The SLC16 gene family-from Pinheiro C, Baltazar F. SLC16A1 (solute carrier family 16, monocarboxylate transporters (MCTs) to aromatic amino member 1 (monocarboxylic acid transporter 1)). Atlas acid transporters and beyond. Pflugers Arch. 2004 Genet Cytogenet Oncol Haematol. 2010; 14(12):1118- Feb;447(5):619-28 1120. Mathupala SP, Parajuli P, Sloan AE. Silencing of monocarboxylate transporters via small interfering

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1120

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

STOML2 (stomatin (EPB72)-like 2) Wenfeng Cao, Liyong Zhang, Fang Ding, Zhumei Cui, Zhihua Liu State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (WC, LZ, DF, ZL); Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China (WC); Department of Obstetrics and Gynecology, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266011, China (ZC)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/STOML2ID44346ch9p13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI STOML2ID44346ch9p13.txt

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

frame however, commonly forms a 356 amino acid Identity residue polypeptide with a predicted molecular Other names: HSPC108, SLP-2 weight of 38.5 kDa. Similar to other family HGNC (Hugo): STOML2 members, SLP-2 as well as the stomatin from other species shares a characteristic NH2-terminal Location: 9p13.3 hydrophobic domain as well as a consensus cognate stomatin signature sequence that defines the DNA/RNA stomatin gene family, however, it lacks the NH2- Description terminal hydrophobic domain (Wang et al., 2000). The SLP-2 protein contains an alanine-rich domain The gene encoding SLP-2 was 3250 bp long and and a number of potential protein kinase C consisted of ten exons interrupted by nine introns. phosphorylation sites, cAMP-and-cGMP-dependent Transcription protein kinase phosphorylation sites and casein kinase II phosphorylation sites. There are 5 transcripts in this gene. However, a single 1.3 kb mRNA transcript encoding SLP-2 was Expression ubiquitously expressed, and the translation length is SLP-2 is widely expressed in many tissues and 356 residues (Owczarek et al., 2001). thought as a new component of the peripheral Pseudogene membrane skeleton. Especially, in the erythrocyte membrane, it also appears to exist at least partially No known pseudogenes. as an oligomeric protein complex. The overexpression of SLP-2 can be found in many Protein kinds of human tumors, such as esophageal Description squamous cell carcinoma, laryngeal squamous cell carcinoma, endometrial adenocarcinoma, and lung NP_038470; 356 aa. cancer. Human SLP-2 is presented on chromosome 9p13. The sequence at the 5'-end of the mRNA is Localisation interesting for the presence of three potential ATG Predominantly on plasma membrane and in the initiator sites, all sharing the same open reading cytoplasm.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1121

STOML2 (stomatin (EPB72)-like 2) Cao W, et al.

Figure A. ALA-RICH, Alanine-rich region profile: 224-274: score = 8.657. Figure B. MYRISTYL, N-myristoylation site: 16- 21: GSllAS, 31-36: GLprNT, 209-214: GTreSA, 314-319: GVvgAL, 326-331: GTpdSL, 341-346: GtdaSL; PKC-PHOSPHO-SITE, Protein kinase C phosphorylation site: 21-23: SgR, 78-80: SlK, 133-135: TmR, 335-337: SsR; CAMP-PHOSPHO-SITE, Camp-and-cGMP-dependent protein kinase phosphorylation site: 26-29: RRaS, 200-203: KRaT; CK2-PHOSPHO-SITE, Casein kinase II phosphorylation site: 78-81: SlkE, 156-159: SivD, 203-206: TvlE, 229-232: SeaE, 277-280: TvaE, 335-338: SsrD, 345-348: SldE; ASN-GLYCOSYLATION, N-glycosylation site: 96-99: NVTL, 154-157: NASI; AMIDATION, amidation site: 219-222: eGKK.

Function Implicated in Human SLP-2 protein with unknown function, we hypothesize that SLP-2 may link stomatin or other Various cancers integral membrane proteins to the peripheral Note cytoskeleton and thereby play a role in regulating SLP-2 has been shown to be over-expressed in a ion channel conductances or the organization of number of different cancers, including esophageal sphingolipid and cholesterol-rich lipid rafts. squamous cell carcinoma (ESCC), laryngeal Some recent results indicated that SLP-2 protein squamous cell carcinoma (LSCC), endometrial can significantly influence on multi-tumor adenocarcinoma (EAC), lung cancer (LC) and progression, which allowed us to identify this breast cancer (see below). unwell-known gene that maybe modulate invasion and metastasis of different cancers. Esophageal squamous cell carcinoma (ESCC) Homology Prognosis SLP-2 is one of the members of the Stomatin As shown in human ESCC, a significant correlation superfamily, among which identified vertebrate exists between SLP-2 protein high expression and homologues are SLP-1, SLP-2, and SLP-3. SLP-1 the depth of ESCC invasion (P=0.033) (Wang et al., is most abundant in brain and shares many 2009). similarities with UNC-24 (STOML1). Also, decreased cell growth and tumorigenesis in SLP-3 is specifically expressed in olfactory sensory the antisense transfectants revealed that SLP-2 may neurons (Seidel et al., 1998; Goldstein et al., 2003). be important in ESCC tumorigenesis (Zhang et al., Mutations 2006). No mutations have been reported for SLP-2. Laryngeal squamous cell carcinoma Mutation detection of SLP-2 exons was done using (LSCC) PCR and automated sequencing with 30 patient- Prognosis matched human esophageal cancer tissues. No In addition, SLP-2 takes part in human LSCC mutation was found within the open-reading frame malignant phenotype formation and development. of SLP-2 after sequencing results were aligned by High-level expression of SLP-2 protein could the procedure SeqMan of DNAStar software contribute to the prognostic characteristics of lymph (Zhang et al., 2006). node metastasis in human LSCC (Cao et al., 2007).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1122

STOML2 (stomatin (EPB72)-like 2) Cao W, et al.

Breast cancer family similar to Caenorhabditis elegans UNC-24. Gene. 1998 Dec 28;225(1-2):23-9 Prognosis Wang Y, Morrow JS. Identification and characterization of High-level expression of SLP-2 protein shows a human SLP-2, a novel homologue of stomatin (band 7.2b) worse prognosis, including increase in tumor size, present in erythrocytes and other tissues. J Biol Chem. progress in clinical stage, and appearance of lymph 2000 Mar 17;275(11):8062-71 node and/or distant metastasis and is associated Owczarek CM, Treutlein HR, Portbury KJ, Gulluyan LM, with decreased overall survival (P=0.011). Kola I, Hertzog PJ. A novel member of the Moreover, SLP-2 can be strongly associated with STOMATIN/EPB72/mec-2 family, stomatin-like 2 (STOML2), is ubiquitously expressed and localizes to HSA another important prognostic factor, HER-2/neu chromosome 9p13.1. Cytogenet Cell Genet. 2001;92(3- protein expression, which shows that they may act 4):196-203 as dependent prognostic factors to indicate poor Goldstein BJ, Kulaga HM, Reed RR. Cloning and prognosis (Cao et al., 2007). characterization of SLP3: a novel member of the stomatin Endometrial adenocarcinoma family expressed by olfactory receptor neurons. J Assoc Res Otolaryngol. 2003 Mar;4(1):74-82 Prognosis Zhang L, Ding F, Cao W, Liu Z, Liu W, Yu Z, Wu Y, Li W, Similarly, SLP-2 is also overexpressed in human Li Y, Liu Z. Stomatin-like protein 2 is overexpressed in endometrial adenocarcinoma (EAC) at both mRNA cancer and involved in regulating cell growth and cell and protein level. Sense transfection of SLP-2 in adhesion in human esophageal squamous cell carcinoma. EAC cell line accelerated cell growth whereas the Clin Cancer Res. 2006 Mar 1;12(5):1639-46 antisense transfection reduced cell growth in vitro Cao W, Zhang B, Liu Y, Li H, Zhang S, Fu L, Niu Y, Ning (Cui et al., 2007). L, Cao X, Liu Z, Sun B. High-level SLP-2 expression and HER-2/neu protein expression are associated with Lung cancer decreased breast cancer patient survival. Am J Clin Pathol. 2007 Sep;128(3):430-6 Prognosis At last, SLP-2 was overexpressed in human lung Cao WF, Zhang LY, Liu MB, Tang PZ, Liu ZH, Sun BC. Prognostic significance of stomatin-like protein 2 cancer (Zhang et al., 2006). High-level SLP-2 overexpression in laryngeal squamous cell carcinoma: expression was significantly correlated with distant clinical, histologic, and immunohistochemistry analyses metastasis, decreased overall survival and disease- with tissue microarray. Hum Pathol. 2007 May;38(5):747- free survival. SLP-2 overexpression was an 52 independent prognostic factor in multivariate Cui Z, Zhang L, Hua Z, Cao W, Feng W, Liu Z. Stomatin- analysis using the Cox regression model (p<0.05) like protein 2 is overexpressed and related to cell growth in human endometrial adenocarcinoma. Oncol Rep. 2007 (Chang et al., 2009). Apr;17(4):829-33 Mitochondrial component Wang Y, Cao W, Yu Z, Liu Z. Downregulation of a Note mitochondria associated protein SLP-2 inhibits tumor cell motility, proliferation and enhances cell sensitivity to SLP-2 localizes in mitochondria, affects chemotherapeutic reagents. Cancer Biol Ther. 2009 mitochondrial membrane potential (MMP) and Sep;8(17):1651-8 ATP production. Hence, SLP-2 is a mitochondrial Chang D, Ma K, Gong M, Cui Y, Liu ZH, Zhou XG, Zhou protein and therefore, functions in energy process CN, Wang TY. SLP-2 overexpression is associated with by MMP maintenance, and subsequently affecting tumour distant metastasis and poor prognosis in cell motility, proliferation and chemosensitivity pulmonary squamous cell carcinoma. Biomarkers. 2010 (Wang et al., 2009). Mar;15(2):104-10 References This article should be referenced as such: Cao W, Zhang L, Ding F, Cui Z, Liu Z. STOML2 (stomatin Seidel G, Prohaska R. Molecular cloning of hSLP-1, a (EPB72)-like 2). Atlas Genet Cytogenet Oncol Haematol. novel human brain-specific member of the band 7/MEC-2 2010; 14(12):1121-1123.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1123

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

AMOT (angiomotin) Roshan Mandrawalia, Ranjan Tamuli Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati-781 039, Assam, India (RM, RT)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/AMOTID632chXq23.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI AMOTID632chXq23.txt

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

(261), 721-751 (31), a PDZ-binding motif 1081- Identity 1084 (4), a SMC_prok_B region 429-549 (121), Other names: KIAA1071 and an angiomotin_C terminal 599-794 (196). HGNC (Hugo): AMOT Phosphorylations occur on S305, S312, S712, S714, T717, Y719, and T1061. Phosphorylated upon Location: Xq23 DNA damage, probably by ATM or ATR. Isoforms: DNA/RNA - Isoform 1: p130 angiomotin Description 1084 amino acids, 118085 Da. This isoform has been chosen as the 'canonical' sequence. DNA size 66.31 kb, mRNA size 6888 bp, 12 exons. - Isoform 2: p80 angiomotin 675 amino acids, 72540 Da. The isoform differs Protein from the canonical sequence with N-terminal alternative splicing region 1-409 (409) missing, Description which mediates the binding of angiomotin to F- Angiomotin protein is 1084 amino acid residues in actin stress fibres. The SMC_prok_B region is also length. It contains two coiled coil domains 429-689 missing in this isoform.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1124

AMOT (angiomotin) Mandrawalia R, Tamuli R

Expression Implicated in Expressed in placenta and skeletal muscle. Predominantly expressed in endothelial cells of Breast cancer capillaries, larger vessels of the placenta. Note Localisation Angiomotin is linked to angiogenesis and aggressive nature of breast tumours. Angiomotin Cell junction, tight junction. Localized on the cell shows high level of expression in mammary tissues . May act as a transmembrane protein. during tumour stages as compared to normal Function expression level (33.1 ± 11 in normal versus 86.5 ± 13.7 in tumour tissues, p=0.0003). Significant high Mediates inhibitory effect of angiostatin on tube expression was found in aggressive tumours (grade formation and the migration of endothelial cells 2, grade 3 and with nodal involvement) compared toward growth factors during the formation of new with less aggressive grade 1 tumour (p<0.001 and blood vessels in the larger vessels of the placenta. p=0.05 respectively). Angiogenesis is the essential Isoform-1 is found to control cell shape by process in the development and spread of breast association with F-actin fibres through N-terminal cancer, by providing blood supply to tumours and part of protein. The isoform 2 (p80) promotes escape route for tumour cells to other part of the angiogenesis, in part, by conferring a body. hypermigratory phenotype to endothelial cells. Hemangioendothelioma invasion Homology Disease The percent identity below represents identity of Angiomotin expression promotes AMOT over an aligned region in Unigene. hemangioendothelioma invasion. Expression of Mus musculus: 88.1 (percent identity) human angiomotin in mouse aortic endothelial Oryctolagus cuniculus: 79 (MAE) cells results in stabilization of tubes in the Sus scrofa: 72 Matrigel assay. Cells from the established tubes Danio rerio: 68.9 invaded into the solidified matrigel, however, cells Fugu rubripes: 65 expressing a functional mutant lacking the PDZ Xenopus laevis: 61.8 protein interaction motif did not migrate and form Caenorhabditis elegans: 46 tubes. Angiomotin may promote angiogenesis by Saccharomyces cerevisiae: 47 both stimulating invasion as well as stabilizing Drosophila melanogaster: 36 established tubes. Mutations Endothelial cell migration and tube formation Note Note Several polymorphisms have been found but none Upon expression of angiomotin in HeLa cells, of them has shown any association with a disease. angiomotin bound and internalized fluorescein- Furthermore, endothelial cells expressing mutated labeled angiostatin, a circulating inhibitor of angiomotins have been reported failure in their angiogenesis. In endothelial cells, angiomotin function, including failure to migrate and inhibition protein is localized to the leading edge of migrating of angiogenesis. Mutation with deletion of three cells and results in increased cell migration. amino acids from PDZ-binding motif results in Angiomotin-transfected MAE cells bind and inhibition of chemotaxis, embryos with this respond to angiostatin by inhibition of cell mutation may lead to death on embryonic day 9.5. migration and tube formation, which suggest that

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1125

AMOT (angiomotin) Mandrawalia R, Tamuli R

angiomotin regulates endothelial cell migration and Levchenko T, Carlson LM, Musiani P, Iezzi M, Curcio C, tube formation. Forni G, Cavallo F, Kiessling R. A DNA vaccine targeting angiomotin inhibits angiogenesis and suppresses tumor growth. Proc Natl Acad Sci U S A. 2006 Jun References 13;103(24):9208-13 Troyanovsky B, Levchenko T, Månsson G, Matvijenko O, Jiang WG, Watkins G, Douglas-Jones A, Holmgren L, Holmgren L. Angiomotin: an angiostatin binding protein Mansel RE. Angiomotin and angiomotin like proteins, their that regulates endothelial cell migration and tube expression and correlation with angiogenesis and clinical formation. J Cell Biol. 2001 Mar 19;152(6):1247-54 outcome in human breast cancer. BMC Cancer. 2006 Jan 23;6:16 Zetter BR. Hold that line. Angiomotin regulates endothelial cell motility. J Cell Biol. 2001 Mar 19;152(6):F35-6 Wells CD, Fawcett JP, Traweger A, Yamanaka Y, Goudreault M, Elder K, Kulkarni S, Gish G, Virag C, Lim C, Bratt A, Wilson WJ, Troyanovsky B, Aase K, Kessler R, Colwill K, Starostine A, Metalnikov P, Pawson T. A Van Meir EG, Holmgren L. Angiomotin belongs to a novel Rich1/Amot complex regulates the Cdc42 GTPase and protein family with conserved coiled-coil and PDZ binding apical-polarity proteins in epithelial cells. Cell. 2006 May domains. Gene. 2002 Sep 18;298(1):69-77 5;125(3):535-48 Levchenko T, Aase K, Troyanovsky B, Bratt A, Holmgren Ernkvist M, Luna Persson N, Audebert S, Lecine P, Sinha L. Loss of responsiveness to chemotactic factors by I, Liu M, Schlueter M, Horowitz A, Aase K, Weide T, Borg deletion of the C-terminal protein interaction site of JP, Majumdar A, Holmgren L. The Amot/Patj/Syx signaling angiomotin. J Cell Sci. 2003 Sep 15;116(Pt 18):3803-10 complex spatially controls RhoA GTPase activity in migrating endothelial cells. Blood. 2009 Jan 1;113(1):244- Levchenko T, Bratt A, Arbiser JL, Holmgren L. Angiomotin 53 expression promotes hemangioendothelioma invasion. Oncogene. 2004 Feb 19;23(7):1469-73 Gagné V, Moreau J, Plourde M, Lapointe M, Lord M, Gagnon E, Fernandes MJ. Human angiomotin-like 1 Bratt A, Birot O, Sinha I, Veitonmäki N, Aase K, Ernkvist associates with an angiomotin protein complex through its M, Holmgren L. Angiomotin regulates endothelial cell-cell coiled-coil domain and induces the remodeling of the actin junctions and cell motility. J Biol Chem. 2005 Oct cytoskeleton. Cell Motil Cytoskeleton. 2009 Sep;66(9):754- 14;280(41):34859-69 68 Ernkvist M, Aase K, Ukomadu C, Wohlschlegel J, Blackman R, Veitonmäki N, Bratt A, Dutta A, Holmgren L. This article should be referenced as such: p130-angiomotin associates to actin and controls endothelial cell shape. FEBS J. 2006 May;273(9):2000-11 Mandrawalia R, Tamuli R. AMOT (angiomotin). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12):1124- Holmgren L, Ambrosino E, Birot O, Tullus C, Veitonmäki N, 1126.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1126

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

BRCA2 (breast cancer 2, early onset) Frédéric Guénard, Francine Durocher Cancer Genomics Laboratory, Oncology and Molecular Endocrinology Research Centre, CRCHUL, CHUQ and Laval University, Quebec, G1V 4G2, Canada (FG, FD)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/BRCA2ID164ch13q13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI BRCA2ID164ch13q13.txt

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

The N-terminal part of the BRCA2 protein contains Identity a transcriptional activation domain (aa 18-105). Other names: BRCC2, BROVCA2, FACD, FAD, BRCA2 exon 11 encodes eight conserved motifs FAD1, FANCB, FANCD, FANCD1, GLM3 termed BRC repeats. Each of these repeats is HGNC (Hugo): BRCA2 composed of about 30 residues. A DNA-binding domain has been located in the C- Location: 13q13.1 terminal region of the BRCA2 protein (aa 2478- 3185). It is composed of a conserved helical DNA/RNA domain and three OB folds. Description Two nuclear localization signals (NLS) have been identified in the C-terminal region of BRCA2. The BRCA2 gene is composed of 27 exons and spans approximately 84.2 kb of genomic DNA. Expression BRCA2 expression is proportional to the rate of cell Transcription proliferation. Non-dividing cells do not express The BRCA2 gene encodes a 11386 bp mRNA BRCA2 while wide expression of BRCA2 was transcript. Transcription site is located 227 bp observed in actively dividing tissues, including the upstream the first ATG of the BRCA2 ORF. The epithelium of the breast during puberty and translation start site is located in exon 2. pregnancy. Pseudogene The BRCA2 expression is regulated during the cell cycle, with highest expression during the S phase of No pseudogene reported. the cell cycle. Most of the BRCA2 proteins are associated to Protein DSS1. The presence of DSS1 was demonstrated to Description stabilize the BRCA2 protein. Human BRCA2 protein is composed of 3418 amino Localisation acids (384 kDa). BRCA2 is a nuclear protein.

Structure of BRCA2. BRCA2 is a 3418 aa protein. BRC repeats: BRCA C-terminal repeats; NLS: Nuclear localization signals.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1127

BRCA2 (breast cancer 2, early onset) Guénard F, Durocher F

Function 11%. Increased risk of several other cancers are associated with BRCA2 mutations, especially for BRCA2 has been implicated in maintenance of prostate and pancreatic cancer. genomic integrity and in the cellular response to DNA damage. The BRCA2 protein interacts with Somatic the RAD51 recombinase to regulate homologous Somatic mutations in BRCA2 are infrequent in recombination (HR). BRCA2 regulates the sporadic breast cancer. Methylation of the BRCA2 intracellular localization of RAD51. It also targets promoter has not been detected in normal tissues the RAD51 to ssDNA and inhibits dsDNA binding, nor in breast and ovarian cancers. Loss of thus regulating/enhancing DNA strand exchange heterozygosity at the BRCA2 locus has been activity of RAD51. CHEK1 and CHEK2 both frequently found in sporadic breast and ovarian phosphorylate the RAD51/BRCA2 complex and tumors. regulate the functional association of this complex in response to DNA damage. Implicated in BRCA2 is also implicated in cell cycle checkpoints. Following exposure to X-rays or UV light, cells Breast cancer expressing truncated BRCA2 protein exhibit arrest Note in the G1 and G2/M phases. BRCA2 protein plays a Informations regarding breast cancer and BRCA2 role in mitotic spindle assembly checkpoints mutations and polymorphisms are available in a through modulation of the level of spindle assembly central repository formed by the National Human checkpoint proteins including Aurora A and Aurora Genome Research; National Institute of Health. B. This repository, named Breast Cancer Information A role in regulation of transcription has been Core (BIC) - NHGRI, is available at the following attributed to BRCA2. BRCA2 binding to the DSS1 address: http://research.nhgri.nih.gov/bic/. protein appears to be required for proper Disease completion of cell division in yeast. Breast tumors in BRCA2 carriers are found at The BRCA2 protein demonstrated the ability to higher histologic grade (2 and 3) than sporadic stimulate transcription. For example, exogenous tumors. Tumors from BRCA2 carriers are more expression of BRCA2 can stimulate transcription of commonly found to be stage IV than sporadic androgen receptor-regulated genes. This function of control tumors and BRCA2-associated breast BRCA2 is regulated by the binding of the EMSY cancer cases are more often node-positive than protein to the region of BRCA2 responsible for control breast cancer cases. transcriptional activation. An excess of EMSY results in silencing of BRCA2-driven Prognosis transcriptional activation. BRCA2 mutation carriers show younger mean age BRCA2 localizes to meiotic chromosomes during at diagnosis than sporadic breast cancer cases. early meiotic prophase I when homologous Bilateral breast cancer is found more commonly in chromosomes undergo synapsis. Moreover, BRCA2 BRCA2-associated breast cancer than in sporadic interacts with the meiosis-specific recombinase breast cancer. DMC1, thus implicating BRCA2 in meiotic ER and PR expression in BRCA2 tumors are recombination. similar than in control tumors, which contrasts with Homology ER and PR expression found in BRCA1 tumors. Oncogenesis BRCA2 homologs have been found in a diverse It was suggested that genomic rearrangements range of organisms. In addition to zebrafish and C. account for 7.7% of the BRCA2 mutation spectrum. elegans, homologs exist in diverse eukaryotes, from Loss of the wild-type allele is not required for plants to parasitic organisms. breast tumorigenesis in BRCA2 mutation carriers. Low general conservation is found in BRCA2. Somatic mutations of the BRCA2 gene are an Higher level of homology is observed for several infrequent event in sporadic breast cancer tumors. segments, including transactivation domain, BRC Loss of heterozygosity at the BRCA2 locus on repeats and nuclear localization signals located chromosome 13q12-q13 was observed in within C-terminal region. approximately 30% of sporadic breast cancer. Methylation of the CpG dinucleotide within the Mutations BRCA2 promoter is not found in normal and Germinal neoplastic breast tissues. High risk of breast and ovarian cancer is associated Male breast cancer with germline BRCA2 mutations. Cumulative risk Note of breast cancer in BRCA2 mutation carriers was A cumulative risk of 6% and 7% of developing estimated to 45% by the age of 70 years while breast cancer by the age of 70 and 80, respectively, ovarian cancer risk in carriers was estimated to has been estimated for male BRCA2 mutation

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1128

BRCA2 (breast cancer 2, early onset) Guénard F, Durocher F

carriers. BRCA2 mutations have been found in 14% less than 1% of early-onset prostate cancer in the of familial male breast cancer and 4% of unselected US Caucasian population while such mutations are male breast cancer cases. responsible for 2.3% of early-onset prostate cancer Disease diagnosed in United Kingdom. Male breast cancers are mostly ductal or Most studies conducted on hereditary prostate unclassified carcinomas. Papillary, mucinous and cancer families did not revealed a contribution of lobular carcinomas each represent less than 3% of BRCA2 truncating mutations in these families. male breast cancers. Estrogen receptor and However, a small study conducted on a limited receptor expression is found in number of families found BRCA2 mutations in two approximately 90% and 81% of male breast families. Incomplete segregation of the mutation cancers, respectively. with the disease was found in these families as affected brothers did not carry these mutations. Prognosis Overall survival rates for male breast cancers are Prognosis lower than for female breast cancers due to the BRCA2 mutation carriers have a significantly lower older age and more advances disease at the time of mean age at diagnosis of prostate cancer and shorter diagnosis. Male breast cancers associated with mean survival time than non-carriers. BRCA2 BRCA2 mutation are diagnosed at younger age mutation carriers show more advanced tumor stage than sporadic male breast cancer cases. and higher grade at diagnosis. Prostate cancer carriers of a BRCA2 mutation show poorer survival Ovarian cancer than BRCA1 carriers. Prostate cancer patients Note which are carriers of the 999del5 Icelandic founder Carriers of mutations in the central portion of mutation appear to have worse prognosis than non- BRCA2, termed OCCR (ovarian cancer cluster carriers of this mutation. region; aa 1012-2210), are at higher risk of ovarian Histopathological features of prostate cancer in cancer and lower breast cancer risk than carriers of BRCA2 mutation carriers revealed that prostate mutations outside the OCCR. cancer developed in mutation carriers show higher Gleason scores in than non-carriers. Disease Ovarian cancer is mostly epithelial tumors (90%) Oncogenesis and lifetime risk of ovarian cancer in the general Allelic loss at the BRCA2 locus was identified in a population is estimated to be 1-1.5%. Risk of majority of prostate tumor samples from carriers of ovarian cancer in BRCA2 mutation carriers is the c.999del5 mutation, thus suggesting that no estimated to be 10%. functional BRCA2 protein is found in these tumors. Prognosis Stomach cancer BRCA2 ovarian tumors are similar to BRCA1 Note ovarian tumors as these two types of tumors are Stomach cancer was reported in family members of more likely to be serous adenocarcinomas and women with ovarian cancer carrying a BRCA2 higher grade than control tumors. BRCA2- mutation within the OCCR. On the other hand, the associated ovarian cancers occur later in life than presence of stomach cancer in relatives of ovarian BRCA1-related or control ovarian tumors. cancer cases is strongly predictive of the presence Oncogenesis of a BRCA2 mutation. Specifically, the BRCA2 Complete loss of the wild-type BRCA2-allele is 999del5 mutation is associated with an increased observed in BRCA2-associated ovarian cancers. risk of stomach cancer in first- and second-degree Loss of heterozygosity at 13q12-q14 is also relatives. observed in sporadic epithelial ovarian tumors. On Assessment of the presence of non-breast or ovarian the other hand, CpG dinucleotide methylation of the cancers in BRCA2 mutation carriers estimated a BRCA2 promoter is not found in sporadic ovarian relative risk of stomach cancer of 2.59 to be cancers. associated with BRCA2 mutations. Meta-analysis Prostate cancer of published studies latter confirmed increased risk of stomach cancer in BRCA2 carriers. Note Different studies conducted on BRCA2 mutation Pharyngeal cancer carriers revealed an increased risk of prostate Note cancer in BRCA2 mutation carriers. Relative risk An increased risk of buccal cavity and pharynx associated with BRCA2 mutations is estimated to cancer was suggested during the assessment of be approximately 2.5 to 5. cancers other than breast and ovarian cancer in Protein-truncating BRCA2 mutations are associated BRCA2 mutation carriers. This was thereafter with early-onset prostate cancer. Different studies confirmed in a cohort of BRCA2 mutation carriers revealed that BRCA2 mutations are responsible for leading to the estimation of a relative risk of 7.3

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1129

BRCA2 (breast cancer 2, early onset) Guénard F, Durocher F

(95% CI = 2.0 - 18.6). Higher relative risk of pancreatic tumorigenesis in patients with germline pharyngeal cancer is found for carriers younger BRCA2 mutation. than 65 years old. Malignant melanoma Gallbladder and bile duct cancer Note Note BRCA2 mutation carriers were estimated to be at Evaluation of risks of cancers other than breast and higher risk of developing malignant melanoma (RR ovarian cancers in BRCA2 carriers found a higher = 2.58; 95% CI = 1.28-5.17). Despite many studies risk of gallbladder and bile duct cancer in BRCA2 reported malignant melanoma in mutation carriers carriers (RR = 4.97; 95% CI = 1.50-16.52). or in their relatives, other studies did not confirm Specifically, the 6167delT Jewish Ashkenazi this association. founder BRCA2 mutation was observed at Bone cancer significantly higher rate in bile duct cancer cases than in population controls. Note An excess risk of bone cancer (RR = 14.4; 95% CI Colon cancer = 2.9 - 42.1) was observed in a cohort of BRCA2 Note mutation carriers from the Netherlands. It was reported that risk of colorectal cancer in first- Fanconi anemia (complementation degree relatives of BRCA2 mutation carriers affected with ovarian cancer is increased by group D1) threefold for BRCA2 mutations located within the Note OCCR. Biallelic mutations of the BRCA2 gene are Analysis of a BRCA2 mutation in different families responsible for Fanconi anemia subgroup D1 (FA- led to the suggestion that BRCA2 mutations D1). predispose to colon cancer. It was thereafter Disease reported that BRCA2 mutation carriers are at Fanconi anemia (FA) is a recessive disease increased risk of colon cancer before the age of 65 characterized by various clinical features. Many years old. The association of BRCA2 mutations developmental defects are found in FA patients. with colon cancer was latter confirmed in a meta- Radial aplasia, microcephaly, microphthalmia, analysis. small stature, skin hyperpigmentation and Pancreas cancer malformation of the kidneys are encountered in FA Note patients. Very high frequency of bone marrow failure, leukemia and squamous cell carcinoma of Different studies suggested that BRCA2 mutations the head and neck as well as gynecological are associated with less than 1% of sporadic squamous cell carcinoma are associated with FA. pancreatic cancer in Caucasians while such Bone marrow failure generally leads to aplastic mutations could account for 10% of sporadic anemia during the first decade of life. Esophageal pancreatic cancer in Ashkenazi Jewish population. carcinoma and liver, brain, skin and renal tumors Approximately 10% of patients developing are also found in FA patients. pancreatic cancer show patterns of hereditary predisposition. Screening of BRCA2 mutations in Prognosis familial pancreatic cancer cases suggested that The FA-D1 and FA-N subgroups are clinically BRCA2 mutations account for 6-17% of these different from other FA subgroups as these families. Following the identification of germline subgroups are associated with increased BRCA2 mutations in pancreatic cancer, it was predisposition to solid childhood malignancies such evaluated that BRCA2 mutations confer roughly a as medulloblastoma and Wilms tumor. 3.5-folds increased risk. Relative risk of pancreatic Cytogenetics cancer was found to be higher at younger age At the cellular level, FA is a chromosomal fragility (younger than 65 years old). Different studies syndrome. FA cells are hypersensitive to DNA evaluated the lifetime risk of pancreatic cancer in interstrand crosslinking agents such as mitomycin BRCA2 mutation carriers to be approximately 5%. C, diepoxybutane and cisplatin. In addition to Prognosis hypersensitivity to these agents, FA cells show an Among human malignancies, pancreatic cancer has increased number of spontaneous breaks. one of the worst prognoses. Oncogenesis References Pancreatic intraepithelial neoplasia (PanIN) Wooster R, Neuhausen SL, Mangion J, Quirk Y, Ford D, analysis in BRCA2 mutation carriers revealed that Collins N, Nguyen K, Seal S, Tran T, Averill D. Localization loss of the wild type BRCA2 allele is found solely of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science. 1994 Sep in high-grade PanIN, thus suggesting that biallelic 30;265(5181):2088-90 inactivation of the BRCA2 gene is a late event in

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1130

BRCA2 (breast cancer 2, early onset) Guénard F, Durocher F

Cleton-Jansen AM, Collins N, Lakhani SR, Weissenbach J, and sporadic cases. Breast Cancer Linkage Consortium. Devilee P, Cornelisse CJ, Stratton MR. Loss of Lancet. 1997 May 24;349(9064):1505-10 heterozygosity in sporadic breast tumours at the BRCA2 locus on chromosome 13q12-q13. Br J Cancer. 1995 Bertwistle D, Swift S, Marston NJ, Jackson LE, Crossland Nov;72(5):1241-4 S, Crompton MR, Marshall CJ, Ashworth A. Nuclear location and cell cycle regulation of the BRCA2 protein. Wooster R, Bignell G, Lancaster J, Swift S, Seal S, Cancer Res. 1997 Dec 15;57(24):5485-8 Mangion J, Collins N, Gregory S, Gumbs C, Micklem G. Identification of the breast cancer susceptibility gene Bignell G, Micklem G, Stratton MR, Ashworth A, Wooster BRCA2. Nature. 1995 Dec 21-28;378(6559):789-92 R. The BRC repeats are conserved in mammalian BRCA2 proteins. Hum Mol Genet. 1997 Jan;6(1):53-8 Berman DB, Costalas J, Schultz DC, Grana G, Daly M, Godwin AK. A common mutation in BRCA2 that Collins N, Wooster R, Stratton MR. Absence of methylation predisposes to a variety of cancers is found in both Jewish of CpG dinucleotides within the promoter of the breast Ashkenazi and non-Jewish individuals. Cancer Res. 1996 cancer susceptibility gene BRCA2 in normal tissues and in Aug 1;56(15):3409-14 breast and ovarian cancers. Br J Cancer. 1997;76(9):1150- 6 Bork P, Blomberg N, Nilges M. Internal repeats in the BRCA2 protein sequence. Nat Genet. 1996 May;13(1):22- Easton DF, Steele L, Fields P, Ormiston W, Averill D, Daly 3 PA, McManus R, Neuhausen SL, Ford D, Wooster R, Cannon-Albright LA, Stratton MR, Goldgar DE. Cancer Couch FJ, Farid LM, DeShano ML, Tavtigian SV, Calzone risks in two large breast cancer families linked to BRCA2 K, Campeau L, Peng Y, Bogden B, Chen Q, Neuhausen S, on chromosome 13q12-13. Am J Hum Genet. 1997 Shattuck-Eidens D, Godwin AK, Daly M, Radford DM, Jul;61(1):120-8 Sedlacek S, Rommens J, Simard J, Garber J, Merajver S, Weber BL. BRCA2 germline mutations in male breast Friedman LS, Gayther SA, Kurosaki T, Gordon D, Noble B, cancer cases and breast cancer families. Nat Genet. 1996 Casey G, Ponder BA, Anton-Culver H. Mutation analysis of May;13(1):123-5 BRCA1 and BRCA2 in a male breast cancer population. Am J Hum Genet. 1997 Feb;60(2):313-9 Donegan WL, Redlich PN. Breast cancer in men. Surg Clin North Am. 1996 Apr;76(2):343-63 Gayther SA, Mangion J, Russell P, Seal S, Barfoot R, Ponder BA, Stratton MR, Easton D. Variation of risks of Goggins M, Schutte M, Lu J, Moskaluk CA, Weinstein CL, breast and ovarian cancer associated with different Petersen GM, Yeo CJ, Jackson CE, Lynch HT, Hruban germline mutations of the BRCA2 gene. Nat Genet. 1997 RH, Kern SE. Germline BRCA2 gene mutations in patients Jan;15(1):103-5 with apparently sporadic pancreatic carcinomas. Cancer Res. 1996 Dec 1;56(23):5360-4 McAllister KA, Haugen-Strano A, Hagevik S, Brownlee HA, Collins NK, Futreal PA, Bennett LM, Wiseman RW. Lancaster JM, Wooster R, Mangion J, Phelan CM, Characterization of the rat and mouse homologues of the Cochran C, Gumbs C, Seal S, Barfoot R, Collins N, Bignell BRCA2 breast cancer susceptibility gene. Cancer Res. G, Patel S, Hamoudi R, Larsson C, Wiseman RW, 1997 Aug 1;57(15):3121-5 Berchuck A, Iglehart JD, Marks JR, Ashworth A, Stratton MR, Futreal PA. BRCA2 mutations in primary breast and Milner J, Ponder B, Hughes-Davies L, Seltmann M, ovarian cancers. Nat Genet. 1996 Jun;13(2):238-40 Kouzarides T. Transcriptional activation functions in BRCA2. Nature. 1997 Apr 24;386(6627):772-3 Lynch HT, Smyrk T, Kern SE, Hruban RH, Lightdale CJ, Lemon SJ, Lynch JF, Fusaro LR, Fusaro RM, Ghadirian P. Mizuta R, LaSalle JM, Cheng HL, Shinohara A, Ogawa H, Familial pancreatic cancer: a review. Semin Oncol. 1996 Copeland N, Jenkins NA, Lalande M, Alt FW. RAB22 and Apr;23(2):251-75 RAB163/mouse BRCA2: proteins that specifically interact with the RAD51 protein. Proc Natl Acad Sci U S A. 1997 Tavtigian SV, Simard J, Rommens J, Couch F, Shattuck- Jun 24;94(13):6927-32 Eidens D, Neuhausen S, Merajver S, Thorlacius S, Offit K, Stoppa-Lyonnet D, Belanger C, Bell R, Berry S, Bogden R, Ozçelik H, Schmocker B, Di Nicola N, Shi XH, Langer B, Chen Q, Davis T, Dumont M, Frye C, Hattier T, Moore M, Taylor BR, Narod SA, Darlington G, Andrulis IL, Jammulapati S, Janecki T, Jiang P, Kehrer R, Leblanc JF, Gallinger S, Redston M. Germline BRCA2 6174delT Mitchell JT, McArthur-Morrison J, Nguyen K, Peng Y, mutations in Ashkenazi Jewish pancreatic cancer patients. Samson C, Schroeder M, Snyder SC, Steele L, Nat Genet. 1997 May;16(1):17-8 Stringfellow M, Stroup C, Swedlund B, Swense J, Teng D, Rajan JV, Marquis ST, Gardner HP, Chodosh LA. Thomas A, Tran T, Tranchant M, Weaver-Feldhaus J, Developmental expression of Brca2 colocalizes with Brca1 Wong AK, Shizuya H, Eyfjord JE, Cannon-Albright L, and is associated with proliferation and differentiation in Tranchant M, Labrie F, Skolnick MH, Weber B, Kamb A, multiple tissues. Dev Biol. 1997 Apr 15;184(2):385-401 Goldgar DE. The complete BRCA2 gene and mutations in chromosome 13q-linked kindreds. Nat Genet. 1996 Sharan SK, Morimatsu M, Albrecht U, Lim DS, Regel E, Mar;12(3):333-7 Dinh C, Sands A, Eichele G, Hasty P, Bradley A. Embryonic lethality and radiation hypersensitivity mediated Teng DH, Bogden R, Mitchell J, Baumgard M, Bell R, by Rad51 in mice lacking Brca2. Nature. 1997 Apr Berry S, Davis T, Ha PC, Kehrer R, Jammulapati S, Chen 24;386(6627):804-10 Q, Offit K, Skolnick MH, Tavtigian SV, Jhanwar S, Swedlund B, Wong AK, Kamb A. Low incidence of BRCA2 Sigurdsson S, Thorlacius S, Tomasson J, Tryggvadottir L, mutations in breast carcinoma and other cancers. Nat Benediktsdottir K, Eyfjörd JE, Jonsson E. BRCA2 mutation Genet. 1996 Jun;13(2):241-4 in Icelandic prostate cancer patients. J Mol Med. 1997 Oct;75(10):758-61 Thorlacius S, Olafsdottir G, Tryggvadottir L, Neuhausen S, Jonasson JG, Tavtigian SV, Tulinius H, Ogmundsdottir Wong AK, Pero R, Ormonde PA, Tavtigian SV, Bartel PL. HM, Eyfjörd JE. A single BRCA2 mutation in male and RAD51 interacts with the evolutionarily conserved BRC female breast cancer families from Iceland with varied motifs in the human breast cancer susceptibility gene cancer phenotypes. Nat Genet. 1996 May;13(1):117-9 brca2. J Biol Chem. 1997 Dec 19;272(51):31941-4 . Pathology of familial breast cancer: differences between Agnarsson BA, Jonasson JG, Björnsdottir IB, Barkardottir breast cancers in carriers of BRCA1 or BRCA2 mutations RB, Egilsson V, Sigurdsson H. Inherited BRCA2 mutation

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1131

BRCA2 (breast cancer 2, early onset) Guénard F, Durocher F

associated with high grade breast cancer. Breast Cancer Gayther SA, de Foy KA, Harrington P, Pharoah P, Res Treat. 1998 Jan;47(2):121-7 Dunsmuir WD, Edwards SM, Gillett C, Ardern-Jones A, Dearnaley DP, Easton DF, Ford D, Shearer RJ, Kirby RS, Chen J, Silver DP, Walpita D, Cantor SB, Gazdar AF, Dowe AL, Kelly J, Stratton MR, Ponder BA, Barnes D, Tomlinson G, Couch FJ, Weber BL, Ashley T, Livingston Eeles RA. The frequency of germ-line mutations in the DM, Scully R. Stable interaction between the products of breast cancer predisposition genes BRCA1 and BRCA2 in the BRCA1 and BRCA2 tumor suppressor genes in mitotic familial prostate cancer. The Cancer Research and meiotic cells. Mol Cell. 1998 Sep;2(3):317-28 Campaign/British Prostate Group United Kingdom Familial Chen PL, Chen CF, Chen Y, Xiao J, Sharp ZD, Lee WH. Prostate Cancer Study Collaborators. Cancer Res. 2000 The BRC repeats in BRCA2 are critical for RAD51 binding Aug 15;60(16):4513-8 and resistance to methyl methanesulfonate treatment. Goggins M, Hruban RH, Kern SE. BRCA2 is inactivated Proc Natl Acad Sci U S A. 1998 Apr 28;95(9):5287-92 late in the development of pancreatic intraepithelial Chodosh LA. Expression of BRCA1 and BRCA2 in normal neoplasia: evidence and implications. Am J Pathol. 2000 and neoplastic cells. J Mammary Gland Biol Neoplasia. May;156(5):1767-71 1998 Oct;3(4):389-402 Loman N, Johannsson O, Bendahl P, Dahl N, Einbeigi Z, Ford D, Easton DF, Stratton M, Narod S, Goldgar D, Gerdes A, Borg A, Olsson H. Prognosis and clinical Devilee P, Bishop DT, Weber B, Lenoir G, Chang-Claude presentation of BRCA2-associated breast cancer. Eur J J, Sobol H, Teare MD, Struewing J, Arason A, Scherneck Cancer. 2000 Jul;36(11):1365-73 S, Peto J, Rebbeck TR, Tonin P, Neuhausen S, Scully R, Livingston DM. In search of the tumour- Barkardottir R, Eyfjord J, Lynch H, Ponder BA, Gayther suppressor functions of BRCA1 and BRCA2. Nature. 2000 SA, Zelada-Hedman M. Genetic heterogeneity and Nov 23;408(6811):429-32 penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Sinclair CS, Berry R, Schaid D, Thibodeau SN, Couch FJ. Consortium. Am J Hum Genet. 1998 Mar;62(3):676-89 BRCA1 and BRCA2 have a limited role in familial prostate cancer. Cancer Res. 2000 Mar 1;60(5):1371-5 Lynch BJ, Holden JA, Buys SS, Neuhausen SL, Gaffney DK. Pathobiologic characteristics of hereditary breast Venkitaraman AR. The breast cancer susceptibility gene, cancer. Hum Pathol. 1998 Oct;29(10):1140-4 BRCA2: at the crossroads between DNA replication and recombination? Philos Trans R Soc Lond B Biol Sci. 2000 Marmorstein LY, Ouchi T, Aaronson SA. The BRCA2 gene Feb 29;355(1394):191-8 product functionally interacts with p53 and RAD51. Proc Natl Acad Sci U S A. 1998 Nov 10;95(23):13869-74 Yu VP, Koehler M, Steinlein C, Schmid M, Hanakahi LA, van Gool AJ, West SC, Venkitaraman AR. Gross Patel KJ, Yu VP, Lee H, Corcoran A, Thistlethwaite FC, chromosomal rearrangements and genetic exchange Evans MJ, Colledge WH, Friedman LS, Ponder BA, between nonhomologous chromosomes following BRCA2 Venkitaraman AR. Involvement of Brca2 in DNA repair. inactivation. Genes Dev. 2000 Jun 1;14(11):1400-6 Mol Cell. 1998 Feb;1(3):347-57 Davies AA, Masson JY, McIlwraith MJ, Stasiak AZ, Stasiak Zhang H, Tombline G, Weber BL. BRCA1, BRCA2, and A, Venkitaraman AR, West SC. Role of BRCA2 in control DNA damage response: collision or collusion? Cell. 1998 of the RAD51 recombination and DNA repair protein. Mol Feb 20;92(4):433-6 Cell. 2001 Feb;7(2):273-82 . Cancer risks in BRCA2 mutation carriers. The Breast Figer A, Irmin L, Geva R, Flex D, Sulkes J, Sulkes A, Cancer Linkage Consortium. J Natl Cancer Inst. 1999 Aug Friedman E. The rate of the 6174delT founder Jewish 4;91(15):1310-6 mutation in BRCA2 in patients with non-colonic Armes JE, Trute L, White D, Southey MC, Hammet F, gastrointestinal tract tumours in Israel. Br J Cancer. 2001 Tesoriero A, Hutchins AM, Dite GS, McCredie MR, Giles Feb;84(4):478-81 GG, Hopper JL, Venter DJ. Distinct molecular Gras E, Cortes J, Diez O, Alonso C, Matias-Guiu X, Baiget pathogeneses of early-onset breast cancers in BRCA1 and M, Prat J. Loss of heterozygosity on chromosome 13q12- BRCA2 mutation carriers: a population-based study. q14, BRCA-2 mutations and lack of BRCA-2 promoter Cancer Res. 1999 Apr 15;59(8):2011-7 hypermethylation in sporadic epithelial ovarian tumors. Chen CF, Chen PL, Zhong Q, Sharp ZD, Lee WH. Cancer. 2001 Aug 15;92(4):787-95 Expression of BRC repeats in breast cancer cells disrupts Marmorstein LY, Kinev AV, Chan GK, Bochar DA, Beniya the BRCA2-Rad51 complex and leads to radiation H, Epstein JA, Yen TJ, Shiekhattar R. A human BRCA2 hypersensitivity and loss of G(2)/M checkpoint control. J complex containing a structural DNA binding component Biol Chem. 1999 Nov 12;274(46):32931-5 influences cell cycle progression. Cell. 2001 Jan Marston NJ, Richards WJ, Hughes D, Bertwistle D, 26;104(2):247-57 Marshall CJ, Ashworth A. Interaction between the product Moynahan ME, Pierce AJ, Jasin M. BRCA2 is required for of the breast cancer susceptibility gene BRCA2 and DSS1, homology-directed repair of chromosomal breaks. Mol a protein functionally conserved from yeast to mammals. Cell. 2001 Feb;7(2):263-72 Mol Cell Biol. 1999 Jul;19(7):4633-42 Risch HA, McLaughlin JR, Cole DE, Rosen B, Bradley L, Spain BH, Larson CJ, Shihabuddin LS, Gage FH, Verma Kwan E, Jack E, Vesprini DJ, Kuperstein G, Abrahamson IM. Truncated BRCA2 is cytoplasmic: implications for JL, Fan I, Wong B, Narod SA. Prevalence and penetrance cancer-linked mutations. Proc Natl Acad Sci U S A. 1999 of germline BRCA1 and BRCA2 mutations in a population Nov 23;96(24):13920-5 series of 649 women with ovarian cancer. Am J Hum Verhoog LC, Brekelmans CT, Seynaeve C, Dahmen G, Genet. 2001 Mar;68(3):700-10 van Geel AN, Bartels CC, Tilanus-Linthorst MM, Wagner Thompson D, Easton D. Variation in cancer risks, by A, Devilee P, Halley DJ, van den Ouweland AM, Meijers- mutation position, in BRCA2 mutation carriers. Am J Hum Heijboer EJ, Klijn JG. Survival in hereditary breast cancer Genet. 2001 Feb;68(2):410-9 associated with germline mutations of BRCA2. J Clin Oncol. 1999 Nov;17(11):3396-402 Howlett NG, Taniguchi T, Olson S, Cox B, Waisfisz Q, De Die-Smulders C, Persky N, Grompe M, Joenje H, Pals G,

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1132

BRCA2 (breast cancer 2, early onset) Guénard F, Durocher F

Ikeda H, Fox EA, D'Andrea AD. Biallelic inactivation of B, Fischbein A, Gruber SB, Rennert G, Ronchetti RD, BRCA2 in Fanconi anemia. Science. 2002 Jul Hewitt SM, Struewing JP, Iscovich J. A twofold increase in 26;297(5581):606-9 BRCA mutation related prostate cancer among Ashkenazi Israelis is not associated with distinctive histopathology. J Kojic M, Kostrub CF, Buchman AR, Holloman WK. BRCA2 Med Genet. 2003 Oct;40(10):787-92 homolog required for proficiency in DNA repair, recombination, and genome stability in Ustilago maydis. Hahn SA, Greenhalf B, Ellis I, Sina-Frey M, Rieder H, Mol Cell. 2002 Sep;10(3):683-91 Korte B, Gerdes B, Kress R, Ziegler A, Raeburn JA, Campra D, Grützmann R, Rehder H, Rothmund M, Lakhani SR, Van De Vijver MJ, Jacquemier J, Anderson Schmiegel W, Neoptolemos JP, Bartsch DK. BRCA2 TJ, Osin PP, McGuffog L, Easton DF. The pathology of germline mutations in familial pancreatic carcinoma. J Natl familial breast cancer: predictive value of Cancer Inst. 2003 Feb 5;95(3):214-21 immunohistochemical markers estrogen receptor, progesterone receptor, HER-2, and p53 in patients with Hughes-Davies L, Huntsman D, Ruas M, Fuks F, Bye J, mutations in BRCA1 and BRCA2. J Clin Oncol. 2002 May Chin SF, Milner J, Brown LA, Hsu F, Gilks B, Nielsen T, 1;20(9):2310-8 Schulzer M, Chia S, Ragaz J, Cahn A, Linger L, Ozdag H, Cattaneo E, Jordanova ES, Schuuring E, Yu DS, Murphy KM, Brune KA, Griffin C, Sollenberger JE, Venkitaraman A, Ponder B, Doherty A, Aparicio S, Bentley Petersen GM, Bansal R, Hruban RH, Kern SE. Evaluation D, Theillet C, Ponting CP, Caldas C, Kouzarides T. EMSY of candidate genes MAP2K4, MADH4, ACVR1B, and links the BRCA2 pathway to sporadic breast and ovarian BRCA2 in familial pancreatic cancer: deleterious BRCA2 cancer. Cell. 2003 Nov 26;115(5):523-35 mutations in 17%. Cancer Res. 2002 Jul 1;62(13):3789-93 Jakubowska A, Scott R, Menkiszak J, Gronwald J, Byrski Pellegrini L, Yu DS, Lo T, Anand S, Lee M, Blundell TL, T, Huzarski T, Górski B, Cybulski C, Debniak T, Kowalska Venkitaraman AR. Insights into DNA recombination from E, Starzyńska T, Ławniczak M, Narod S, Lubinski J. A high the structure of a RAD51-BRCA2 complex. Nature. 2002 frequency of BRCA2 gene mutations in Polish families with Nov 21;420(6913):287-93 ovarian and stomach cancer. Eur J Hum Genet. 2003 Takata M, Tachiiri S, Fujimori A, Thompson LH, Miki Y, Dec;11(12):955-8 Hiraoka M, Takeda S, Yamazoe M. Conserved domains in Kutler DI, Singh B, Satagopan J, Batish SD, Berwick M, the chicken homologue of BRCA2. Oncogene. 2002 Feb Giampietro PF, Hanenberg H, Auerbach AD. A 20-year 7;21(7):1130-4 perspective on the International Fanconi Anemia Registry Tulinius H, Olafsdottir GH, Sigvaldason H, Arason A, (IFAR). Blood. 2003 Feb 15;101(4):1249-56 Barkardottir RB, Egilsson V, Ogmundsdottir HM, Lo T, Pellegrini L, Venkitaraman AR, Blundell TL. Tryggvadottir L, Gudlaugsdottir S, Eyfjord JE. The effect of Sequence fingerprints in BRCA2 and RAD51: implications a single BRCA2 mutation on cancer in Iceland. J Med for DNA repair and cancer. DNA Repair (Amst). 2003 Sep Genet. 2002 Jul;39(7):457-62 18;2(9):1015-28 Venkitaraman AR. Cancer susceptibility and the functions Offit K, Levran O, Mullaney B, Mah K, Nafa K, Batish SD, of BRCA1 and BRCA2. Cell. 2002 Jan 25;108(2):171-82 Diotti R, Schneider H, Deffenbaugh A, Scholl T, Proud VK, Warren M, Smith A, Partridge N, Masabanda J, Griffin D, Robson M, Norton L, Ellis N, Hanenberg H, Auerbach AD. Ashworth A. Structural analysis of the chicken BRCA2 Shared genetic susceptibility to breast cancer, brain gene facilitates identification of functional domains and tumors, and Fanconi anemia. J Natl Cancer Inst. 2003 Oct disease causing mutations. Hum Mol Genet. 2002 Apr 15;95(20):1548-51 1;11(7):841-51 Palacios J, Honrado E, Osorio A, Cazorla A, Sarrió D, Yang H, Jeffrey PD, Miller J, Kinnucan E, Sun Y, Thoma Barroso A, Rodríguez S, Cigudosa JC, Diez O, Alonso C, NH, Zheng N, Chen PL, Lee WH, Pavletich NP. BRCA2 Lerma E, Sánchez L, Rivas C, Benítez J. function in DNA binding and recombination from a BRCA2- Immunohistochemical characteristics defined by tissue DSS1-ssDNA structure. Science. 2002 Sep microarray of hereditary breast cancer not attributable to 13;297(5588):1837-48 BRCA1 or BRCA2 mutations: differences from breast carcinomas arising in BRCA1 and BRCA2 mutation Alter BP. Cancer in Fanconi anemia, 1927-2001. Cancer. carriers. Clin Cancer Res. 2003 Sep 1;9(10 Pt 1):3606-14 2003 Jan 15;97(2):425-40 Rosenberg PS, Greene MH, Alter BP. Cancer incidence in Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE, persons with Fanconi anemia. Blood. 2003 Feb Hopper JL, Loman N, Olsson H, Johannsson O, Borg A, 1;101(3):822-6 Pasini B, Radice P, Manoukian S, Eccles DM, Tang N, Olah E, Anton-Culver H, Warner E, Lubinski J, Gronwald J, Shin S, Verma IM. BRCA2 cooperates with histone Gorski B, Tulinius H, Thorlacius S, Eerola H, Nevanlinna acetyltransferases in androgen receptor-mediated H, Syrjäkoski K, Kallioniemi OP, Thompson D, Evans C, transcription. Proc Natl Acad Sci U S A. 2003 Jun Peto J, Lalloo F, Evans DG, Easton DF. Average risks of 10;100(12):7201-6 breast and ovarian cancer associated with BRCA1 or Daniels MJ, Wang Y, Lee M, Venkitaraman AR. Abnormal BRCA2 mutations detected in case Series unselected for cytokinesis in cells deficient in the breast cancer family history: a combined analysis of 22 studies. Am J susceptibility protein BRCA2. Science. 2004 Oct Hum Genet. 2003 May;72(5):1117-30 29;306(5697):876-9 Edwards SM, Kote-Jarai Z, Meitz J, Hamoudi R, Hope Q, Giordano SH, Cohen DS, Buzdar AU, Perkins G, Osin P, Jackson R, Southgate C, Singh R, Falconer A, Hortobagyi GN. Breast carcinoma in men: a population- Dearnaley DP, Ardern-Jones A, Murkin A, Dowe A, Kelly J, based study. Cancer. 2004 Jul 1;101(1):51-7 Williams S, Oram R, Stevens M, Teare DM, Ponder BA, Gayther SA, Easton DF, Eeles RA. Two percent of men Hirsch B, Shimamura A, Moreau L, Baldinger S, Hag- with early-onset prostate cancer harbor germline mutations alshiekh M, Bostrom B, Sencer S, D'Andrea AD. in the BRCA2 gene. Am J Hum Genet. 2003 Jan;72(1):1- Association of biallelic BRCA2/FANCD1 mutations with 12 spontaneous chromosomal instability and solid tumors of childhood. Blood. 2004 Apr 1;103(7):2554-9 Giusti RM, Rutter JL, Duray PH, Freedman LS, Konichezky M, Fisher-Fischbein J, Greene MH, Maslansky

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1133

BRCA2 (breast cancer 2, early onset) Guénard F, Durocher F

Lakhani SR, Manek S, Penault-Llorca F, Flanagan A, Couch FJ, Johnson MR, Rabe KG, Brune K, de Andrade Arnout L, Merrett S, McGuffog L, Steele D, Devilee P, Klijn M, Goggins M, Rothenmund H, Gallinger S, Klein A, JG, Meijers-Heijboer H, Radice P, Pilotti S, Nevanlinna H, Petersen GM, Hruban RH. The prevalence of BRCA2 Butzow R, Sobol H, Jacquemier J, Lyonet DS, Neuhausen mutations in familial pancreatic cancer. Cancer Epidemiol SL, Weber B, Wagner T, Winqvist R, Bignon YJ, Monti F, Biomarkers Prev. 2007 Feb;16(2):342-6 Schmitt F, Lenoir G, Seitz S, Hamman U, Pharoah P, Lane G, Ponder B, Bishop DT, Easton DF. Pathology of ovarian Esashi F, Galkin VE, Yu X, Egelman EH, West SC. cancers in BRCA1 and BRCA2 carriers. Clin Cancer Res. Stabilization of RAD51 nucleoprotein filaments by the C- 2004 Apr 1;10(7):2473-81 terminal region of BRCA2. Nat Struct Mol Biol. 2007 Jun;14(6):468-74 Narod SA, Foulkes WD. BRCA1 and BRCA2: 1994 and beyond. Nat Rev Cancer. 2004 Sep;4(9):665-76 Jacquemont C, Taniguchi T. The Fanconi anemia pathway and . BMC Biochem. 2007 Nov 22;8 Suppl 1:S10 Siaud N, Dray E, Gy I, Gérard E, Takvorian N, Doutriaux MP. Brca2 is involved in meiosis in Arabidopsis thaliana as King TA, Li W, Brogi E, Yee CJ, Gemignani ML, Olvera N, suggested by its interaction with Dmc1. EMBO J. 2004 Mar Levine DA, Norton L, Robson ME, Offit K, Borgen PI, Boyd 24;23(6):1392-401 J. Heterogenic loss of the wild-type BRCA allele in human breast tumorigenesis. Ann Surg Oncol. 2007 Wagner JE, Tolar J, Levran O, Scholl T, Deffenbaugh A, Sep;14(9):2510-8 Satagopan J, Ben-Porat L, Mah K, Batish SD, Kutler DI, MacMillan ML, Hanenberg H, Auerbach AD. Germline Palli D, Falchetti M, Masala G, Lupi R, Sera F, Saieva C, mutations in BRCA2: shared genetic susceptibility to D'Amico C, Ceroti M, Rizzolo P, Caligo MA, Zanna I, Ottini breast cancer, early onset leukemia, and Fanconi anemia. L. Association between the BRCA2 N372H variant and Blood. 2004 Apr 15;103(8):3226-9 male breast cancer risk: a population-based case-control study in Tuscany, Central Italy. BMC Cancer. 2007 Sep Friedenson B. BRCA1 and BRCA2 pathways and the risk 3;7:170 of cancers other than breast or ovarian. MedGenMed. 2005 Jun 29;7(2):60 Thorslund T, Esashi F, West SC. Interactions between human BRCA2 protein and the meiosis-specific Martin JS, Winkelmann N, Petalcorin MI, McIlwraith MJ, recombinase DMC1. EMBO J. 2007 Jun 20;26(12):2915- Boulton SJ. RAD-51-dependent and -independent roles of 22 a Caenorhabditis elegans BRCA2-related protein during DNA double-strand break repair. Mol Cell Biol. 2005 Tryggvadóttir L, Vidarsdóttir L, Thorgeirsson T, Jonasson Apr;25(8):3127-39 JG, Olafsdóttir EJ, Olafsdóttir GH, Rafnar T, Thorlacius S, Jonsson E, Eyfjord JE, Tulinius H. Prostate cancer Reid S, Renwick A, Seal S, Baskcomb L, Barfoot R, progression and survival in BRCA2 mutation carriers. J Jayatilake H, Pritchard-Jones K, Stratton MR, Ridolfi-Lüthy Natl Cancer Inst. 2007 Jun 20;99(12):929-35 A, Rahman N. Biallelic BRCA2 mutations are associated with multiple malignancies in childhood including familial Bahassi EM, Ovesen JL, Riesenberg AL, Bernstein WZ, Wilms tumour. J Med Genet. 2005 Feb;42(2):147-51 Hasty PE, Stambrook PJ. The checkpoint kinases Chk1 and Chk2 regulate the functional associations between van Asperen CJ, Brohet RM, Meijers-Heijboer EJ, hBRCA2 and Rad51 in response to DNA damage. Hoogerbrugge N, Verhoef S, Vasen HF, Ausems MG, Oncogene. 2008 Jun 26;27(28):3977-85 Menko FH, Gomez Garcia EB, Klijn JG, Hogervorst FB, van Houwelingen JC, van't Veer LJ, Rookus MA, van Mitra A, Fisher C, Foster CS, Jameson C, Barbachanno Y, Leeuwen FE. Cancer risks in BRCA2 families: estimates Bartlett J, Bancroft E, Doherty R, Kote-Jarai Z, Peock S, for sites other than breast and ovary. J Med Genet. 2005 Easton D, Eeles R. Prostate cancer in male BRCA1 and Sep;42(9):711-9 BRCA2 mutation carriers has a more aggressive phenotype. Br J Cancer. 2008 Jan 29;98(2):502-7 Boulton SJ. Cellular functions of the BRCA tumour- suppressor proteins. Biochem Soc Trans. 2006 Nov;34(Pt Narod SA, Neuhausen S, Vichodez G, Armel S, Lynch HT, 5):633-45 Ghadirian P, Cummings S, Olopade O, Stoppa-Lyonnet D, Couch F, Wagner T, Warner E, Foulkes WD, Saal H, Casilli F, Tournier I, Sinilnikova OM, Coulet F, Soubrier F, Weitzel J, Tulman A, Poll A, Nam R, Sun P, Danquah J, Houdayer C, Hardouin A, Berthet P, Sobol H, Bourdon V, Domchek S, Tung N, Ainsworth P, Horsman D, Kim-Sing Muller D, Fricker JP, et al. The contribution of germline C, Maugard C, Eisen A, Daly M, McKinnon W, Wood M, rearrangements to the spectrum of BRCA2 mutations. J Isaacs C, Gilchrist D, Karlan B, Nedelcu R, Meschino W, Med Genet. 2006 Sep;43(9):e49 Garber J, Pasini B, Manoukian S, Bellati C. Rapid progression of prostate cancer in men with a BRCA2 Li J, Zou C, Bai Y, Wazer DE, Band V, Gao Q. DSS1 is mutation. Br J Cancer. 2008 Jul 22;99(2):371-4 required for the stability of BRCA2. Oncogene. 2006 Feb 23;25(8):1186-94 Carreira A, Hilario J, Amitani I, Baskin RJ, Shivji MK, Venkitaraman AR, Kowalczykowski SC. The BRC repeats Mathew CG. Fanconi anaemia genes and susceptibility to of BRCA2 modulate the DNA-binding selectivity of RAD51. cancer. Oncogene. 2006 Sep 25;25(43):5875-84 Cell. 2009 Mar 20;136(6):1032-43 Taniguchi T, D'Andrea AD. Molecular pathogenesis of Shivji MK, Mukund SR, Rajendra E, Chen S, Short JM, Fanconi anemia: recent progress. Blood. 2006 Jun Savill J, Klenerman D, Venkitaraman AR. The BRC 1;107(11):4223-33 repeats of human BRCA2 differentially regulate RAD51 Agalliu I, Karlins E, Kwon EM, Iwasaki LM, Diamond A, binding on single- versus double-stranded DNA to Ostrander EA, Stanford JL. Rare germline mutations in the stimulate strand exchange. Proc Natl Acad Sci U S A. BRCA2 gene are associated with early-onset prostate 2009 Aug 11;106(32):13254-9 cancer. Br J Cancer. 2007 Sep 17;97(6):826-31 This article should be referenced as such: Agalliu I, Kwon EM, Zadory D, McIntosh L, Thompson J, Stanford JL, Ostrander EA. Germline mutations in the Guénard F, Durocher F. BRCA2 (breast cancer 2, early BRCA2 gene and susceptibility to hereditary prostate onset). Atlas Genet Cytogenet Oncol Haematol. 2010; cancer. Clin Cancer Res. 2007 Feb 1;13(3):839-43 14(12):1127-1134.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1134

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

FST (follistatin) Michael Grusch Medical University of Vienna, Department of Medicine I, Institute of Cancer Research, Borschkegasse 8a, A-1090 Vienna, Austria (MG)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/FSTID44477ch5q11.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI FSTID44477ch5q11.txt

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

rise to two main transcripts of 1122 bp (transcript Identity variant FST344) and 1386 bp (transcript variant Other names: FS FST317). The first exon encodes the signal peptide, HGNC (Hugo): FST the second exon the N-terminal domain and exons 3-5 each code for a follistatin module. Alternative Location: 5q11.2 splicing leads to usage of either exon 6A, which Local order codes for an acidic region in FST344 or exon 6B, RPS19P4 (ribosomal protein S19 pseudogene 4) - which contains two bases of the stop codon of FST - NDUFS4 (NADH dehydrogenase FST317 (Shimasaki et al., 1988). (ubiquinone) Fe-S protein 4). Transcription DNA/RNA Transcription of FST mRNA was shown to be stimulated by TGF beta and activin A via Smad Description proteins (Bartholin et al., 2002), which seems to be The human FST gene is comprised of six exons part of a negative feedback loop as FST can spanning 5329 bp on chromosome 5q11.2 and gives antagonize activin A (see below).

Intron/exon structure of the FST gene and domain architecture of FST proteins. 1, 2, 3, 4, 5, 6A, 6B: exon number; SP: signal peptide; NTD: N-terminal domain; FSD: follistatin domain; AT: acidic tail.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1135

FST (follistatin) Grusch M

Other factors and pathways that have been Hashimoto et al., 2002; Wada et al., 2004). demonstrated to stimulate follistatin gene Follistatin-bound activin is unable to initiate signal transcription are -releasing hormone transduction and consequently follistatin is a potent (GnRH) acting via cAMP and CREB (Winters et antagonist of physiological activin signals. Of the al., 2007), GLI2, a transcription factor activated by three follistatin domains present in all follistatin hedgehog signaling (Eichberger et al., 2008), isoforms, (Shimasaki et al., 1988) the first two, but dexamethasone (Hayashi et al., 2009), androgens not the third, are necessary for activin A binding and activators of wnt signaling (Willert et al., 2002; (Keutmann et al., 2004; Harrington et al., 2006). Yao et al., 2004; Singh et al., 2009). Repression of Aside from activins, follistatin also binds several the follistatin promoter in response to peroxisome bone morphogenetic proteins (BMP) including proliferator-activated receptor gamma was BMP2, BMP4, BMP6 and BMP7 (Iemura et al., mediated via SP1 (Necela et al., 2008). 1998; Glister et al., 2004). In 2004 it was shown that follistatin binds myostatin (also known as Protein growth and differentiation factor 8, GDF8) with high affinity and thereby is able to antagonize the Description inhibitory effect of myostatin on muscle growth Mature secreted follistatin protein exists in three (Amthor et al., 2004). main forms consisting of 288, 303, and 315 amino The functional significance of the interaction acids (Sugino et al., 1993). The FST344 transcript between follistatin and angiogenin, a pro- gives rise to a protein precursor of 344 amino acids, angiogenic factor unrelated to the TGF beta family, which results in the mature 315 amino acid form remains to be determined (Gao et al., 2007). The after removal of the signal peptide. A fraction of interaction of follistatin with heparin and heparan follistatin 315 is further converted to the 303 amino sulfates is isoform specific. Follistatin 288 binds to acid form by proteolytic cleavage at the C-terminus. heparan sulfates, whereas this binding is blocked by Signal peptide removal of FST317 leads to the the acidic tail of follistatin 315 (Sugino et al., mature 288 amino acid form of follistatin. All 1993). forms of follistatin contain three follistatin domains Knock-out mice for follistatin die within hours after (FSD) characterized by a conserved arrangement of birth and show multiple abnormalities of muscles, 10 cysteine residues. The N-terminal subdomains of skin and skeleton (Matzuk et al., 1995). Evidence the FSD have similarity with EGF-like modules, from many organs and tissues shows that whereas the C-terminal regions resemble the Kazal counterbalancing of signals from TGF beta family domains found in multiple serine protease members by follistatin is crucial for normal tissue inhibitors. The follistatin protein contains two development, architecture and function (de Kretser potential N-glycosilation sites on asparagines 124 et al., 2004; McDowall et al., 2008; Kreidl et al., and 288. 2009; Antsiferova et al., 2009). Due to the capability for efficient antagonization of Localisation signals from activin and myostatin, the therapeutic Follistatin is expressed in a wide variety of tissues application of follistatin has been discussed in and organs with the highest expression in the several clinical conditions involving elevated and testes (Phillips and de Kretser, 1998; activin/myostatin activity. Potential areas of Tortoriello et al., 2001). The signal peptide directs application include blocking increased activin the nascent protein to the secretory pathway and expression in inflammation (Phillips et al., 2009) follistatin has been detected in human serum and in and fibrotic disorders (Aoki and Kojima, 2007) and cell culture supernatants of multiple cell lines inhibition of myostatin in muscle diseases (Rodino- (Phillips and de Kretser, 1998). Among the Klapac et al., 2009). follistatin isoforms FST315 was secreted faster than FST288 (Schneyer et al., 2003) and due to the lack Homology of binding to cell-surface heparin-sulfated The follistatin module with its characteristic proteoglycans, a larger fraction of FST315 enters spacing of cysteines represents a conserved protein the circulation (Schneyer et al., 1996). domain. Follistatin modules are found in varying numbers in a wider set of secreted proteins Function including FSTL1, SPARC/osteonectin, or agrin Follistatin binds to several members of the TGF (Ullman and Perkins, 1997). Among these, beta family and blocks the interaction of these follistatin-like 3 (FSTL3, FLRG) shares a similar cytokines with their cognate receptors. Follistatin overall domain architecture with follistatin, but was first identified as a factor that could inhibit the harbors only two instead of three follistatin release of follicle-stimulating hormone from modules (Tortoriello et al., 2001). With respect to pituitary cells (Ueno et al., 1987). It binds activins activin binding ability, functional homology among A, B and AB with high affinity and was also follistatin domain-containing proteins is only found reported to bind activin E but not activin C between follistatin and FSTL3, whereas all other (Nakamura et al., 1990; Schneyer et al., 1994; follistatin family proteins have not been

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1136

FST (follistatin) Grusch M

demonstrated to bind proteins of the TGF beta structure of the human follistatin precursor and its genomic family (Tsuchida et al., 2000). Follistatin is also organization. Proc Natl Acad Sci U S A. 1988 Jun;85(12):4218-22 highly conserved between species with around 97% amino acid identity in human, mouse and rat. Nakamura T, Takio K, Eto Y, Shibai H, Titani K, Sugino H. Activin-binding protein from rat ovary is follistatin. Science. Implicated in 1990 Feb 16;247(4944):836-8 Sugino K, Kurosawa N, Nakamura T, Takio K, Shimasaki Malignancy S, Ling N, Titani K, Sugino H. Molecular heterogeneity of follistatin, an activin-binding protein. Higher affinity of the Note carboxyl-terminal truncated forms for heparan sulfate Overexpression of follistatin has been found in rat proteoglycans on the ovarian granulosa cell. J Biol Chem. 1993 Jul 25;268(21):15579-87 and mouse models of hepatocellular carcinoma (HCC) (Rossmanith et al., 2002; Fujiwara et al., Schneyer AL, Rzucidlo DA, Sluss PM, Crowley WF Jr. 2008) as well as in tumor tissue and serum of HCC Characterization of unique binding kinetics of follistatin and activin or inhibin in serum. Endocrinology. 1994 patients (Yuen et al., 2002; Grusch et al., 2006; Aug;135(2):667-74 Beale et al., 2008). However, follistatin had no Matzuk MM, Lu N, Vogel H, Sellheyer K, Roop DR, benefit as surveillance biomarker for HCC Bradley A. Multiple defects and perinatal death in mice development in patients with alcoholic and non- deficient in follistatin. Nature. 1995 Mar 23;374(6520):360- alcoholic liver disease (ALD and NAFLD) due to 3 the already elevated levels in the underlying liver Schneyer AL, Hall HA, Lambert-Messerlian G, Wang QF, pathologies (Beale et al., 2008). Follistatin Sluss P, Crowley WF Jr. Follistatin-activin complexes in overexpression was also demonstrated in human human serum and follicular fluid differ immunologically and melanoma cell lines (Stove et al., 2004) and has biochemically. Endocrinology. 1996 Jan;137(1):240-7 been suggested as candidate biomarker for lung Ullman CG, Perkins SJ. The Factor I and follistatin domain cancer (Planque et al., 2009). families: the return of a prodigal son. Biochem J. 1997 Sep 15;326 ( Pt 3):939-41 Endometriosis Iemura S, Yamamoto TS, Takagi C, Uchiyama H, Natsume Note T, Shimasaki S, Sugino H, Ueno N. Direct binding of Follistatin was increased in serum of women with follistatin to a complex of bone-morphogenetic protein and its receptor inhibits ventral and epidermal cell fates in early ovarian endometriosis and suggested as biomarker Xenopus embryo. Proc Natl Acad Sci U S A. 1998 Aug for endometrioma (Florio et al., 2009). 4;95(16):9337-42 Polycystic ovary syndrome Phillips DJ, de Kretser DM. Follistatin: a multifunctional regulatory protein. Front Neuroendocrinol. 1998 Note Oct;19(4):287-322 A genetic linkage analysis found evidence for Urbanek M, et al. Thirty-seven candidate genes for linkage of follistatin with polycystic ovary polycystic ovary syndrome: strongest evidence for linkage syndrome (PCOS) (Urbanek et al., 1999). Another is with follistatin. Proc Natl Acad Sci U S A. 1999 Jul study reported that the follistatin gene is not a 20;96(15):8573-8 susceptibility locus for PCOS but a single Tsuchida K, Arai KY, Kuramoto Y, Yamakawa N, nucleotide polymorphism of the gene may be Hasegawa Y, Sugino H. Identification and characterization involved in the hyperandrogenaemia of the disease of a novel follistatin-like protein as a binding protein for the (Jones et al., 2007). TGF-beta family. J Biol Chem. 2000 Dec 29;275(52):40788-96 Liver failure Tortoriello DV, Sidis Y, Holtzman DA, Holmes WE, Note Schneyer AL. Human follistatin-related protein: a structural homologue of follistatin with nuclear localization. Serum levels of follistatin and activin A were Endocrinology. 2001 Aug;142(8):3426-34 increased in patients with acute liver failure and it was suggested that a decreased follistatin/activin A Bartholin L, Maguer-Satta V, Hayette S, Martel S, Gadoux M, Corbo L, Magaud JP, Rimokh R. Transcription ratio in the blood may be an indicator for the activation of FLRG and follistatin by activin A, through severity of liver injury in hepatitis-related acute Smad proteins, participates in a negative feedback loop to liver disease (Hughes and Evans, 2003; Lin et al., modulate activin A function. Oncogene. 2002 Mar 2006). 28;21(14):2227-35 Hashimoto O, Tsuchida K, Ushiro Y, Hosoi Y, Hoshi N, References Sugino H, Hasegawa Y. cDNA cloning and expression of human activin betaE subunit. Mol Cell Endocrinol. 2002 Ueno N, Ling N, Ying SY, Esch F, Shimasaki S, Guillemin Aug 30;194(1-2):117-22 R. Isolation and partial characterization of follistatin: a Rossmanith W, et al. Follistatin overexpression in rodent single-chain Mr 35,000 monomeric protein that inhibits the liver tumors: a possible mechanism to overcome activin release of follicle-stimulating hormone. Proc Natl Acad Sci growth control. Mol Carcinog. 2002 Sep;35(1):1-5 U S A. 1987 Dec;84(23):8282-6 Willert J, Epping M, Pollack JR, Brown PO, Nusse R. A Shimasaki S, Koga M, Esch F, Cooksey K, Mercado M, transcriptional response to Wnt protein in human Koba A, Ueno N, Ying SY, Ling N, Guillemin R. Primary embryonic carcinoma cells. BMC Dev Biol. 2002 Jul 2;2:8

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1137

FST (follistatin) Grusch M

Yuen MF, Norris S, Evans LW, Langley PG, Hughes RD. Winters SJ, Ghooray D, Fujii Y, Moore JP Jr, Nevitt JR, Transforming growth factor-beta 1, activin and follistatin in Kakar SS. Transcriptional regulation of follistatin patients with hepatocellular carcinoma and patients with expression by GnRH in mouse gonadotroph cell lines: alcoholic cirrhosis. Scand J Gastroenterol. 2002 evidence for a role for cAMP signaling. Mol Cell Feb;37(2):233-8 Endocrinol. 2007 Jun 15;271(1-2):45-54 Hughes RD, Evans LW. Activin A and follistatin in acute Beale G, Chattopadhyay D, Gray J, Stewart S, Hudson M, liver failure. Eur J Gastroenterol Hepatol. 2003 Day C, Trerotoli P, Giannelli G, Manas D, Reeves H. AFP, Feb;15(2):127-31 PIVKAII, GP3, SCCA-1 and follisatin as surveillance biomarkers for hepatocellular cancer in non-alcoholic and Schneyer A, Schoen A, Quigg A, Sidis Y. Differential alcoholic fatty liver disease. BMC Cancer. 2008 Jul binding and neutralization of activins A and B by follistatin 18;8:200 and follistatin like-3 (FSTL-3/FSRP/FLRG). Endocrinology. 2003 May;144(5):1671-4 Eichberger T, et al. GLI2-specific transcriptional activation of the bone morphogenetic protein/activin antagonist Amthor H, Nicholas G, McKinnell I, Kemp CF, Sharma M, follistatin in human epidermal cells. J Biol Chem. 2008 May Kambadur R, Patel K. Follistatin complexes Myostatin and 2;283(18):12426-37 antagonises Myostatin-mediated inhibition of myogenesis. Dev Biol. 2004 Jun 1;270(1):19-30 Fujiwara M, Marusawa H, Wang HQ, Iwai A, Ikeuchi K, Imai Y, Kataoka A, Nukina N, Takahashi R, Chiba T. de Kretser DM, et al. The role of activin, follistatin and Parkin as a tumor suppressor gene for hepatocellular inhibin in testicular physiology. Mol Cell Endocrinol. 2004 carcinoma. Oncogene. 2008 Oct 9;27(46):6002-11 Oct 15;225(1-2):57-64 McDowall M, Edwards NM, Jahoda CA, Hynd PI. The role Glister C, Kemp CF, Knight PG. Bone morphogenetic of activins and follistatins in skin and hair follicle protein (BMP) ligands and receptors in bovine ovarian development and function. Cytokine Growth Factor Rev. follicle cells: actions of BMP-4, -6 and -7 on granulosa cells 2008 Oct-Dec;19(5-6):415-26 and differential modulation of Smad-1 phosphorylation by follistatin. Reproduction. 2004 Feb;127(2):239-54 Necela BM, Su W, Thompson EA. Peroxisome proliferator- activated receptor gamma down-regulates follistatin in Keutmann HT, Schneyer AL, Sidis Y. The role of follistatin intestinal epithelial cells through SP1. J Biol Chem. 2008 domains in follistatin biological action. Mol Endocrinol. Oct 31;283(44):29784-94 2004 Jan;18(1):228-40 Antsiferova M, Klatte JE, Bodó E, Paus R, Jorcano JL, Stove C, Vanrobaeys F, Devreese B, Van Beeumen J, Matzuk MM, Werner S, Kögel H. Keratinocyte-derived Mareel M, Bracke M. Melanoma cells secrete follistatin, an follistatin regulates epidermal homeostasis and wound antagonist of activin-mediated growth inhibition. repair. Lab Invest. 2009 Feb;89(2):131-41 Oncogene. 2004 Jul 8;23(31):5330-9 Florio P, et al. High serum follistatin levels in women with Wada W, Maeshima A, Zhang YQ, Hasegawa Y, Kuwano ovarian endometriosis. Hum Reprod. 2009 H, Kojima I. Assessment of the function of the betaC- Oct;24(10):2600-6 subunit of activin in cultured hepatocytes. Am J Physiol Endocrinol Metab. 2004 Aug;287(2):E247-54 Hayashi K, Yamaguchi T, Yano S, Kanazawa I, Yamauchi M, Yamamoto M, Sugimoto T. BMP/Wnt antagonists are Yao HH, Matzuk MM, Jorgez CJ, Menke DB, Page DC, upregulated by dexamethasone in osteoblasts and Swain A, Capel B. Follistatin operates downstream of reversed by alendronate and PTH: potential therapeutic Wnt4 in mammalian ovary organogenesis. Dev Dyn. 2004 targets for glucocorticoid-induced osteoporosis. Biochem Jun;230(2):210-5 Biophys Res Commun. 2009 Feb 6;379(2):261-6 Grusch M, Drucker C, Peter-Vörösmarty B, Erlach N, Kreidl E, et al. Activins and follistatins: Emerging roles in Lackner A, Losert A, Macheiner D, Schneider WJ, liver physiology and cancer. World J Hepatol Rev 2009 Oct Hermann M, Groome NP, Parzefall W, Berger W, Grasl- 31; 1(1): 17-27. (REVIEW) Kraupp B, Schulte-Hermann R. Deregulation of the activin/follistatin system in hepatocarcinogenesis. J Phillips DJ, de Kretser DM, Hedger MP. Activin and related Hepatol. 2006 Nov;45(5):673-80 proteins in inflammation: not just interested bystanders. Cytokine Growth Factor Rev. 2009 Apr;20(2):153-64 Harrington AE, Morris-Triggs SA, Ruotolo BT, Robinson CV, Ohnuma S, Hyvönen M. Structural basis for the Planque C, Kulasingam V, Smith CR, Reckamp K, inhibition of activin signalling by follistatin. EMBO J. 2006 Goodglick L, Diamandis EP. Identification of five candidate Mar 8;25(5):1035-45 lung cancer biomarkers by proteomics analysis of conditioned media of four lung cancer cell lines. Mol Cell Lin SD, Kawakami T, Ushio A, Sato A, Sato S, Iwai M, Proteomics. 2009 Dec;8(12):2746-58 Endo R, Takikawa Y, Suzuki K. Ratio of circulating follistatin and activin A reflects the severity of acute liver Rodino-Klapac LR, Haidet AM, Kota J, Handy C, Kaspar injury and prognosis in patients with acute liver failure. J BK, Mendell JR. Inhibition of myostatin with emphasis on Gastroenterol Hepatol. 2006 Feb;21(2):374-80 follistatin as a therapy for muscle disease. Muscle Nerve. 2009 Mar;39(3):283-96 Aoki F, Kojima I. Therapeutic potential of follistatin to promote tissue regeneration and prevent tissue fibrosis. Singh R, Bhasin S, Braga M, Artaza JN, Pervin S, Taylor Endocr J. 2007;54(6):849-54 WE, Krishnan V, Sinha SK, Rajavashisth TB, Jasuja R. Regulation of myogenic differentiation by androgens: cross Gao X, Hu H, Zhu J, Xu Z. Identification and talk between androgen receptor/ beta-catenin and characterization of follistatin as a novel angiogenin-binding follistatin/transforming growth factor-beta signaling protein. FEBS Lett. 2007 Nov 27;581(28):5505-10 pathways. Endocrinology. 2009 Mar;150(3):1259-68 Jones MR, Wilson SG, Mullin BH, Mead R, Watts GF, Stuckey BG. Polymorphism of the follistatin gene in This article should be referenced as such: polycystic ovary syndrome. Mol Hum Reprod. 2007 Grusch M. FST (follistatin). Atlas Genet Cytogenet Oncol Apr;13(4):237-41 Haematol. 2010; 14(12):1135-1138.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1138

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

GATA6 (GATA binding protein 6) Rosalyn M Adam, Joshua R Mauney Urological Diseases Research Center, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA (RMA, JRM)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/GATA6ID40690ch18q11.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI GATA6ID40690ch18q11.txt

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

GATA6 genes contain two alternative non-coding Identity upstream exons, transcribed from two distinct Other names: GATA-6 promoters (Brewer et al., 1999), similar to other HGNC (Hugo): GATA6 GATA family members. The non-coding exons possess regulatory capability and may act to Location: 18q11.2 promote transcription. Two isoforms of GATA6 are Local order: GATA-6 is flanked in the direction of expressed from two distinct open reading frames the centromere by: and distinct initiator Met codons as a result of leaky LOC100128893, hypothetical protein ribosome scanning. LOC100128893 - RNU7-17P, RNA U7 small There are no apparent differences in the amounts or nuclear 17 pseudogene - LOC100287318 - sites of expression of the two transcripts that result RPL34P32, ribosomal protein L34 pseudogene 32 - from initiation at different Met codons. MIB1, mindbomb homolog 1 - MIR1-2 - MIR133A1. Protein GATA6 is flanked in the direction of the telomere by: Description CTAGE1, cutaneous T-cell lymphoma-associated The GATA6 protein products that result from antigen 1 - RPS4P18, ribosomal protein S4X different initiation codons comprise a long isoform pseudogene 18 - RBBP8, retinoblastoma binding of 595 aa (64 kDa) and a short isoform of 449 aa protein 8 - CABLES1, Cdk5 and Abl enzyme (52 kDa). substrate 1 - C18orf45, open Both isoforms possess an N-terminal reading frame 45 - RIOK3, RIO kinase 3. transactivation domain and two zinc finger Note: GATA6 is one of a family of 6 related domains, all of which are essential for activity GATA binding proteins. All six proteins possess (Takeda et al., 2004). The two isoforms display zinc finger-type DNA binding domains and act as different transactivation potential on GATA6- transcription factors. dependent promoters with long GATA6 showing higher activity than short GATA6. DNA/RNA Expression Description GATA6 is expressed predominantly in tissues of mesodermal and endodermal origin. In early Genomic DNA encoding GATA6 encompasses development, high levels are detected in the 33088 bp on the long arm of chromosome 18. The precardiac mesoderm, embryonic heart tube and gene is encoded on the plus (forward) strand. primitive gut. As development proceeds GATA6 Transcription expression is observed in vascular smooth muscle The pre-mRNA comprises 7 exons, one of which is cells, the developing airways, urogenital ridge and non-coding, and 6 introns. The mouse and human bladder (Morrisey et al., 1996).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1139

GATA6 (GATA binding protein 6) Adam RM, Mauney JR

Localisation factors and signaling molecules, including FOG factors, GATA4 (Xin et al., 2006; Zhao et al., Nuclear. 2008), Tbx5 (Maitra et al., 2009), members of the Function Nkx2 family (Peterkin et al., 2003) and Wnt family GATA6 binds to a 5'-(T/A)GATA(A/G)-3' proteins. The complexity of these interactions is consensus sequence in the promoters of target exemplified by the functional cooperation of Wnt2 genes to regulate their transcription. GATA6 is and GATA6 in regulating heart development. In post-translationallly modified by MEK/Erk- this case, GATA6 not only regulates Wnt2 dependent phosphorylation at Ser120 (S266 in long transcription during heart development through GATA6). Ser120Ala mutation abolished GATA6 direct binding to the Wnt2 promoter DNA-binding activity and GATA6-mediated Nox1 (Alexandrovich et al., 2006), but is itself regulated promoter activation, and also suppressed growth of by a Wnt2-dependent mechanism, since GATA6 CaCo-2 colon carcinoma cells (Adachi et al., 2008). expression is markedly reduced in Wnt2-null mice GATA6 activity is also regulated through (Tian et al., 2010). interaction with members of the Friend of GATA GATA6 has also been implicated in regulating (FOG) family of proteins. Two FOG proteins have development of other organs including the lung and been identified in mice and humans, FOG-1 and pancreas. In the lung, GATA6 has been shown to FOG-2, and their interaction with GATA factors regulate specification, differentiation and can promote or inhibit GATA activity, depending maturation of the pulmonary epithelium as well as on context (Cantor and Orkin, 2005). branching morphogenesis (Keijzer et al., 2001; GATA6 is essential for normal development, since Yang et al., 2002; Liu et al., 2002; Zhang et al., genetic knockout in mice leads to embryonic 2008). Inhibition of GATA6 at E6.0 prevented lethality as early as E6.5. The underlying defect in alveolar maturation and also diminished expression GATA6-null mice was determined to be a failure of of surfactant proteins required for normal endoderm differentiation resulting in attenuated pulmonary function. In the pancreas, GATA6 is co- expression of GATA6 target genes including expressed with GATA4 in the epithelium early in GATA4, HNF3beta and HNF4 (Morrisey et al., development, but as development progresses is 1998). GATA6 was shown subsequently to be expressed only in endocrine cells. Ablation of essential for early extraembryonic development GATA6 function using a dominant inhibitory (Koutsourakis et al., 1999). Partial rescue of engrailed fusion protein strategy led to a reduction GATA6-deficient embryos by tetraploid embryo or complete loss of pancreatic tissue, consistent complementation demonstrated additional functions with a critical role for GATA6 in pancreatic for GATA6 in liver development. The early development (Decker et al., 2006). lethality in GATA6-null embryos could be GATA6 has also been implicated in postnatal overcome by providing wild type extraembryonic maintenance of the differentiated phenotype in endoderm and allowed embryos to proceed through various tissues including bladder smooth muscle gastrulation. However, although hepatic (Kanematsu, 2007), gut mucosa (Fang, 2006) and specification occurred normally in rescued GATA6- airway epithelium (Zhang, 2008). /- embryos, normal differentiation did not occur and Homology hepatic development arrested at E10.5 (Zhao et al., GATA6 shares homology with the other 5 GATA 2005). factors, all of which are evolutionarily conserved Early development of other organ systems was across multiple species. All 6 GATA factors unaffected in rescued GATA6-null embryos, possess two zinc fingers of the Cys-X -Cys-X - including the heart and vasculature. Interestingly, 2 17 Cys-X -Cys configuration. The C-terminal zinc conditional deletion of GATA6 using SM22alpha 2 finger mediates high affinity DNA binding and the promoter-driven Cre recombinase led to perinatal N-terminal zinc finger stabilizes the interaction lethality as a result of cardiovascular defects with DNA. emerging later in embryonic development. In that analysis, the underlying mechanism was determined to be diminished expression of the vascular and Mutations neuronal guidance molecule semaphorin 3C, a Germinal direct target of GATA6 (Lepore et al., 2006). Consistent with GATA6-dependent regulation of None known. Sema3C in mice, mutations in GATA6 were found Somatic to cause cardiac outflow tract defects in humans by Two mutations in GATA6 were identified in dysregulating semaphorin-dependent signaling patients with persistent truncus arteriosus, as (Kodo et al., 2009). In general, GATA6 does not follows (Kodo et al., 2009). act alone in regulating developmental processes, but GATA6-E486del resulted in conversion of P489 to rather achieves its effects through physical and a stop codon, disruption of the nuclear localization functional interaction with other transcription signal and truncation of the C-terminus by 100 aa.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1140

GATA6 (GATA binding protein 6) Adam RM, Mauney JR

The encoded protein showed abnormal nuclear Expression of GATA4 and GATA6 was shown to localization, no transcriptional activity against atrial correlate with specific histological subtypes of natriuretic factor and WNT2 promoters and was ovarian cancer. In particular, although expression of dominant negative. both factors was lost in the over 80% of GATA6-N466H contained a point mutation in the endometrioid, clear cell and serous tumors, GATA4 C-terminal zinc finger domain. Despite normal and GATA6 expression persisted in mucinous nuclear localization, the encoded protein had no carcinomas (Cai et al., 2009). Loss of GATA factor transcriptional activity against atrial natriuretic expression preceded neoplastic transformation, factor and WNT2 promoters. consistent with an important role for these proteins in tumor development. The mechanism underlying Implicated in loss of GATA6 and GATA4 expression in ovarian cancer cell lines was demonstrated to be histone Pancreatic cancer, pancreatobiliary deacetylation at the GATA factor promoter regions. cancer Inhibition of histone deacetylase activity with Disease trichostatin A restored GATA6 and GATA4 Genomic profiling of pancreatic and bile duct expression in cell lines (Caslini et al., 2006). cancers revealed focal amplification at 18q11.2 that Prognosis encoded GATA6. Amplification led to Loss of GATA6 expression precedes neoplastic overexpression of GATA6 at both mRNA and transformation in ovarian surface epithelia (Cai et protein levels in nearly 50% of tumor samples, al., 2009) and is correlated with loss of markers of whereas no normal pancreatic tissues showed differentiated epithelia (Capo-chichi et al., 2003). overexpression (Kwei et al., 2008; Fu et al., 2008). Although a majority of ovarian carcinomas retained Consistent with an oncogenic role for GATA6 in GATA4 expression, most had either aberrantly pancreatic cancer, RNAi-mediated silencing in localized or absent GATA6 expression. pancreatic cancer cell lines in which GATA6 was Cytoplasmic expression of GATA6 showed a amplified decreased cell cycle transit, growth and correlation with overall survival, but this clonogenic ability (Kwei et al., 2008). Conversely association did not reach statistical significance forced expression of GATA6 in a pancreatic cancer (McEachin, 2008). cell line stimulated anchorage-independent growth Gastrointestinal cancer and proliferation (Fu et al., 2008). Disease Prognosis Expression of GATA6 has been linked, both GATA6 silencing by RNAi in pancreatic cancer positively and negatively, to development of cells in vitro reduced proliferation, cell cycle transit gastrointestinal tract tumors. and colony formation, whereas forced overexpression promoted colony formation in soft Prognosis agar and enhanced proliferation, consistent with a GATA6 expression was found to be decreased in role for GATA6 in driving the tumorigenic colon carcinoma compared to normal intestinal phenotype. tissue or benign intestinal lesions (Haveri et al., 2008), which showed robust expression, especially Cytogenetics in cells with proliferative capacity. Conversely Focal amplification of the locus encoding GATA6 GATA6 was reported to be overexpressed in human at 18q11.2 was identified by array-based genomic colon cancer cells, where it contributes to silencing profiling and validated by fluorescence in situ of 15-lipoxygenase-1 (Shureiqi et al., 2007). The hybridization, quantitative PCR, biological significance of this discrepancy in immunohistochemical analysis and GATA6 expression between colon cancer cells and immunoblotting. tissues has not been determined. Expression Ovarian cancer profiling of Barrett's esophagus and Disease adenocarcinoma to identify genes whose expression Consistent with their expression in the mouse correlated with disease progression revealed ovary, GATA6, GATA4 and FOG2 are also changes in GATA6 expression among other genes, expressed in human ovary and in tumors derived consistent with upregulation of GATA6 in the from granulosa and thecal cells (Laitinen, 2000). transition from normal esophageal epithelium to Under normal conditions, both GATA4 and carcinoma (Kimchi et al., 2005). GATA6 are robustly expressed in ovarian surface Lung cancer epithelial cells. However, in a majority of ovarian Disease carcinomas, GATA6 is lost or mislocalized to the Despite substantial evidence linking GATA6 to cytoplasm (Capo-chichi et al., 2003; McEachin et pulmonary development, only one study has al., 2008), leading to irreversible epithelial investigated the potential role of GATA6 in lung dedifferentiation (Capo-chichi et al., 2003). cancer. Specifically, expression of GATA6 was

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1141

GATA6 (GATA binding protein 6) Adam RM, Mauney JR

evaluated in malignant mesothelioma and pleural metastases of lung adenocarcinomas and staining References patterns correlated with biological and clinical Morrisey EE, Ip HS, Lu MM, Parmacek MS. GATA-6: a outcomes. Nuclear immunoreactivity for GATA-6 zinc finger transcription factor that is expressed in multiple cell lineages derived from lateral mesoderm. Dev Biol. was stronger and more frequent in malignant 1996 Jul 10;177(1):309-22 mesothelioma than in metastatic lung adenocarcinoma (Lindholm et al., 2009). However, Morrisey EE, Tang Z, Sigrist K, Lu MM, Jiang F, Ip HS, Parmacek MS. GATA6 regulates HNF4 and is required for no relationship was found between GATA6 differentiation of visceral endoderm in the mouse embryo. expression and growth or apoptotic endpoints. Genes Dev. 1998 Nov 15;12(22):3579-90 Prognosis Brewer A, Gove C, Davies A, McNulty C, Barrow D, Prognosis was better in malignant mesothelioma Koutsourakis M, Farzaneh F, Pizzey J, Bomford A, Patient patients whose tumors expressed GATA-6 R. The human and mouse GATA-6 genes utilize two promoters and two initiation codons. J Biol Chem. 1999 compared to those whose tumors had no GATA-6 Dec 31;274(53):38004-16 expression, and the relationship was highly statistically significant. Kiiveri S, Siltanen S, Rahman N, Bielinska M, Lehto VP, Huhtaniemi IT, Muglia LJ, Wilson DB, Heikinheimo M. Reciprocal changes in the expression of transcription Adrenocortical cancer factors GATA-4 and GATA-6 accompany adrenocortical Disease tumorigenesis in mice and humans. Mol Med. 1999 GATA6 has been implicated in development of the Jul;5(7):490-501 normal . GATA6 mRNA, although Koutsourakis M, Langeveld A, Patient R, Beddington R, expressed in the normal adrenal cortex was found to Grosveld F. The transcription factor GATA6 is essential for early extraembryonic development. Development. 1999 be absent from experimental mouse adrenocortical May;126(9):723-32 tumors, whereas GATA-4 showed the opposite Laitinen MP, Anttonen M, Ketola I, Wilson DB, Ritvos O, pattern (Kiiveri, 1999; Rahman et al., 2001). Butzow R, Heikinheimo M. Transcription factors GATA-4 GATA-6 expression was also decreased in human and GATA-6 and a GATA family cofactor, FOG-2, are adrenocortical carcinomas compared to normal expressed in human ovary and sex cord-derived ovarian adrenal tissue and adenomas (Kiiveri et al., 2004). tumors. J Clin Endocrinol Metab. 2000 Sep;85(9):3476-83 The physiologic relevance of altered GATA6 Keijzer R, van Tuyl M, Meijers C, Post M, Tibboel D, expression in adrenocortical tumorigenesis has not Grosveld F, Koutsourakis M. The transcription factor yet been elucidated. However, based on expression GATA6 is essential for branching morphogenesis and epithelial cell differentiation during fetal pulmonary of the CDK inhibitor p21 and proliferation marker development. Development. 2001 Feb;128(4):503-11 Ki67, GATA-6 expression in adrenocortical tumors does not appear to be linked to regulation of cell Rahman NA, Kiiveri S, Siltanen S, Levallet J, Kero J, Lensu T, Wilson DB, Heikinheimo MT, Huhtaniemi IT. proliferation. Adrenocortical tumorigenesis in transgenic mice: the role Prognosis of receptor and transcription factors GATA-4 and GATA-61. Reprod Biol. 2001 Jul;1(1):5-9 The prognostic significance of GATA-6 in adrenocortical tumors has not been determined. Liu C, Morrisey EE, Whitsett JA. GATA-6 is required for maturation of the lung in late gestation. Am J Physiol Lung Germ cell tumors Cell Mol Physiol. 2002 Aug;283(2):L468-75 Disease Yang H, Lu MM, Zhang L, Whitsett JA, Morrisey EE. GATA6 regulates differentiation of distal lung epithelium. Germ cell tumors comprise a heterogeneous group Development. 2002 May;129(9):2233-46 of lesions, including teratomas, yolk sac tumors and Capo-chichi CD, Roland IH, Vanderveer L, Bao R, embryonal carcinoma. Using in situ hybridization Yamagata T, Hirai H, Cohen C, Hamilton TC, Godwin AK, and immunohistochemical staining, GATA6 was Xu XX. Anomalous expression of epithelial differentiation- evaluated in pediatric germ cell tumors and was determining GATA factors in ovarian tumorigenesis. found to be expressed in a majority of yolk sac Cancer Res. 2003 Aug 15;63(16):4967-77 tumors. GATA6 expression was also evident in Peterkin T, Gibson A, Patient R. GATA-6 maintains BMP-4 distinct cell types comprising teratomas, including and Nkx2 expression during cardiomyocyte precursor gut and airway epithelia (Siltanen et al., 2003), but maturation. EMBO J. 2003 Aug 15;22(16):4260-73 was variable in carcinoma in situ of the testis and Siltanen S, Heikkilä P, Bielinska M, Wilson DB, absent from embryonal carcinomas and Heikinheimo M. Transcription factor GATA-6 is expressed choriocarcinomas (Salonen et al., 2010). in malignant endoderm of pediatric yolk sac tumors and in teratomas. Pediatr Res. 2003 Oct;54(4):542-6 Prognosis Takeda M, Obayashi K, Kobayashi A, Maeda M. A unique The prognostic role of GATA6 in germ cell tumors role of an amino terminal 16-residue region of long-type is unknown. GATA-6. J Biochem. 2004 May;135(5):639-50 Cantor AB, Orkin SH. Coregulation of GATA factors by the

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1142

GATA6 (GATA binding protein 6) Adam RM, Mauney JR

Friend of GATA (FOG) family of multitype zinc finger Haveri H, Westerholm-Ormio M, Lindfors K, Mäki M, proteins. Semin Cell Dev Biol. 2005 Feb;16(1):117-28 Savilahti E, Andersson LC, Heikinheimo M. Transcription factors GATA-4 and GATA-6 in normal and neoplastic Kiiveri S, Liu J, Arola J, Heikkilä P, Kuulasmaa T, human gastrointestinal mucosa. BMC Gastroenterol. 2008 Lehtonen E, Voutilainen R, Heikinheimo M. Transcription Apr 11;8:9 factors GATA-6, SF-1, and cell proliferation in human adrenocortical tumors. Mol Cell Endocrinol. 2005 Apr Kwei KA, Bashyam MD, Kao J, Ratheesh R, Reddy EC, 15;233(1-2):47-56 Kim YH, Montgomery K, Giacomini CP, Choi YL, Chatterjee S, Karikari CA, Salari K, Wang P, Hernandez- Kimchi ET, Posner MC, Park JO, Darga TE, Kocherginsky Boussard T, Swarnalata G, van de Rijn M, Maitra A, M, Karrison T, Hart J, Smith KD, Mezhir JJ, Weichselbaum Pollack JR. Genomic profiling identifies GATA6 as a RR, Khodarev NN. Progression of Barrett's metaplasia to candidate oncogene amplified in pancreatobiliary cancer. adenocarcinoma is associated with the suppression of the PLoS Genet. 2008 May 23;4(5):e1000081 transcriptional programs of epidermal differentiation. Cancer Res. 2005 Apr 15;65(8):3146-54 McEachin MD, Xu XX, Santoianni RA, Lawson D, Cotsonis G, Cohen C. GATA-4 and GATA-6 expression in human Zhao R, Watt AJ, Li J, Luebke-Wheeler J, Morrisey EE, ovarian surface epithelial carcinoma. Appl Duncan SA. GATA6 is essential for embryonic Immunohistochem Mol Morphol. 2008 Mar;16(2):153-8 development of the liver but dispensable for early heart formation. Mol Cell Biol. 2005 Apr;25(7):2622-31 Zhang Y, Goss AM, Cohen ED, Kadzik R, Lepore JJ, Muthukumaraswamy K, Yang J, DeMayo FJ, Whitsett JA, Alexandrovich A, Arno M, Patient RK, Shah AM, Pizzey Parmacek MS, Morrisey EE. A Gata6-Wnt pathway JA, Brewer AC. Wnt2 is a direct downstream target of required for epithelial stem cell development and airway GATA6 during early cardiogenesis. Mech Dev. 2006 regeneration. Nat Genet. 2008 Jul;40(7):862-70 Apr;123(4):297-311 Zhao R, Watt AJ, Battle MA, Li J, Bondow BJ, Duncan SA. Caslini C, Capo-chichi CD, Roland IH, Nicolas E, Yeung Loss of both GATA4 and GATA6 blocks cardiac myocyte AT, Xu XX. Histone modifications silence the GATA differentiation and results in acardia in mice. Dev Biol. transcription factor genes in ovarian cancer. Oncogene. 2008 May 15;317(2):614-9 2006 Aug 31;25(39):5446-61 Cai KQ, Caslini C, Capo-chichi CD, Slater C, Smith ER, Decker K, Goldman DC, Grasch CL, Sussel L. Gata6 is an Wu H, Klein-Szanto AJ, Godwin AK, Xu XX. Loss of important regulator of mouse pancreas development. Dev GATA4 and GATA6 expression specifies ovarian cancer Biol. 2006 Oct 15;298(2):415-29 histological subtypes and precedes neoplastic Fang R, Olds LC, Sibley E. Spatio-temporal patterns of transformation of ovarian surface epithelia. PLoS One. intestine-specific transcription factor expression during 2009 Jul 31;4(7):e6454 postnatal mouse gut development. Gene Expr Patterns. Kodo K, Nishizawa T, Furutani M, Arai S, Yamamura E, 2006 Apr;6(4):426-32 Joo K, Takahashi T, Matsuoka R, Yamagishi H. GATA6 Lepore JJ, Mericko PA, Cheng L, Lu MM, Morrisey EE, mutations cause human cardiac outflow tract defects by Parmacek MS. GATA-6 regulates semaphorin 3C and is disrupting semaphorin-plexin signaling. Proc Natl Acad Sci required in cardiac neural crest for cardiovascular U S A. 2009 Aug 18;106(33):13933-8 morphogenesis. J Clin Invest. 2006 Apr;116(4):929-39 Lindholm PM, Soini Y, Myllärniemi M, Knuutila S, Xin M, Davis CA, Molkentin JD, Lien CL, Duncan SA, Heikinheimo M, Kinnula VL, Salmenkivi K. Expression of Richardson JA, Olson EN. A threshold of GATA4 and GATA-6 transcription factor in pleural malignant GATA6 expression is required for cardiovascular mesothelioma and metastatic pulmonary adenocarcinoma. development. Proc Natl Acad Sci U S A. 2006 Jul J Clin Pathol. 2009 Apr;62(4):339-44 25;103(30):11189-94 Maitra M, Schluterman MK, Nichols HA, Richardson JA, Lo Kanematsu A, Ramachandran A, Adam RM. GATA-6 CW, Srivastava D, Garg V. Interaction of Gata4 and Gata6 mediates human bladder smooth muscle differentiation: with Tbx5 is critical for normal cardiac development. Dev involvement of a novel enhancer element in regulating Biol. 2009 Feb 15;326(2):368-77 alpha-smooth muscle actin gene expression. Am J Physiol Salonen J, Rajpert-De Meyts E, Mannisto S, Nielsen JE, Cell Physiol. 2007 Sep;293(3):C1093-102 Graem N, Toppari J, Heikinheimo M. Differential Shureiqi I, Zuo X, Broaddus R, Wu Y, Guan B, Morris JS, developmental expression of transcription factors GATA-4 Lippman SM. The transcription factor GATA-6 is and GATA-6, their cofactor FOG-2 and downstream target overexpressed in vivo and contributes to silencing 15-LOX- genes in testicular carcinoma in situ and germ cell tumors. 1 in vitro in human colon cancer. FASEB J. 2007 Eur J Endocrinol. 2010 Mar;162(3):625-31 Mar;21(3):743-53 Tian Y, Yuan L, Goss AM, Wang T, Yang J, Lepore JJ, Adachi Y, Shibai Y, Mitsushita J, Shang WH, Hirose K, Zhou D, Schwartz RJ, Patel V, Cohen ED, Morrisey EE. Kamata T. Oncogenic Ras upregulates NADPH oxidase 1 Characterization and in vivo pharmacological rescue of a gene expression through MEK-ERK-dependent Wnt2-Gata6 pathway required for cardiac inflow tract phosphorylation of GATA-6. Oncogene. 2008 Aug development. Dev Cell. 2010 Feb 16;18(2):275-87 21;27(36):4921-32 This article should be referenced as such: Fu B, Luo M, Lakkur S, Lucito R, Iacobuzio-Donahue CA. Frequent genomic copy number gain and overexpression Adam RM, Mauney JR. GATA6 (GATA binding protein 6). of GATA-6 in pancreatic carcinoma. Cancer Biol Ther. Atlas Genet Cytogenet Oncol Haematol. 2010; 2008 Oct;7(10):1593-601 14(12):1139-1143.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1143

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

HIPK2 (homeodomain interacting protein kinase 2) Dirk Sombroek, Thomas G Hofmann Deutsches Krebsforschungszentrum (dkfz.), Cellular Senescence Unit A210, Cell and Tumor Biology Program, Heidelberg, Germany (DS, TGH)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/HIPK2ID40824ch7q34.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI HIPK2ID40824ch7q34.txt

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

HIPK2-002 [ENST00000428878]; 3969 bp linear Identity mRNA; 1171 amino acids, Other names: DKFZp686K02111, FLJ23711, HIPK2-201 [ENST00000263551]; 14953 bp linear hHIPk2, PRO0593 mRNA; 1198 amino acids, HGNC (Hugo): HIPK2 HIPK2-202 [ENST00000342645]; 2757 bp linear mRNA; 918 amino acids. Location: 7q34 Pseudogene DNA/RNA Nothing known. Description Protein Zhang et al. (2005) reported 13 exons that span around 60 kb; however, up to 15 exons are listed in Description different databases. HIPK2 is a protein kinase of 1198 amino acids (131 kDa); posttranslational modifications: Transcription phosphorylation, ubiquitination, sumoylation at Around 15 kb mRNA (full-length); 3594 bp open K25, caspase cleavage at D916 and D977. reading frame. Contains several motifs and domains (from N- to C- At least two alternative transcripts. terminus): a nuclear localisation signal (NLS)1 (97- Entrez Nucleotide: 157), a kinase domain (192-520), an interaction [NM_022740.4] Homo sapiens HIPK2, transcript domain for homeodomain transcription factors variant 1; 15245 bp linear mRNA; full-length (583-798), a NLS2 (780-840) and a NLS3 within a isoform, speckle-retention signal (SRS) (860-967), a PEST [NM_001113239.2] Homo sapiens HIPK2, sequence (839-934) and an autoinhibitory domain transcript variant 2; 15164 bp linear mRNA; this (935-1050). variant lacks an internal segment in the CDS, the resulting isoform is shorter. Expression UniProtHB/Swiss-Prot [Q9H2X6]: HIPK2 is ubiquitously expressed (high mRNA [Q9H2X6-1] full-length isoform (1), levels in neuronal tissues, heart, muscle and [Q9H2X6-2] isoform (2), kidney); but barely detectable at protein levels in [Q9H2X6-3] isoform (3). unstressed cells. Protein levels increase upon Ensemble Gene [ENSG00000064393]; 4 genotoxic stress. transcripts: Localisation HIPK2-001 [ENST00000406875]; 15049 bp linear Mainly nuclear localisation, in nuclear bodies; but mRNA; 1198 amino acids, also found in nucleoplasm and cytoplasm.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1144

HIPK2 (homeodomain interacting protein kinase 2) Sombroek D, Hofmann TG

Function Overexpression of HMGA1 was reported to inhibit p53-mediated apoptosis by removing HIPK2 from HIPK2 is a potential tumour suppressor; in vivo the nucleus and retaining it in the cytoplasm. data suggest at least a role as an haploinsufficient Observations could be correlated with in vivo data, tumour suppressor in the skin of mice. at least in breast cancer. WT p53-expressing breast HIPK2 is a protein kinase that interacts with carcinomas showed a low spontaneous apoptotic numerous transcription factors (such as p53, index in case of HIPK2-relocalisation (Pierantoni et AML1(RUNX1), PAX6, c-MYB or NK3) as well al., 2007). as transcriptional regulators (such as CBP, p300, Groucho, CtBP, HMGA1 or Smads). In this way Epithelial tumours (with altered beta4 HIPK2 can activate or repress transcription and integrin expression) thereby influence differentiation, development and Oncogenesis the DNA damage response. HIPK2 was reported to repress beta4 integrin HIPK2 is an unstable protein in unstressed cells. It expression and thereby beta4-mediated tumour is constantly degraded via the ubiquitin-proteasome progression in a p53-dependent manner. Beta4 system (mediated by the E3 ubiquitin ligases overexpression correlates in vivo with a SIAH1/SIAH2, WSB1 and MDM2). Various types cytoplasmic relocalisation of HIPK2, at least in of DNA damage (e.g. UV, IR or breast cancer: HIPK2 showed a cytoplasmic pattern chemotherapeutics) lead to stabilisation of the in 62.5% of the beta4-positive tumours (Bon et al., kinase and an HIPK2-mediated induction of 2009). apoptosis or presumably also senescence. HIPK2 can promote the apoptotic program via p53- Juvenile pilocytic astrocytomas dependent and -independent pathways through (JPA) phosphorylation of p53 at Ser46 or phosphorylation Note of the anti-apoptotic co-repressor CtBP at Ser422 Benign childhood brain tumors. (both actions leading to the transcription of pro- apoptotic target genes). Disease HIPK2 plays a role in the transcriptional regulation A frequent amplification of HIPK2 along with at low oxigen concentrations (hypoxia). BRAF rearrangements in JPA (35 out of 53 cases) Interestingly, HIPK2 also seems to have pro- through 7q34 duplication was reported. This survival functions, at least in neurons. duplication was more specific for JPA that Homology originated from the cerebellum or the optic chiasm. It was absent in other brain tumours. If (and how) HIPK2 is conserved from flies to man. HIPK2 contributes to JPA development is currently Mutations unclear (Jacob et al., 2009). Cervical cancer Somatic Note HIPK2 is rarely mutated (2 out of 130 cases) in Surprisingly, a significant overexpression of HIPK2 acute myeloid leukemia (AML) and myelodyplastic in cervical cancer was reported. But if (and how) syndrome (MDS) patients. Two missense mutations HIPK2 contributes to the development of cervical (R868W and N958I) within the speckle-retention carcinomas remains unclear. No correlation signal (SRS) were reported. These mutations led to between HIPK2 expression and grade or prognosis a changed nuclear localisation of HIPK2 and a of the disease could be demonstrated so far (Al- decreased transactivation potential in AML1- and Beiti et al., 2008). p53-dependent transcription. The mutants showed AML(RUNX1)-associated leukemias dominant-negative effects (Li et al., 2007). Oncogenesis Implicated in HIPK2 is inactivated on protein level by relocalisation through a PEBP2-beta-SMMHC and breast cancer fusion protein. Targeting of HIPK2 to cytoplasmic filaments and thereby prevention of Oncogenesis AML1(RUNX1) activation was reported. HIPK2 is frequently inactivated by transcriptional Specifically, phosphorylation of RUNX1 and its downregulation in thyroid carcinomas (8 out of 14 cofactor p300 seems to be inhibited by HIPK2 cases) and breast carcinomas (8 out of 20 cases) relocalisation (Wee et al., 2008). (Pierantoni et al., 2002). Breast cancer References Oncogenesis Kim YH, Choi CY, Lee SJ, Conti MA, Kim Y. HIPK2 is inactivated on protein level by Homeodomain-interacting protein kinases, a novel family cytoplasmic relocalisation through HMGA1. of co-repressors for homeodomain transcription factors. J Biol Chem. 1998 Oct 2;273(40):25875-9

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1145

HIPK2 (homeodomain interacting protein kinase 2) Sombroek D, Hofmann TG

Choi CY, Kim YH, Kwon HJ, Kim Y. The homeodomain Gresko E, Möller A, Roscic A, Schmitz ML. Covalent protein NK-3 recruits Groucho and a histone deacetylase modification of human homeodomain interacting protein complex to repress transcription. J Biol Chem. 1999 Nov kinase 2 by SUMO-1 at lysine 25 affects its stability. 19;274(47):33194-7 Biochem Biophys Res Commun. 2005 Apr 22;329(4):1293- 9 Kim YH, Choi CY, Kim Y. Covalent modification of the homeodomain-interacting protein kinase 2 (HIPK2) by the Zhang D, Li K, Erickson-Miller CL, Weiss M, Wojchowski ubiquitin-like protein SUMO-1. Proc Natl Acad Sci U S A. DM. DYRK gene structure and erythroid-restricted features 1999 Oct 26;96(22):12350-5 of DYRK3 gene expression. Genomics. 2005 Jan;85(1):117-30 Hofmann TG, Mincheva A, Lichter P, Dröge W, Schmitz ML. Human homeodomain-interacting protein kinase-2 Aikawa Y, Nguyen LA, Isono K, Takakura N, Tagata Y, (HIPK2) is a member of the DYRK family of protein Schmitz ML, Koseki H, Kitabayashi I. Roles of HIPK1 and kinases and maps to chromosome 7q32-q34. Biochimie. HIPK2 in AML1- and p300-dependent transcription, 2000 Dec;82(12):1123-7 hematopoiesis and blood vessel formation. EMBO J. 2006 Sep 6;25(17):3955-65 Pierantoni GM, Fedele M, Pentimalli F, Benvenuto G, Pero R, Viglietto G, Santoro M, Chiariotti L, Fusco A. High Gresko E, Roscic A, Ritterhoff S, Vichalkovski A, del Sal mobility group I (Y) proteins bind HIPK2, a serine- G, Schmitz ML. Autoregulatory control of the p53 response threonine kinase protein which inhibits cell growth. by caspase-mediated processing of HIPK2. EMBO J. 2006 Oncogene. 2001 Sep 27;20(43):6132-41 May 3;25(9):1883-94 Wang Y, Hofmann TG, Runkel L, Haaf T, Schaller H, Isono K, Nemoto K, Li Y, Takada Y, Suzuki R, Katsuki M, Debatin K, Hug H. Isolation and characterization of cDNAs Nakagawara A, Koseki H. Overlapping roles for for the protein kinase HIPK2. Biochim Biophys Acta. 2001 homeodomain-interacting protein kinases hipk1 and hipk2 Mar 19;1518(1-2):168-72 in the mediation of cell growth in response to morphogenetic and genotoxic signals. Mol Cell Biol. 2006 D'Orazi G, Cecchinelli B, Bruno T, Manni I, Higashimoto Y, Apr;26(7):2758-71 Saito S, Gostissa M, Coen S, Marchetti A, Del Sal G, Piaggio G, Fanciulli M, Appella E, Soddu S. Kim EA, Noh YT, Ryu MJ, Kim HT, Lee SE, Kim CH, Lee Homeodomain-interacting protein kinase-2 phosphorylates C, Kim YH, Choi CY. Phosphorylation and transactivation p53 at Ser 46 and mediates apoptosis. Nat Cell Biol. 2002 of Pax6 by homeodomain-interacting protein kinase 2. J Jan;4(1):11-9 Biol Chem. 2006 Mar 17;281(11):7489-97 Hofmann TG, Möller A, Sirma H, Zentgraf H, Taya Y, Dauth I, Krüger J, Hofmann TG. Homeodomain-interacting Dröge W, Will H, Schmitz ML. Regulation of p53 activity by protein kinase 2 is the ionizing radiation-activated p53 its interaction with homeodomain-interacting protein serine 46 kinase and is regulated by ATM. Cancer Res. kinase-2. Nat Cell Biol. 2002 Jan;4(1):1-10 2007 Mar 1;67(5):2274-9 Pierantoni GM, Bulfone A, Pentimalli F, Fedele M, Iuliano Li XL, Arai Y, Harada H, Shima Y, Yoshida H, Rokudai S, R, Santoro M, Chiariotti L, Ballabio A, Fusco A. The Aikawa Y, Kimura A, Kitabayashi I. Mutations of the HIPK2 homeodomain-interacting protein kinase 2 gene is gene in acute myeloid leukemia and myelodysplastic expressed late in embryogenesis and preferentially in syndrome impair AML1- and p53-mediated transcription. retina, muscle, and neural tissues. Biochem Biophys Res Oncogene. 2007 Nov 8;26(51):7231-9 Commun. 2002 Jan 25;290(3):942-7 Pierantoni GM, Rinaldo C, Mottolese M, Di Benedetto A, Harada J, Kokura K, Kanei-Ishii C, Nomura T, Khan MM, Esposito F, Soddu S, Fusco A. High-mobility group A1 Kim Y, Ishii S. Requirement of the co-repressor inhibits p53 by cytoplasmic relocalization of its homeodomain-interacting protein kinase 2 for ski-mediated proapoptotic activator HIPK2. J Clin Invest. 2007 inhibition of bone morphogenetic protein-induced Mar;117(3):693-702 transcriptional activation. J Biol Chem. 2003 Oct 3;278(40):38998-9005 Rinaldo C, Prodosmo A, Mancini F, Iacovelli S, Sacchi A, Moretti F, Soddu S. MDM2-regulated degradation of Zhang Q, Yoshimatsu Y, Hildebrand J, Frisch SM, HIPK2 prevents p53Ser46 phosphorylation and DNA Goodman RH. Homeodomain interacting protein kinase 2 damage-induced apoptosis. Mol Cell. 2007 Mar promotes apoptosis by downregulating the transcriptional 9;25(5):739-50 corepressor CtBP. Cell. 2003 Oct 17;115(2):177-86 Wei G, Ku S, Ma GK, Saito S, Tang AA, Zhang J, Mao JH, Di Stefano V, Rinaldo C, Sacchi A, Soddu S, D'Orazi G. Appella E, Balmain A, Huang EJ. HIPK2 represses beta- Homeodomain-interacting protein kinase-2 activity and p53 catenin-mediated transcription, epidermal stem cell phosphorylation are critical events for cisplatin-mediated expansion, and skin tumorigenesis. Proc Natl Acad Sci U apoptosis. Exp Cell Res. 2004 Feb 15;293(2):311-20 S A. 2007 Aug 7;104(32):13040-5 Doxakis E, Huang EJ, Davies AM. Homeodomain- Zhang J, Pho V, Bonasera SJ, Holtzman J, Tang AT, interacting protein kinase-2 regulates apoptosis in Hellmuth J, Tang S, Janak PH, Tecott LH, Huang EJ. developing sensory and sympathetic neurons. Curr Biol. Essential function of HIPK2 in TGFbeta-dependent survival 2004 Oct 5;14(19):1761-5 of midbrain dopamine neurons. Nat Neurosci. 2007 Jan;10(1):77-86 Kanei-Ishii C, Ninomiya-Tsuji J, Tanikawa J, Nomura T, Ishitani T, Kishida S, Kokura K, Kurahashi T, Ichikawa- Al-Beiti MA, Lu X. Expression of HIPK2 in cervical cancer: Iwata E, Kim Y, Matsumoto K, Ishii S. Wnt-1 signal induces correlation with clinicopathology and prognosis. Aust N Z J phosphorylation and degradation of c-Myb protein via Obstet Gynaecol. 2008 Jun;48(3):329-36 TAK1, HIPK2, and NLK. Genes Dev. 2004 Apr 1;18(7):816-29 Choi DW, Seo YM, Kim EA, Sung KS, Ahn JW, Park SJ, Lee SR, Choi CY. Ubiquitination and degradation of Choi CY, Kim YH, Kim YO, Park SJ, Kim EA, homeodomain-interacting protein kinase 2 by WD40 Riemenschneider W, Gajewski K, Schulz RA, Kim Y. repeat/SOCS box protein WSB-1. J Biol Chem. 2008 Feb Phosphorylation by the DHIPK2 protein kinase modulates 22;283(8):4682-9 the corepressor activity of Groucho. J Biol Chem. 2005 Jun 3;280(22):21427-36 Wee HJ, Voon DC, Bae SC, Ito Y. PEBP2-beta/CBF-beta-

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1146

HIPK2 (homeodomain interacting protein kinase 2) Sombroek D, Hofmann TG

dependent phosphorylation of RUNX1 and p300 by HIPK2: Jacob K, Albrecht S, Sollier C, Faury D, Sader E, Montpetit implications for leukemogenesis. Blood. 2008 Nov A, Serre D, Hauser P, Garami M, Bognar L, Hanzely Z, 1;112(9):3777-87 Montes JL, Atkinson J, Farmer JP, Bouffet E, Hawkins C, Tabori U, Jabado N. Duplication of 7q34 is specific to Winter M, Sombroek D, Dauth I, Moehlenbrink J, juvenile pilocytic astrocytomas and a hallmark of cerebellar Scheuermann K, Crone J, Hofmann TG. Control of HIPK2 and optic pathway tumours. Br J Cancer. 2009 Aug stability by ubiquitin ligase Siah-1 and checkpoint kinases 18;101(4):722-33 ATM and ATR. Nat Cell Biol. 2008 Jul;10(7):812-24 Nardinocchi L, Puca R, Guidolin D, Belloni AS, Bossi G, Bon G, Di Carlo SE, Folgiero V, Avetrani P, Lazzari C, Michiels C, Sacchi A, Onisto M, D'Orazi G. Transcriptional D'Orazi G, Brizzi MF, Sacchi A, Soddu S, Blandino G, regulation of hypoxia-inducible factor 1alpha by HIPK2 Mottolese M, Falcioni R. Negative regulation of beta4 suggests a novel mechanism to restrain tumor growth. integrin transcription by homeodomain-interacting protein Biochim Biophys Acta. 2009 Feb;1793(2):368-77 kinase 2 and p53 impairs tumor progression. Cancer Res. 2009 Jul 15;69(14):5978-86 This article should be referenced as such: Calzado MA, de la Vega L, Möller A, Bowtell DD, Schmitz Sombroek D, Hofmann TG. HIPK2 (homeodomain ML. An inducible autoregulatory loop between HIPK2 and interacting protein kinase 2). Atlas Genet Cytogenet Oncol Siah2 at the apex of the hypoxic response. Nat Cell Biol. Haematol. 2010; 14(12):1144-1147. 2009 Jan;11(1):85-91

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1147

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

RAD9A (RAD9 homolog A (S. pombe)) Vivian Chan Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, Hong Kong, China (VC)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/RAD9AID42031ch11q13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI RAD9AID42031ch11q13.txt

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

hRad9 forms ring-shape heterotrimeric complex Identity with hRad1 and hHus1 proteins (9-1-1 complex). Other names: RAD9, hRAD9 All 3 proteins have sequence homology with HGNC (Hugo): RAD9A proliferating cell nuclear antigen (PCNA). The 9-1- 1 complex is recruited onto DNA-lesion by RAD17 Location: 11q13.2 and ATR - triggering checkpoint signaling pathway Note: Accession No. NM_004584. and acts to repair DNA damage (Volkmer and Karnitz, 1999; Rauen et al., 2000; Zou et al., 2002; DNA/RNA Medhurst et al., 2008). Phosphorylation of hRad9 by protein kinase C delta (PKCD) is necessary for Description the formation of the 9-1-1 complex (Yoshida et al., 6461 bp, 11 exons. 2003). Transcription NH2 terminus of hRad9 contains BH3-like domain which binds antiapoptotic proteins BCL2 and Bcl- The transcript length is 1176 bp, full open reading x2, thereby promoting apoptosis (Komatsu et al., frame cDNA clone, encodes a 391 amino acid, 2000). This interaction of hRad9 to Bcl2 is 42520 Da protein (Lieberman et al., 1996). regulated also by PKCdelta (Yoshida et al., 2003). RAD9, like P53 can regulate P21 at the Protein transcriptional level. Overexpression of hRad was shown to cause an Function increase in P21 RNA and the encoded protein level The gene product is highly similar to Rad9 protein in P53-null H1299 cells (Yin et al., 2004). This from S pombe. A cell cycle checkpoint protein with suggests that hRAD9 and P53 coregulate P21 to multiple functions for preserving genomic integrity direct cell cycle progression. hRAD9 may also (Ishikawa et al., 2006), such as the regulation of modulate transcription of other down-stream target DNA damage response, cell cycle checkpoint, DNA genes. repair, apoptosis, transcriptional regulation, C-terminal region of hRad9 protein acts to transport exonuclease activity, ribonucleotide synthesis and 9-1-1 complex into the nucleus (Hirai and Wang, embryogenesis. 2002; Sohn and Cho, 2009).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1148

RAD9A (RAD9 homolog A (S. pombe)) Chan V

(Adapted from Ishikawa K et al., Current Genomics.2006:7:477-80). hRad9 and ATM rapidly colocalize to regions structurally related to hRad9 protein (55% similar containing DNA double-stranded breaks after and 35% identical). HRad9B gene is expressed DNA-damage (Greer et al., 2003; Medhurst et al., predominantly in the testis and found in decreased 2008) and Atm can phosphorylate Rad9 directly at amount in testicular tumours, particularly Ser-272 during ionizing radiation (IR)-induced seminomas (Hopkins et al., 2003). G1/S checkpoint activation (Chen et al., 2001). Prostate cancer The 9-1-1 complex may attract DNA polymerase beta to sites of DNA damage, thus connecting Oncogenesis checkpoint and DNA repair (Toueille et al., 2004). Carboxy terminus of hRad9 contains a FXXLF Thr-292 of hRad9 is subject to CDC2-dependent motif which interrupts the androgen-induced phosphorylation in mitosis. Four other hRad9 interaction between the C and N terminus of phosphorylation sites (Ser-277, Ser-328, Ser-336 androgen receptor (AR), acting as a co-regulator to and Thr-355) are regulated in part by Cdc2 (St suppress androgen-AR transactivation in prostrate Onge et al., 2001; St Onge et al., 2003; Ishikawa et cancer cells (Wang et al., 2004). This denotes a al., 2006). possible tumour suppressor function of hRad9. Phosphorylation sites of the C-terminal region of Recent study has confirmed that high levels of hRad9 are essential for CHK1 activation following Rad9 expression is found in prostate cancer cells hydroxyurea, ionizing radiation and ultraviolet and the high protein levels in prostate treatment (Roos-Mattjus et al., 2003). adenocarcinomas were generally associated with Crystal structure of the human Rad9-Hus1-Rad1 more advanced disease (Zhu et al., 2008). Similar complex reveals a single repair enzyme binding site to previous findings in breast cancer (Cheng et al., (Doré et al., 2009) and suggests that the C-terminal 2005), the increased expression of Rad9 in prostate end of Rad9 protein is involved in the regulation of cancer cells was in part due to aberrant methylation the complex in DNA binding (Sohn and Cho, or gene amplification (Zhu et al., 2008). The study 2009). failed to show that the role of Rad9 in prostate hRad9 possesses 3'-5' exonuclease activity which tumorigenesis was androgen dependent, since both may contribute to its role in sensing and repairing androgen dependent CWR22 and LNCaP cell lines DNA damage (Bessho and Sancar, 2000). The exact as well as androgen independent DU145 and PC-3 mechanism of this exonucleolytic processing is still cell lines were found to contain high levels of Rad9 unclear. protein (Zhu et al., 2008). Implicated in Lung cancer Oncogenesis Various cancers Presence of hyperphosphorylated forms of hRad9 has been found in the nuclei of surgically resected Oncogenesis primary lung carcinoma cells (Maniwa et al., 2005). Checkpoint genes are known to be involved in the No mutation of the hRad9 gene was found in lung maintenance of genomic integrity and their aberrant cancer cells, but a nonsynonymous single expression can lead to cancer. Paralogue of human nucleotide polymorphism (SNP), His239Arg was HRad9 is called HRad9B. Gene product is found in 8 out of 50 lung adenocarcinoma patients,

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1149

RAD9A (RAD9 homolog A (S. pombe)) Chan V

suggesting a possible association of this SNP with DNA damage-responsive protein complex. J Biol Chem. the development of cancer (Maniwa et al., 2006). 1999 Jan 8;274(2):567-70 Bessho T, Sancar A. Human DNA damage checkpoint Breast cancer protein hRAD9 is a 3' to 5' exonuclease. J Biol Chem. Oncogenesis 2000 Mar 17;275(11):7451-4 Over-expression of hRad9 mRNA was found in Komatsu K, Miyashita T, Hang H, Hopkins KM, Zheng W, breast cancer, which was shown to be correlated Cuddeback S, Yamada M, Lieberman HB, Wang HG. with tumour size (p = 0.037) and local recurrence (p Human homologue of S. pombe Rad9 interacts with BCL- 2/BCL-xL and promotes apoptosis. Nat Cell Biol. 2000 = 0.033). Over-expression of Rad9 mRNA was Jan;2(1):1-6 partly due to increase in RAD9 gene amplification and aberrant DNA methylation at a putative Sp 1/3 Rauen M, Burtelow MA, Dufault VM, Karnitz LM. The human checkpoint protein hRad17 interacts with the binding site within the second intron of the RAD9 PCNA-like proteins hRad1, hHus1, and hRad9. J Biol gene. Promoter assays indicate that the Sp 1/3 site Chem. 2000 Sep 22;275(38):29767-71 in intron 2 may act as a silencer. Further Chen MJ, Lin YT, Lieberman HB, Chen G, Lee EY. ATM- experiments in silencing Rad9 expression by RNAi dependent phosphorylation of human Rad9 is required for inhibit the proliferation of MCF-7 cell line in vitro. ionizing radiation-induced checkpoint activation. J Biol These findings suggested that Rad9 is a new Chem. 2001 May 11;276(19):16580-6 oncogene candidate on Ch11q13 with a role in Lindsey-Boltz LA, Bermudez VP, Hurwitz J, Sancar A. breast cancer progression (Cheng et al., 2005). Purification and characterization of human DNA damage In contrast to previous findings in testicular checkpoint Rad complexes. Proc Natl Acad Sci U S A. 2001 Sep 25;98(20):11236-41 tumours, increased hRad9 protein was found in the nuclei of breast cancer cells. These were shown to St Onge RP, Besley BD, Park M, Casselman R, Davey S. DNA damage-dependent and -independent exist as hyperphosphorylated forms, with molecular phosphorylation of the hRad9 checkpoint protein. J Biol weights of 65 and 50 kDa. Since the theoretical Chem. 2001 Nov 9;276(45):41898-905 molecular weight of hRad9 is 45 kDa (Lindsey- Hirai I, Wang HG. A role of the C-terminal region of human Boltz et al., 2001), these larger forms most likely Rad9 (hRad9) in nuclear transport of the hRad9 represent hyperphosphorylated hRad9 and its checkpoint complex. J Biol Chem. 2002 Jul hRad9-hRad1-hHus1 complex (Chan et al., 2008; 12;277(28):25722-7 St Onge et al., 1999). Localization of Zou L, Cortez D, Elledge SJ. Regulation of ATR substrate hyperphosphorylated forms of hRad in the nucleus selection by Rad17-dependent loading of Rad9 complexes of cancer cells is in keeping with its function in onto chromatin. Genes Dev. 2002 Jan 15;16(2):198-208 ameliorating DNA instability, whereby it Greer DA, Besley BD, Kennedy KB, Davey S. hRad9 inadvertently assists tumour growth. rapidly binds DNA containing double-strand breaks and is required for damage-dependent topoisomerase II beta Colorectal cancer binding protein 1 focus formation. Cancer Res. 2003 Aug Oncogenesis 15;63(16):4829-35 Rad9 interacts physically within the DNA Hopkins KM, Wang X, Berlin A, Hang H, Thaker HM, mismatch repair (MMR) protein MLH1. Disruption Lieberman HB. Expression of mammalian paralogues of HRAD9 and Mrad9 checkpoint control genes in normal and of the interaction by a single point mutation in cancerous testicular tissue. Cancer Res. 2003 Sep Rad9 leads to significantly reduced mismatch repair 1;63(17):5291-8 activity (He et al., 2008). The Rad9-MHL1 Roos-Mattjus P, Hopkins KM, Oestreich AJ, Vroman BT, interaction might be a hotspot for mutation in Johnson KL, Naylor S, Lieberman HB, Karnitz LM. tumour cells. The hMLH1 mutations lead to Phosphorylation of human Rad9 is required for genotoxin- hereditary non-polyposis colorectal cancer activated checkpoint signaling. J Biol Chem. 2003 Jul (HNPCC) (Avdievich et al., 2008; Peltomäki et al., 4;278(27):24428-37 2004) and various types of tumours (Avdievich et St Onge RP, Besley BD, Pelley JL, Davey S. A role for the al., 2008; Hu et al., 2008). However, hRad9's phosphorylation of hRad9 in checkpoint signaling. J Biol Chem. 2003 Jul 18;278(29):26620-8 function in MMR is not in the 9-1-1-complex form (He et al., 2008). Yoshida K, Wang HG, Miki Y, Kufe D. Protein kinase Cdelta is responsible for constitutive and DNA damage- induced phosphorylation of Rad9. EMBO J. 2003 Mar References 17;22(6):1431-41 Lieberman HB, Hopkins KM, Nass M, Demetrick D, Davey Peltomäki P, Vasen H. Mutations associated with HNPCC S. A human homolog of the Schizosaccharomyces pombe predisposition -- Update of ICG-HNPCC/INSiGHT mutation rad9+ checkpoint control gene. Proc Natl Acad Sci U S A. database. Dis Markers. 2004;20(4-5):269-76 1996 Nov 26;93(24):13890-5 Toueille M, El-Andaloussi N, Frouin I, Freire R, Funk D, St Onge RP, Udell CM, Casselman R, Davey S. The Shevelev I, Friedrich-Heineken E, Villani G, Hottiger MO, human G2 checkpoint control protein hRAD9 is a nuclear Hübscher U. The human Rad9/Rad1/Hus1 damage sensor phosphoprotein that forms complexes with hRAD1 and clamp interacts with DNA polymerase beta and increases hHUS1. Mol Biol Cell. 1999 Jun;10(6):1985-95 its DNA substrate utilisation efficiency: implications for DNA repair. Nucleic Acids Res. 2004;32(11):3316-24 Volkmer E, Karnitz LM. Human homologs of Schizosaccharomyces pombe rad1, hus1, and rad9 form a Wang L, Hsu CL, Ni J, Wang PH, Yeh S, Keng P, Chang

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1150

RAD9A (RAD9 homolog A (S. pombe)) Chan V

C. Human checkpoint protein hRad9 functions as a Chan V, Khoo US, Wong MS, Lau K, Suen D, Li G, Kwong negative coregulator to repress androgen receptor A, Chan TK. Localization of hRad9 in breast cancer. BMC transactivation in prostate cancer cells. Mol Cell Biol. 2004 Cancer. 2008 Jul 11;8:196 Mar;24(5):2202-13 He W, Zhao Y, Zhang C, An L, Hu Z, Liu Y, Han L, Bi L, Yin Y, Zhu A, Jin YJ, Liu YX, Zhang X, Hopkins KM, Xie Z, Xue P, Yang F, Hang H. Rad9 plays an important Lieberman HB. Human RAD9 checkpoint role in DNA mismatch repair through physical interaction control/proapoptotic protein can activate transcription of with MLH1. Nucleic Acids Res. 2008 Nov;36(20):6406-17 p21. Proc Natl Acad Sci U S A. 2004 Jun 15;101(24):8864- 9 Hu Z, Liu Y, Zhang C, Zhao Y, He W, Han L, Yang L, Hopkins KM, Yang X, Lieberman HB, Hang H. Targeted Cheng CK, Chow LW, Loo WT, Chan TK, Chan V. The cell deletion of Rad9 in mouse skin keratinocytes enhances cycle checkpoint gene Rad9 is a novel oncogene activated genotoxin-induced tumor development. Cancer Res. 2008 by 11q13 amplification and DNA methylation in breast Jul 15;68(14):5552-61 cancer. Cancer Res. 2005 Oct 1;65(19):8646-54 Medhurst AL, Warmerdam DO, Akerman I, Verwayen EH, Maniwa Y, Yoshimura M, Bermudez VP, Yuki T, Okada K, Kanaar R, Smits VA, Lakin ND. ATR and Rad17 Kanomata N, Ohbayashi C, Hayashi Y, Hurwitz J, Okita Y. collaborate in modulating Rad9 localisation at sites of DNA Accumulation of hRad9 protein in the nuclei of nonsmall damage. J Cell Sci. 2008 Dec 1;121(Pt 23):3933-40 cell lung carcinoma cells. Cancer. 2005 Jan 1;103(1):126- 32 Zhu A, Zhang CX, Lieberman HB. Rad9 has a functional role in human prostate carcinogenesis. Cancer Res. 2008 Ishikawa K, Ishii H, Saito T, Ichimura K. Multiple functions Mar 1;68(5):1267-74 of rad9 for preserving genomic integrity. Curr Genomics. 2006;7(8):477-80 Doré AS, Kilkenny ML, Rzechorzek NJ, Pearl LH. Crystal structure of the rad9-rad1-hus1 DNA damage checkpoint Maniwa Y, Yoshimura M, Bermudez VP, Okada K, complex--implications for clamp loading and regulation. Kanomata N, Ohbayashi C, Nishimura Y, Hayashi Y, Mol Cell. 2009 Jun 26;34(6):735-45 Hurwitz J, Okita Y. His239Arg SNP of HRAD9 is associated with lung adenocarcinoma. Cancer. 2006 Mar Sohn SY, Cho Y. Crystal structure of the human rad9- 1;106(5):1117-22 hus1-rad1 clamp. J Mol Biol. 2009 Jul 17;390(3):490-502 Avdievich E, Reiss C, Scherer SJ, Zhang Y, Maier SM, Jin This article should be referenced as such: B, Hou H Jr, Rosenwald A, Riedmiller H, Kucherlapati R, Cohen PE, Edelmann W, Kneitz B. Distinct effects of the Chan V. RAD9A (RAD9 homolog A (S. pombe)). Atlas recurrent Mlh1G67R mutation on MMR functions, cancer, Genet Cytogenet Oncol Haematol. 2010; 14(12):1148- and meiosis. Proc Natl Acad Sci U S A. 2008 Mar 1151. 18;105(11):4247-52

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1151

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

SCAF1 (SR-related CTD-associated factor 1) Christos Kontos, Andreas Scorilas Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Athens, 157 01, Panepistimiopolis, Athens, Greece (CK, AS)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SCAF1ID46074ch19q13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SCAF1ID46074ch19q13.txt

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

The human SCAF1 gene was shown to be Identity expressed widely in many normal tissues, but its Other names: FLJ00034, SCAF, SFRS19, SRA1, mRNA levels vary a lot. The highest levels of SR-A1, SCAF1 transcripts were detected in the fetal brain HGNC (Hugo): SCAF1 and fetal liver and the lowest in salivary gland, skin, heart, uterus and ovary. Location: 19q13.33 In the mammary and prostate gland, SCAF1 mRNA Local order: Telomere to centromere. transcripts are constitutively present at relatively Note: The first name of this gene, discovered and high levels. cloned by Scorilas et al. was SR-A1. After the The mRNA levels of SCAF1 appear to increase in establishment of the name "SRA1" for steroid cancer cell lines treated with various steroid receptor RNA activator 1, the official name of SR- hormones, including , androgens and A1 gene has changed into SCAF1, to avoid glucocorticoids, and to a lesser extent with confusion. progestins (Scorilas et al., 2001). DNA/RNA Pseudogene Not identified so far. Description Spanning 16.5 kb of genomic DNA, the SCAF1 Protein gene consists of 11 exons and 10 intervening Description introns (Scorilas et al., 2001). The SCAF1 protein is composed of 1312 amino Transcription acids, with a calculated molecular mass of 139.1 The unique transcript of SCAF1 gene is 4313 bp. kDa and a theoretical isoelectric point of 9.31.

Schematic representation of the SCAF1 gene. Exons are shown as boxes and introns as connecting lines. Arrows show the positions of the start codon, stop codon, and polyadenylation signal. Roman numerals indicate intron phases. The intron phase refers to the location of the intron within the codon; I denotes that the intron occurs after the first nucleotide of the codon, II that the intron occurs after the second nucleotide, and 0 that the intron occurs between distinct codons. The numbers inside boxes indicate exon lengths and the vertical connecting lines show the intron lengths (in bp). Figure is not drawn to scale.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1152

SCAF1 (SR-related CTD-associated factor 1) Kontos C, Scorilas A

Schematic representation of the amino acid sequence of the SCAF1 protein. The Arg/Ser-rich domain is shown in bold and underlined, and the CTD-binding domain is double-underlined. Additionally, the SCAF1 protein contains two areas with negatively charged polyglutamic acid (E) stretches, shown as underlined with dashes, and an Arg/Asp-rich motif, which is normally underlined. Various putative post-translational modification sites have also been identified, including numerous potential sites for either O- or N-glycosylation, and several possible sites of phosphorylation by cAMP-dependent protein kinase (PKA), protein kinase C (PKC), and casein kinase 2.

The SCAF1 protein contains an Arg/Ser-rich Expression domain (SR) as well as a CTD-binding domain, Currently, there are no data concerning the in vivo present only in a subset of Arg/Ser-rich splicing expression of the human SCAF1 protein. factors. Through interactions with the pre-mRNA and the Localisation C-terminal domain (CTD) of the large subunit of The SCAF1 protein is predicted to be localized to RNA polymerase II, Arg/Ser-rich proteins have the nucleus. been shown to regulate alternative splicing. In addition, we identified two areas with negatively Function charged polyglutamic acid (E) stretches and an SCAF1 interacts with the CTD domain of the RNA Arg/Asp-rich motif in the SCAF1 protein. This polymerase II polypeptide A (POLR2A) and may motif is also present in a number of other RNA- be involved in pre-mRNA splicing. binding proteins such as the U1-70 K, the RD Homology RNA-binding protein and the 68 kDa human pre- Human SCAF1 shares 85% amino acid identity and mRNA cleavage factor Im. Examination of the hydrophobicity profile of the 91% similarity with the mouse and rat Scaf1 SCAF1 protein did not reveal regions with long protein. Moreover, it shows 25% identity and 48% stretches of hydrophobic residues. similarity with the human PHRF1 protein ("PHD SCAF1 is predicted to be a nuclear protein with no and RING finger domain-containing protein 1", transmembrane region. also known as "CTD-binding SR-like protein rA9"), and to a lesser extent with other Arg/Ser-rich Various putative post-translational modification splicing factors. sites have been identified, including numerous potential sites for either O- or N-glycosylation, and Mutations several possible sites of phosphorylation by cAMP- dependent protein kinase (PKA), protein kinase C No germinal or somatic mutations associated with (PKC), and casein kinase 2 (Scorilas et al., 2001). cancer have been identified so far.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1153

SCAF1 (SR-related CTD-associated factor 1) Kontos C, Scorilas A

induced by each drug may be due to distinct Implicated in apoptotic pathways and therefore to distinct cellular Breast and ovarian cancer needs for the splice variants of specific genes. Prognosis Cytogenetics Expression analysis of the SCAF1 gene has showed No cytogenetic abnormalities have been identified that SCAF1 mRNA expression may be considered so far. as a new unfavorable prognostic marker for breast Hybrid/Mutated gene and ovarian cancer. Expression of the SCAF1 gene Not identified so far. in breast cancer tissues is influenced by the tumor size and the existence of lymph node metastases. References Furthermore, high SCAF1 expression is a Scorilas A, Kyriakopoulou L, Katsaros D, Diamandis EP. significant independent prognostic marker of Cloning of a gene (SR-A1), encoding for a new member of disease-free survival (DFS), and low mRNA the human Ser/Arg-rich family of pre-mRNA splicing expression of the gene is associated with long DFS factors: overexpression in aggressive ovarian cancer. Br J and overall survival (OS). Cancer. 2001 Jul 20;85(2):190-8 Regarding SCAF1 gene expression in ovarian Mathioudaki K, Leotsakou T, Papadokostopoulou A, cancer, it is positively related to the histological Paraskevas E, Ardavanis A, Talieri M, Scorilas A. SR-A1, grade and stage of the disease, the size of the tumor, a member of the human pre-mRNA splicing factor family, and its expression in colon cancer progression. Biol Chem. and the debulking success. Additionally, high 2004 Sep;385(9):785-90 SCAF1 expression is a significant independent Katsarou ME, Papakyriakou A, Katsaros N, Scorilas A. prognostic marker of OS, and low mRNA Expression of the C-terminal domain of novel human SR- expression of the gene is related to long DFS and A1 protein: interaction with the CTD domain of RNA OS. polymerase II. Biochem Biophys Res Commun. 2005 Aug 19;334(1):61-8 Colon cancer Leoutsakou T, Talieri M, Scorilas A. Expression analysis Prognosis and prognostic significance of the SRA1 gene, in ovarian SCAF1 mRNA expression seems also to be cancer. Biochem Biophys Res Commun. 2006 Jun associated with colon cancer progression, since its 2;344(2):667-74 expression is higher at the initial stages of Leoutsakou T, Talieri M, Scorilas A. Prognostic tumorigenesis and is reduced as cancer progresses. significance of the expression of SR-A1, encoding a novel SR-related CTD-associated factor, in breast cancer. Biol Leukemia Chem. 2006 Dec;387(12):1613-8 Prognosis Katsarou ME, Thomadaki H, Katsaros N, Scorilas A. Effect Alterations of SCAF1 mRNA expression have been of bleomycin and cisplatin on the expression profile of SRA1, a novel member of pre-mRNA splicing factors, in noticed in the human acute promyelocytic leukemia HL-60 human promyelocytic leukemia cells. Biol Chem. cell line HL-60, after treatment with cisplatin and 2007 Aug;388(8):773-8 bleomycin. mRNA levels of SCAF1 are modulated in both cases as a response to apoptosis induction This article should be referenced as such: by each drug, with up-regulation in bleomycin- Kontos C, Scorilas A. SCAF1 (SR-related CTD-associated induced apoptosis and down-regulation in cisplatin- factor 1). Atlas Genet Cytogenet Oncol Haematol. 2010; induced apoptosis in HL-60 cells. This differential 14(12):1152-1154. response of SCAF1 mRNA levels to apoptosis

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1154

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

SIRT1 (sirtuin (silent mating type information regulation 2 homolog) 1 (S. cerevisiae)) Ruo-Chia Tseng, Yi-Ching Wang Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC (RCT, YCW)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SIRT1ID44006ch10q21.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SIRT1ID44006ch10q21.txt

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

with a predictive molecular weight of 81.7 kDa and Identity an isoelectric point of 4.55 (Alcaín and Villalba, Other names: EC 3.5.1, hSIR2, hSIRT1, 2009). SIR2alpha, SIR2L1 Transcription HGNC (Hugo): SIRT1 SIRT1 transcription is under the control of at least Location: 10q21.3 two negative feedback loops that keep its induction tightly regulated under conditions of oxidative DNA/RNA stress. SIRT1 promoter can be activated by E2F1 and HIC1 during cellular stress. Description E2F1 directly binds to the SIRT1 promoter at a The SIRT1 gene spans about 34 kb including nine consensus site located at bp position -65 and exons. The SIRT1 promoter contains a CCAAT box appears to regulate the basal expression level of and a number of NFkappaB and GATA SIRT1. transcription factor binding sites in addition to a Such high levels of SIRT1 lead to a negative small 350-bp CpG island in the 5' flanking genomic feedback loop where E2F1 activity is inhibited by region. The gene encodes a 747 amino acids protein SIRT1-mediated deacetylation.

SIRT1 gene expression is modulated at both transcriptional and posttranscriptional levels.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1155

SIRT1 (sirtuin (silent mating type information regulation 2 Tseng RC, Wang YC homolog) 1 (S. cerevisiae))

By contrast, the tumor suppressor HIC1 and SIRT1 SIRT1 was the active regulator of SIRT1 (AROS). form a transcriptional repression complex that The AROS protein is known to significantly directly binds SIRT1 promoter via its N-terminal enhance the activity of SIRT1 on acetylated p53 POZ domain and represses SIRT1 transcription both in vitro and in cell lines thereby promoting the thereby inhibiting SIRT1-mediated p53 inhibitory effect of SIRT1 on p53-mediated deacetylation and inactivation. transcriptional activity of pro-apoptotic genes (e.g. Two HIC1 binding sites have been assigned to base Bax and p21Waf-1) under conditions of DNA- pair positions -1116 and -1039 within the SIRT1 damage. A negative regulator of SIRT1, DBC-1 promoter. In addition, two functional p53 binding (deleted in breast cancer-1), has recently been sites (-178 bp and -168 bp), which normally repress identified. DBC1 binds directly to the catalytic SIRT1 expression, have been identified. domain of SIRT1, preventing substrate binding to SIRT1 expression is also regulated at the SIRT1 and inhibiting SIRT1 activity. Reduction of posttranscriptional level by HuR. It has been DBC1 inhibits p53-mediated apoptosis after demonstrated that HuR, a ubiquitously expressed induction of double-stranded DNA breaks owing to RNA binding protein, associates with the 3' UTR of SIRT1-mediated p53 deacetylation. Both factors the SIRT1 mRNA under physiological conditions represent the first endogenous, direct regulators of and helps to stabilize the transcript. This interaction SIRT1 function. results in increased SIRT1 mRNA stability and thus Localisation in elevated protein levels. Conversely, the HuR- SIRT1 mRNA complex is being disrupted upon SIRT1 is predominately in the nucleus (although oxidative stress, which finally leads to decreased SIRT1 does have some important cytoplasmic mRNA stability and therefore decreased SIRT functions as well). In addition to possessing two protein levels. NLSs, SIRT1 contains two nuclear export signals. Thus, the exposure of nuclear localization signals Pseudogene versus nuclear export signals may dictate the None identified. cytosolic versus nuclear localization of SIRT1. Protein Function SIRT1 has been reported to play a key role in a Description variety of physiological processes such as Human SIRT1 encodes 747 amino acids protein metabolism, neurogenesis and cell survival due to with a nuclear localization signal (NLS) at the N- its ability to deacetylate both histone and numerous terminus (aa 41-46) and a sirtuin homology domain nonhistone substrates. at the center (aa 261-447); this domain is a (1) Lysines 9 and 14 in the amino-terminal tail of conserved catalytic domain for deacetylation. histone H3 and lysine 16 of histone H4 are deacetylated by yeast Sir2 and mammalian SIRT1 Expression (Sir2alpha). Expression appears to be ubiquitous in adult tissues (2) Metabolic homeostasis is controlled by SIRT1- (although at different levels). Two proteins have mediated deacetylation and thus activation of the been identified to regulate the SIRT1 activity both peroxisome proliferation activating receptor positively and negatively through complex (PPAR)-gamma co-activator-1a (PGC-1a), which formation in the context of the cellular stress stimulates mitochondrial activity and subsequently response. The first identified direct regulator of increases glucose metabolism, which in turn improves sensitivity.

SIRT1 deacetylase activity is modulated through protein-protein interaction and sumoylation at its three protein domains (Liu T et al., 2009).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1156

SIRT1 (sirtuin (silent mating type information regulation 2 Tseng RC, Wang YC homolog) 1 (S. cerevisiae))

SIRT1 represses PPAR-gamma, a key regulator of carcinoma and lung adenocarcinoma. The lung adipogenesis, by docking with its cofactors NCoR squamous cell carcinoma patients with low p53 (nuclear receptor co-repressor) and SMRT acetylation and SIRT1 expression mostly showed (silencing mediator of retinoid and thyroid hormone low HIC1 expression, confirming deregulation receptors). The upregulation of SIRT1 triggers HIC1-SIRT1-p53 circular loop in clinical model. lipolysis and loss of fat. Expression of DBC1, which blocks the interaction (3) The activation of SIRT1 appears to be between SIRT1 deacetylase and p53, led to neuroprotective in animal models for Alzheimer's acetylated p53 in lung adenocarcinoma patients. disease and amyotrophic lateral sclerosis as well as Prognosis optic neuritis mainly due to decreased deacetylation Lung cancer patients with altered HIC1-SIRT1-p53 of the tumor suppressor p53 and PGC-1a. circular regulation showed poor prognosis. (4) SIRT1 represses p53-dependent apoptosis in response to DNA damage and oxidative stress and Breast cancer promotes cell survival under cellular stress induced Note by etoposide treatment or irradiation. The breast cancer associated protein, BCA3, when (5) SIRT1 activates FOXO1 and FOXO4, which neddylated (modified by NEDD8) interacts with promote cell-cycle arrest by inducing p27kip1; SIRT1 and suppresses NF-kB-dependent SIRT1 also induces cellular resistance to oxidative transcription, also sensitizes human breast cancer stress by increasing the levels of manganese cells (such as MCF7) to TNF-a-induced apoptosis. superoxide dismutase and GADD45 (growth arrest In addition, it has been shown recently that SIRT7 and DNA damage-inducible protein 45). levels of expression increase significantly in breast (6) SIRT1 inhibits the transcriptional activity of cancer, and that SIRT7 and SIRT3 both are highly NF-kappaB by deacetylating NF-kappaB's subunit, transcribed in lymph-node positive breast biopsies, RelA/p65, at lysine 310. Thus, although SIRT1 is a stage in which the tumour size is at least 2 mm capable of protecting cells from p53-induced and the cancer has already spread to the lymph apoptosis, it may augment apoptosis by repressing nodes. NF-kappaB. SIRT1 is reported to bind CTIP2 (BCL11B B-cell CLL/lymphoma 11B) and Brain tumor accelerate the transcriptional repression by this Note molecule. CTIP2 represses the transcription of its SIRT2 resides in a genomic region frequently target genes and is implicated in hematopoietic cell deleted in human gliomas and ectopic expression of development. SIRT2 in glioma-derived cell lines markedly Homology reduces their capacity to form colonies in vitro. Exogenously expressed SIRT2 blocks chromosomal SIRT1 is the mammalian homologue closest to condensation and hyperploidy in glioma cell lines, yeast NAD+-dependent deacetylase Sir2 (silent accompanied by the presence of cyclin B/cdc2 information regulation 2). It was originally activity in response to mitotic stress. Thus, SIRT2 identified as a lifespan extending gene when over- may be a novel metaphase check-point protein that expressed in budding yeast, Caenorhabditis elegans, promotes genomic integrity and inhibits the and Drosophila melanogaster. The SIR2 gene is uncontrolled proliferation of transformed cells. broadly conserved in organisms ranging from bacteria to humans. The accession numbers for the Kidney diseases amino acid sequences used are as follows: yeast Note Sir2 (CAA25667), mouse Sir2alpha (AAF24983), SIRT1 attenuates TGF-beta (transforming growth human Sirt1 (AAD40849). All of the sirtuin factor-beta) apoptotic signaling that is mediated by proteins contain the ~275 residue sirtuin homology the effector molecule Smad7. SIRT1-dependent domain. In many instances a highly conserved deacetylation of Smad7 at Lys60 and Lys70 protein domain represents a conserved functional enhances its ubiquitin-dependent proteasomal binding site for a metabolite or biomolecule and degradation via Smurf1 (Smad ubiquitination such conserved binding site domains are often regulatory factor 1), thus protecting glomerular found within enzymatic catalytic domains. mesangial cells from TGF-beta-dependent apoptosis. Implicated in Cardiac hypertrophy Lung cancer Note Note Decreasing hypertrophy or apoptosis in cardiac Distinct status of p53 acetylation/deacetylation and myocytes can ameliorate the disease, and there is HIC1 alteration mechanism result from different reason to suspect that SIRT1 activation may be SIRT1-DBC1 (deleted in breast cancer 1) control useful in this regard. SIRT1 protects primary and epigenetic alteration in lung squamous cell cultured myocytes from programmed cell death induced by serum starvation or by the activation of

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1157

SIRT1 (sirtuin (silent mating type information regulation 2 Tseng RC, Wang YC homolog) 1 (S. cerevisiae))

PARP-1 [poly(ADP-ribose) polymerase-1] in a protein hSir2(SIRT1). J Biol Chem. 2004 Jul p53-dependent manner. SIRT1 also deacetylates 9;279(28):28873-9 Lys115 and Lys121 of the histone variant H2A.Z, a Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, factor known to promote cardiac hypertrophy. In Frye RA, Mayo MW. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. doing so, SIRT1 promotes the ubiquitination and EMBO J. 2004 Jun 16;23(12):2369-80 proteosome-dependent degradation of H2A.Z, which may help to protect against heart failure. Chen WY, Wang DH, Yen RC, Luo J, Gu W, Baylin SB. Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses. Cell. References 2005 Nov 4;123(3):437-48 Frye RA. Characterization of five human cDNAs with Hisahara S, Chiba S, Matsumoto H, Horio Y. homology to the yeast SIR2 gene: Sir2-like proteins Transcriptional regulation of neuronal genes and its effect (sirtuins) metabolize NAD and may have protein ADP- on neural functions: NAD-dependent histone deacetylase ribosyltransferase activity. Biochem Biophys Res SIRT1 (Sir2alpha). J Pharmacol Sci. 2005 Jul;98(3):200-4 Commun. 1999 Jun 24;260(1):273-9 Kobayashi Y, Furukawa-Hibi Y, Chen C, Horio Y, Isobe K, Kaeberlein M, McVey M, Guarente L. The SIR2/3/4 Ikeda K, Motoyama N. SIRT1 is critical regulator of FOXO- complex and SIR2 alone promote longevity in mediated transcription in response to oxidative stress. Int J Saccharomyces cerevisiae by two different mechanisms. Mol Med. 2005 Aug;16(2):237-43 Genes Dev. 1999 Oct 1;13(19):2570-80 Pillai JB, Isbatan A, Imai S, Gupta MP. Poly(ADP-ribose) Imai S, Armstrong CM, Kaeberlein M, Guarente L. polymerase-1-dependent cardiac myocyte cell death Transcriptional silencing and longevity protein Sir2 is an during heart failure is mediated by NAD+ depletion and NAD-dependent histone deacetylase. Nature. 2000 Feb reduced Sir2alpha deacetylase activity. J Biol Chem. 2005 17;403(6771):795-800 Dec 30;280(52):43121-30 Brennan CM, Steitz JA. HuR and mRNA stability. Cell Mol Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Life Sci. 2001 Feb;58(2):266-77 Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature. Gray SG, Ekström TJ. The human histone deacetylase 2005 Mar 3;434(7029):113-8 family. Exp Cell Res. 2001 Jan 15;262(2):75-83 Ashraf N, Zino S, Macintyre A, Kingsmore D, Payne AP, Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, George WD, Shiels PG. Altered sirtuin expression is Guarente L, Gu W. Negative control of p53 by Sir2alpha associated with node-positive breast cancer. Br J Cancer. promotes cell survival under stress. Cell. 2001 Oct 2006 Oct 23;95(8):1056-61 19;107(2):137-48 Chen IY, Lypowy J, Pain J, Sayed D, Grinberg S, Alcendor Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, RR, Sadoshima J, Abdellatif M. Histone H2A.z is essential Pandita TK, Guarente L, Weinberg RA. hSIR2(SIRT1) for cardiac myocyte hypertrophy but opposed by silent functions as an NAD-dependent p53 deacetylase. Cell. information regulator 2alpha. J Biol Chem. 2006 Jul 2001 Oct 19;107(2):149-59 14;281(28):19369-77 Hiratsuka M, Inoue T, Toda T, Kimura N, Shirayoshi Y, Gao F, Cheng J, Shi T, Yeh ET. Neddylation of a breast Kamitani H, Watanabe T, Ohama E, Tahimic CG, cancer-associated protein recruits a class III histone Kurimasa A, Oshimura M. Proteomics-based identification deacetylase that represses NFkappaB-dependent of differentially expressed genes in human gliomas: down- transcription. Nat Cell Biol. 2006 Oct;8(10):1171-7 regulation of SIRT2 gene. Biochem Biophys Res Commun. 2003 Sep 26;309(3):558-66 Sauve AA, Wolberger C, Schramm VL, Boeke JD. The biochemistry of sirtuins. Annu Rev Biochem. 2006;75:435- Senawong T, Peterson VJ, Avram D, Shepherd DM, Frye 65 RA, Minucci S, Leid M. Involvement of the histone deacetylase SIRT1 in chicken ovalbumin upstream Voelter-Mahlknecht S, Mahlknecht U. Cloning, promoter transcription factor (COUP-TF)-interacting chromosomal characterization and mapping of the NAD- protein 2-mediated transcriptional repression. J Biol Chem. dependent histone deacetylases gene sirtuin 1. Int J Mol 2003 Oct 31;278(44):43041-50 Med. 2006 Jan;17(1):59-67 Alcendor RR, Kirshenbaum LA, Imai S, Vatner SF, Wang C, Chen L, Hou X, Li Z, Kabra N, Ma Y, Nemoto S, Sadoshima J. Silent information regulator 2alpha, a Finkel T, Gu W, Cress WD, Chen J. Interactions between longevity factor and class III histone deacetylase, is an E2F1 and SirT1 regulate apoptotic response to DNA essential endogenous apoptosis inhibitor in cardiac damage. Nat Cell Biol. 2006 Sep;8(9):1025-31 myocytes. Circ Res. 2004 Nov 12;95(10):971-80 Abdelmohsen K, Pullmann R Jr, Lal A, Kim HH, Galban S, Daitoku H, Hatta M, Matsuzaki H, Aratani S, Ohshima T, Yang X, Blethrow JD, Walker M, Shubert J, Gillespie DA, Miyagishi M, Nakajima T, Fukamizu A. Silent information Furneaux H, Gorospe M. Phosphorylation of HuR by Chk2 regulator 2 potentiates Foxo1-mediated transcription regulates SIRT1 expression. Mol Cell. 2007 Feb through its deacetylase activity. Proc Natl Acad Sci U S A. 23;25(4):543-57 2004 Jul 6;101(27):10042-7 Inoue T, Hiratsuka M, Osaki M, Yamada H, Kishimoto I, Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong Yamaguchi S, Nakano S, Katoh M, Ito H, Oshimura M. T, Machado De Oliveira R, Leid M, McBurney MW, SIRT2, a tubulin deacetylase, acts to block the entry to Guarente L. Sirt1 promotes fat mobilization in white chromosome condensation in response to mitotic stress. adipocytes by repressing PPAR-gamma. Nature. 2004 Jun Oncogene. 2007 Feb 15;26(7):945-57 17;429(6993):771-6 Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi van der Horst A, Tertoolen LG, de Vries-Smits LM, Frye F, Rodgers JT, Delalle I, Baur JA, Sui G, Armour SM, RA, Medema RH, Burgering BM. FOXO4 is acetylated Puigserver P, Sinclair DA, Tsai LH. SIRT1 deacetylase upon peroxide stress and deacetylated by the longevity protects against neurodegeneration in models for

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1158

SIRT1 (sirtuin (silent mating type information regulation 2 Tseng RC, Wang YC homolog) 1 (S. cerevisiae))

Alzheimer's disease and amyotrophic lateral sclerosis. Nature. 2008 Jan 31;451(7178):587-90 EMBO J. 2007 Jul 11;26(13):3169-79 Zschoernig B, Mahlknecht U. SIRTUIN 1: regulating the Kim EJ, Kho JH, Kang MR, Um SJ. Active regulator of regulator. Biochem Biophys Res Commun. 2008 Nov SIRT1 cooperates with SIRT1 and facilitates suppression 14;376(2):251-5 of p53 activity. Mol Cell. 2007 Oct 26;28(2):277-90 Alcain FJ, Villalba JM.. Sirtuin inhibitors. Expert Opin Ther Kume S, Haneda M, Kanasaki K, Sugimoto T, Araki S, Pat. 2009 Mar;19(3):283-94. Isshiki K, Isono M, Uzu T, Guarente L, Kashiwagi A, Koya D. SIRT1 inhibits transforming growth factor beta-induced Brooks CL, Gu W. How does SIRT1 affect metabolism, apoptosis in glomerular mesangial cells via Smad7 senescence and cancer? Nat Rev Cancer. 2009 deacetylation. J Biol Chem. 2007 Jan 5;282(1):151-8 Feb;9(2):123-8 Michan S, Sinclair D. Sirtuins in mammals: insights into Fujita Y, Yamaguchi A, Hata K, Endo M, Yamaguchi N, their biological function. Biochem J. 2007 May 15;404(1):1- Yamashita T. Zyxin is a novel interacting partner for 13 SIRT1. BMC Cell Biol. 2009 Jan 27;10:6 Shindler KS, Ventura E, Rex TS, Elliott P, Rostami A. Liu T, Liu PY, Marshall GM. The critical role of the class III SIRT1 activation confers neuroprotection in experimental histone deacetylase SIRT1 in cancer. Cancer Res. 2009 optic neuritis. Invest Ophthalmol Vis Sci. 2007 Mar 1;69(5):1702-5 Aug;48(8):3602-9 Tseng RC, Lee CC, Hsu HS, Tzao C, Wang YC. Distinct Stankovic-Valentin N, Deltour S, Seeler J, Pinte S, HIC1-SIRT1-p53 loop deregulation in lung squamous Vergoten G, Guérardel C, Dejean A, Leprince D. An carcinoma and adenocarcinoma patients. Neoplasia. 2009 acetylation/deacetylation-SUMOylation switch through a Aug;11(8):763-70 phylogenetically conserved psiKXEP motif in the tumor Haigis MC, Sinclair DA. Mammalian sirtuins: biological suppressor HIC1 regulates transcriptional repression insights and disease relevance. Annu Rev Pathol. activity. Mol Cell Biol. 2007 Apr;27(7):2661-75 2010;5:253-95 Tanno M, Sakamoto J, Miura T, Shimamoto K, Horio Y. Nucleocytoplasmic shuttling of the NAD+-dependent This article should be referenced as such: histone deacetylase SIRT1. J Biol Chem. 2007 Mar Tseng RC, Wang YC. SIRT1 (sirtuin (silent mating type 2;282(9):6823-32 information regulation 2 homolog) 1 (S. cerevisiae)). Atlas Zhao W, Kruse JP, Tang Y, Jung SY, Qin J, Gu W. Genet Cytogenet Oncol Haematol. 2010; 14(12):1155- Negative regulation of the deacetylase SIRT1 by DBC1. 1159.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1159

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

SLC16A3 (solute carrier family 16, member 3 (monocarboxylic acid transporter 4)) Céline Pinheiro, Fátima Baltazar Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal (CP, FB)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SLC16A3ID44573ch17q25.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SLC16A3ID44573ch17q25.txt

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

cells, chondrocytes, testis, lung, placenta, heart and Identity some mammalian cell lines (Halestrap and Other names: MCT3, MCT4, MGC138472, Meredith, 2004; Meredith and Christian, 2008). MGC138474 Localisation HGNC (Hugo): SLC16A3 Plasma membrane. Location: 17q25.3 Function DNA/RNA Proton-linked monocarboxylate transporter. Catalyzes plasma membrane transport of Note monocarboxylates such as lactate, pyruvate, SLC16A3 was first cloned from human circulating branched-chain oxo acids derived from leucine, blood by Price et al. (1998). valine and isoleucine, and the ketone bodies Description acetoacetate, beta-hydroxybutyrate and acetate. 11077 bp lenght, 5 exons. Homology Transcription Belongs to the major facilitator superfamily (MFS). Monocarboxylate porter (TC 2.A.1.13) family. The 3 transcripts have been described for this gene (all SLC16A3 gene is conserved in chimpanzee, dog, with protein product): SLC16A3-201, (5 exons; cow, mouse, rat, chicken, zebrafish, and M. grisea. 2033 bps transcript length; 465 residues translation length); SLC16A3-202 (4 exons; 4222 bps Implicated in transcript length; 465 residues translation length); SLC16A3-203 (5 exons; 2054 bps transcript length; Colorectal carcinoma 465 residues translation length). Note SLC16A3/MCT4 protein is overexpressed in Protein colorectal cancer (Pinheiro et al., 2008a). Description Cervical cancer 465 residues; 49469 Da; 12 transmembrane Note domains; intracellular N- and C-terminals. SLC16A3/MCT4 protein is overexpressed in Expression cervical cancer (Pinheiro et al., 2008b). SLC16A3/MCT4 protein overexpression in cervical SLC16A3/MCT4 is expressed in tissues such as cancer correlated with positivity for high-risk HPV white skeletal muscle fibres, astrocytes, white blood (Pinheiro et al., 2008b).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1160

SLC16A3 (solute carrier family 16, member 3 Pinheiro C, Baltazar F (monocarboxylic acid transporter 4))

Protein diagram drawn following UniProtKB/Swiss-Prot database prediction, using TMRPres2D software.

Bladder cancer a patient with a mitochondrial myopathy (Baker et al., 2001). Note SLC16A3 gene expression was upregulated in some Chronic obstructive pulmonary bladder tumours and induced by hypoxia in bladder disease cancer cell lines, but not in cultures of normal Note urothelium (Ord et al., 2005). SLC16A3/MCT4 downregulation was described in Breast cancer the vastus lateralis muscle of patients with chronic Note obstructive pulmonary disease as compared with Induction was also seen in two breast cancer cell healthy controls (Green et al., 2008). lines. Expression of SLC16A3 gene is higher in breast cancer distant metastasis as compared to References primary tumours or regional metastasis. SLC16A3 Price NT, Jackson VN, Halestrap AP. Cloning and gene was then included in the 'VEGF profile' of sequencing of four new mammalian monocarboxylate breast cancer, associated with promotion of vessel transporter (MCT) homologues confirms the existence of a transporter family with an ancient past. Biochem J. 1998 formation, survival under anaerobic conditions and Jan 15;329 ( Pt 2):321-8 loss of dependence upon fibroblasts (Hu et al., 2009). Baker SK, Tarnopolsky MA, Bonen A. Expression of MCT1 and MCT4 in a patient with mitochondrial myopathy. Ovarian cancer Muscle Nerve. 2001 Mar;24(3):394-8 Note Halestrap AP, Meredith D. The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino SLC16A3 gene expression was described to be acid transporters and beyond. Pflugers Arch. 2004 downregulated in malignant ovarian tumours as Feb;447(5):619-28 compared to normal ovarian surface epithelial cells. Ord JJ, Streeter EH, Roberts IS, Cranston D, Harris AL. Additionally, the non-tumorigenic cell line TOV- Comparison of hypoxia transcriptome in vitro with in vivo 81D presented higher expression that tumorigenic gene expression in human bladder cancer. Br J Cancer. cell lines (Wojnarowicz et al., 2008). 2005 Aug 8;93(3):346-54 SLC16A3 gene, among other transporter genes, was Green HJ, Burnett ME, D'Arsigny CL, O'Donnell DE, differentially expressed in a chemotherapy resistant Ouyang J, Webb KA. Altered metabolic and transporter ovarian cancer cell line and tumour tissue as characteristics of vastus lateralis in chronic obstructive compared to a chemosensitive cell line and tumour pulmonary disease. J Appl Physiol. 2008 Sep;105(3):879- 86 tissue. It was suggested that these transporters might be involved in drug influx/efflux, modulating Meredith D, Christian HC. The SLC16 monocaboxylate chemotherapy response (Cheng et al., 2010). transporter family. Xenobiotica. 2008 Jul;38(7-8):1072-106 Pinheiro C, Longatto-Filho A, Ferreira L, Pereira SM, Mitochondrial myopathy Etlinger D, Moreira MA, Jubé LF, Queiroz GS, Schmitt F, Baltazar F. Increasing expression of monocarboxylate Note transporters 1 and 4 along progression to invasive cervical SLC16A3/MCT4 overexpression was described in carcinoma. Int J Gynecol Pathol. 2008 Oct;27(4):568-74

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1161

SLC16A3 (solute carrier family 16, member 3 Pinheiro C, Baltazar F (monocarboxylic acid transporter 4))

Pinheiro C, Longatto-Filho A, Scapulatempo C, Ferreira L, with distant metastases and poor outcomes. BMC Med. Martins S, Pellerin L, Rodrigues M, Alves VA, Schmitt F, 2009 Mar 16;7:9 Baltazar F. Increased expression of monocarboxylate transporters 1, 2, and 4 in colorectal carcinomas. Virchows Cheng L, Lu W, Kulkarni B, Pejovic T, Yan X, Chiang JH, Arch. 2008 Feb;452(2):139-46 Hood L, Odunsi K, Lin B. Analysis of chemotherapy response programs in ovarian cancers by the next- Wojnarowicz PM, Breznan A, Arcand SL, Filali-Mouhim A, generation sequencing technologies. Gynecol Oncol. 2010 Provencher DM, Mes-Masson AM, Tonin PN. Construction May;117(2):159-69 of a chromosome 17 transcriptome in serous ovarian cancer identifies differentially expressed genes. Int J This article should be referenced as such: Gynecol Cancer. 2008 Sep-Oct;18(5):963-75 Pinheiro C, Baltazar F. SLC16A3 (solute carrier family 16, Hu Z, Fan C, Livasy C, He X, Oh DS, Ewend MG, Carey member 3 (monocarboxylic acid transporter 4)). Atlas LA, Subramanian S, West R, Ikpatt F, Olopade OI, van de Genet Cytogenet Oncol Haematol. 2010; 14(12):1160- Rijn M, Perou CM. A compact VEGF signature associated 1162.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1162

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

SPAM1 (sperm adhesion molecule 1 (PH-20 hyaluronidase, zona pellucida binding)) Asli Sade, Sreeparna Banerjee Department of Biology, Middle East Technical University, Ankara 06531, Turkey (AS, SB)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SPAM1ID42361ch7q31.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SPAM1ID42361ch7q31.txt

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

genes. Three of them (HYAL1, HYAL2 and Identity HYAL3) are clustered on chromosome 3p21.3 and Other names: EC 3.2.1.35, HYA1, HYAL1, the other three (HYAL4, SPAM1 and HYALP1) HYAL3, HYAL5, Hyal-PH20, MGC26532, PH-20, are clustered on chromosome 7q31.3. Of the three PH20, SPAG15 genes on chromosome 7q31.3, HYALP1 is an HGNC (Hugo): SPAM1 expressed pseudogene. The extensive homology between the six hyaluronidase genes suggests an Location: 7q31.32 ancient gene duplication event before the Local order: According to NCBI Map Viewer, emergence of modern mammals. genes flanking SPAM1 in centromere to telomere direction on 7q31.3 are: Description - HYALP1 7q31.3 hyaluronoglucosaminidase According to Entrez Gene, SPAM1 gene maps to pseudogene 1 locus NC_000007 and spans a region of 46136 bp. - HYAL4 7q31.3 hyaluronoglucosaminidase 4 According to Spidey (mRNA to genomic sequence - SPAM1 7q31.3 sperm adhesion molecule 1 alignment tool), SPAM1 has 7 exons, the sizes - TMEM229A 7q31.32 transmembrane protein being 78, 112, 1160, 90, 441, 99 and 404. 229A - hCG_1651160 7q31.33 SSU72 RNA polymerase Transcription II CTD phosphatase homolog pseudogene The SPAM1 mRNA has two isoforms; transcript Note: SPAM1 is a glycosyl-phosphatidyl inositol variant 1 (NM_003117) a 2395 bp mRNA and (GPI)-anchored enzyme found in all mammalian transcript variant 2 (NM_153189) a 2009 bp spermatozoa. The protein has a hyaluronidase mRNA. The variant 2 uses an alternate in-frame activity that enables sperm to penetrate the splice site in the 3' coding region, compared to cumulus, a role in zona pellucida binding and also variant 1, resulting in a shorter C-terminus. participates in Ca2+ signaling associated acrosomal The promoter region of SPAM1 has been shown to exocytosis. contain a CRE (cAMP-responsive element) sequence which is a binding site for CREM DNA/RNA (cAMP-responsive element modulator) and thus Spam1 is under a cAMP-dependent transcriptional Note regulation. No TATA or CCAAT boxes were found The human genome contains six hyaluronidase like in the promoter region of SPAM1.

The diagram of SPAM1 transcript variant 1. The red boxes represent the exons (in scale) and exon numbers are given below the boxes.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1163

SPAM1 (sperm adhesion molecule 1 (PH-20 hyaluronidase, Sade A, Banerjee S zona pellucida binding))

The testis-specific promoters of the human and SPAM1, which is GPI anchored, is responsible for mouse SPAM1 genes are derived from a sequence local degradation of the cumulus ECM during that was originally part of an ERV pol gene. sperm penetration. Plasma membrane SPAM1 Pseudogene mediates HA-induced sperm signaling via the HA binding domain. SPAM1 is also secreted by the The human SPAM1 pseudogene HYALP1 is epithelial cells of the epididymis and has a role in located on chromosome 7q31.3. sperm maturation. In addition SPAM1 is implicated in fluid reabsorption and urine concentration in Protein kidney. Note Homology Two sperm adhesion isoforms exist; one is 511 aa - Pan troglodytes sperm adhesion molecule 1 long isoform 1 and the other 509 aa long isoform 2. (SPAM1) When the two isoforms are aligned the sequences - Canis lupus familiaris sperm adhesion molecule 1 are 100% identical and no functional difference has (SPAM1) been reported. - Bos taurus sperm adhesion molecule 1 (SPAM1) Description - Mus musculus hyaluronoglucosaminidase 5 (Hyal5) SPAM1 is a 68 kDa protein that belongs to glycosyl - Mus musculus sperm adhesion molecule 1 hydrolase 56 family. This family of enzymes has (SPAM1) hyaluronidase activity which hydrolyses the - Rattus norvegicus sperm adhesion molecule 1 glycosidic bond between two or more (HYALP_RAT) carbohydrates, or between a carbohydrate and a - Gallus gallus sperm adhesion molecule 1 non-carbohydrate moiety. Sperm hyaluronidase is (SPAM1) active at neutral and acidic pHs which results from - Danio rerio sperm adhesion molecule 1 (Spam1) different active sites in the hyaluronidase domain at the N-terminus of the protein. The hyaluronidase domain also contains a hyaluronic acid (HA) Mutations binding site that plays a role in the signaling Note pathway leading to acrosomal exocytosis. The According to dbSNP, one validated missense SNP protein also contains a zona binding domain at the for SPAM1 is found in the 47th aa position causing C-terminal end. a V to A (rs34633019) substitution. Other SNPs Expression causing synonymous changes are: rs34404662 A/G substitution at the 3rd amino acid residue (Val), According to GNF Expression Atlas 2 Data from rs2285996 A/G substitution at the 184th amino acid U133A and GNF1H Chips, SPAM1 expression is residue (Lys) and rs34978112 C/T substitution at widely limited to testis and epididymis but it was the 330th amino acid residue (Ala). No clinical also found to be expressed in murine kidney and associations with these SNPs have been reported. female reproductive tract. Both rare and abundant SPAM1 transcripts have been found in neoplastic Germinal breast tissue and in a number of other cancers In mice bearing Robertsonian translocation including pharyngeal astatic melanomas and Rb(6.15) and (6.16), reduced Spam1 hyaluronidase gliomas. In normal somatic cells rare transcripts activity was found to cause sperm dysfunction. It have been found in breast tissue and in fetal, was proposed that entrapment of spontaneous placental, and prostate cDNA libraries. Spam1 mutations, owing to recombination Localisation suppression near the Rb junctions was the major effect. SPAM1 is located on the sperm surface and in the According to in vitro mutagenesis experiments the lysosome-derived acrosome, where it is bound to following mutations were detected to have the inner acrosomal membrane. The acrosomal functional consequences: membrane SPAM1 differs biochemically from the - D146N: 80% loss of activity one on the sperm surface. - E148Q: loss of activity Function - R211G: 90% loss of activity SPAM1 is a multifunctional protein; a - E284Q: loss of activity hyaluronidase that acts in penetrating the cumulus, - R287T: loss of activity a receptor for hyaluronic acid induced cell signaling which leads to acrosomal exocytosis and a receptor Implicated in for the zona pellucida surrounding the oocyte. The zona pellucida recognition function is ascribed to Breast cancer the inner acrosomal membrane SPAM1. The Oncogenesis neutral enzyme activity of plasma membrane Increased levels of SPAM1 are noted in invasive

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1164

SPAM1 (sperm adhesion molecule 1 (PH-20 hyaluronidase, Sade A, Banerjee S zona pellucida binding)) and metastatic breast cancer compared to ductal to proximal chromosome 6 and is a candidate for the carcinoma in situ (DCIS). Tumors from African sperm dysfunction in Rb(6.16)24Lub and Rb(6.15)1Ald heterozygotes. Mamm Genome. 1997 Feb;8(2):94-7 American women with invasive and metastatic breast cancer showed higher levels of SPAM1 than Sun L, Feusi E, Sibalic A, Beck-Schimmer B, Wüthrich RP. Expression profile of hyaluronidase mRNA transcripts in Caucasians. Varying levels of SPAM1 in mammary the kidney and in renal cells. Kidney Blood Press Res. tissue may contribute to early invasion and 1998;21(6):413-8 metastasis of breast cancer. Zheng Y, Martin-Deleon PA. Characterization of the Laryngeal cancer genomic structure of the murine Spam1 gene and its promoter: evidence for transcriptional regulation by a Oncogenesis cAMP-responsive element. Mol Reprod Dev. 1999 SPAM1 expression was found to be significantly Sep;54(1):8-16 elevated in primary laryngeal cancer tissue and Godin DA, Fitzpatrick PC, Scandurro AB, Belafsky PC, even higher in metastatic lesions compared with Woodworth BA, Amedee RG, Beech DJ, Beckman BS. normal laryngeal tissue. SPAM1 may therefore be a PH20: a novel tumor marker for laryngeal cancer. Arch useful tumor marker and prognostic tool for Otolaryngol Head Neck Surg. 2000 Mar;126(3):402-4 laryngeal cancer. In squamous cell laryngeal Cherr GN, Yudin AI, Overstreet JW. The dual functions of carcinoma aberrant expression of SPAM1 at late GPI-anchored PH-20: hyaluronidase and intracellular stages of cancer was detected. signaling. Matrix Biol. 2001 Dec;20(8):515-25 Csoka AB, Frost GI, Stern R. The six hyaluronidase-like Colon Cancer genes in the human and mouse genomes. Matrix Biol. Oncogenesis 2001 Dec;20(8):499-508 SPAM1 mRNA was present in mRNA from four Vines CA, Li MW, Deng X, Yudin AI, Cherr GN, Overstreet biopsies obtained from patients with colorectal JW. Identification of a hyaluronic acid (HA) binding domain in the PH-20 protein that may function in cell signaling. Mol cancers. Normal colonic mucosal tissues obtained Reprod Dev. 2001 Dec;60(4):542-52 from the same patients did not express SPAM1 mRNA. In metastatic colon carcinoma cell lines but Zheng Y, Deng X, Zhao Y, Zhang H, Martin-DeLeon PA. Spam1 (PH-20) mutations and sperm dysfunction in mice not in non-metastatic cell lines, SPAM1 expression with the Rb(6.16) or Rb(6.15) translocation. Mamm was detected. Strong angiogenesis developed in Genome. 2001 Nov;12(11):822-9 four of five animals injected with SPAM1+ colon Beech DJ, Madan AK, Deng N. Expression of PH-20 in carcinoma VAC05 cells. However, only one of five normal and neoplastic breast tissue. J Surg Res. 2002 - animals injected with SPAM1 VAC06 cells Apr;103(2):203-7 developed significant angiogenesis. Evans EA, Zhang H, Martin-DeLeon PA. SPAM1 (PH-20) Melanoma protein and mRNA expression in the epididymides of humans and macaques: utilizing laser microdissection/RT- Oncogenesis PCR. Reprod Biol Endocrinol. 2003 Aug 6;1:54 SPAM1 expression is seen in metastatic melanoma Zhang H, Martin-DeLeon PA. Mouse Spam1 (PH-20) is a but not in non-metastatic melanoma cell lines multifunctional protein: evidence for its expression in the (SMMU-2 and SMMU-1 respectively). SPAM1+ female reproductive tract. Biol Reprod. 2003 human melanoma cell line SMMU-2 but not Aug;69(2):446-54 SPAM1- SMMU-1 cells induced angiogenesis in Dunn CA, Mager DL. Transcription of the human and mice cornea although the exact mechanisms of how rodent SPAM1 / PH-20 genes initiates within an ancient endogenous retrovirus. BMC Genomics. 2005 Apr SPAM1 induces angiogenesis is not known. 1;6(1):47 Christopoulos TA, Papageorgakopoulou N, Theocharis References DA, Mastronikolis NS, Papadas TA, Vynios DH. Jones MH, Davey PM, Aplin H, Affara NA. Expression Hyaluronidase and CD44 hyaluronan receptor expression analysis, genomic structure, and mapping to 7q31 of the in squamous cell laryngeal carcinoma. Biochim Biophys human sperm adhesion molecule gene SPAM1. Acta. 2006 Jul;1760(7):1039-45 Genomics. 1995 Oct 10;29(3):796-800 Grigorieva A, Griffiths GS, Zhang H, Laverty G, Shao M, Liu D, Pearlman E, Diaconu E, Guo K, Mori H, Haqqi T, Taylor L, Martin-DeLeon PA. Expression of SPAM1 (PH- Markowitz S, Willson J, Sy MS. Expression of 20) in the murine kidney is not accompanied by hyaluronidase by tumor cells induces angiogenesis in vivo. hyaluronidase activity: evidence for potential roles in fluid Proc Natl Acad Sci U S A. 1996 Jul 23;93(15):7832-7 and water reabsorption. Kidney Blood Press Res. 2007;30(3):145-55 Arming S, Strobl B, Wechselberger C, Kreil G. In vitro mutagenesis of PH-20 hyaluronidase from human sperm. This article should be referenced as such: Eur J Biochem. 1997 Aug 1;247(3):810-4 Sade A, Banerjee S. SPAM1 (sperm adhesion molecule 1 Deng X, Moran J, Copeland NG, Gilbert DJ, Jenkins NA, (PH-20 hyaluronidase, zona pellucida binding)). Atlas Primakoff P, Martin-DeLeon PA. The mouse Spam1 maps Genet Cytogenet Oncol Haematol. 2010; 14(12):1163- 1165.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1165

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Mini Review

TMPRSS2 (transmembrane protease, serine 2) Youngwoo Park Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejon, Korea (YP)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/TMPRSS2ID42592ch21q22.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI TMPRSS2ID42592ch21q22.txt

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

human tissues. Especially, TMPRSS2 is highly Identity expressed in small intestine, but also in lower levels Other names: FLJ41954, PP9284, PRSS10 in several other tissues. Also expressed in prostate, HGNC (Hugo): TMPRSS2 colon, stomach and salivary gland. Location: 21q22.3 Localisation Subcellular location: Cell membrane; Single-pass DNA/RNA type II membrane protein. Description Activated by cleavage and secreted. TMPRSS2 gene approximately extends 43.59 kb- Function long on chromosome 21 in the region q22.3, This gene was demonstrated to be up-regulated by containing 14 exons. androgenic hormones in prostate cancer cells and Transcription down-regulated in androgen-independent prostate Two alternative splicing variants have been cancer tissue. To containing intra- and extracellular described, producing transcripts of 3.25 kb and 3.21 domains, TMPRSS2 could work as a receptor for kb, respectively. specific ligand(s) mediating signals between the environment and the cell. TMPRSS2 has been Protein proposed to regulate epithelial sodium currents in the lung through proteolytic cleavage of the Description epithelial sodium channel and inflammatory TMPRSS2 is a 492 amino acid type II responses in the prostate via the proteolytic transmembrane serine proteases (TTSPs) which are activation of PAR-2. expressed at the cell surface and are thus ideally Homology located to regulate cell-cell and cell-matrix interactions. TTPs (type II transmembrane serine proteases) contain an integral transmembrane domain and Expression remain cell-surface-associated, even after TMPRSS2 is expressed in normal and diseased proteolytic activation of the protease zymogen.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1166

TMPRSS2 (transmembrane protease, serine 2) Park Y

Human TTSPs, which consists of 17 members, analysis of the serine protease domains, namely the were grouped into four subfamilies based on matriptase, corin, hepsin/TMPRSS and HAT/DESC similarity in domain structure and phylogenetic subfamilies.

TMPRSS2 is a 492 amino acid single-pass type II membrane protein. It contains a Serine protease domain (aa 255-492) of the S1 family, followed by a Scavenger receptor cysteine-rich domain (SRDR, aa 149-242) of group A; an LDL receptor class A (LDLRA, aa 113-148) domain forms a binding site for calcium; a predicted transmembrane domain (aa 84-106). Letters H, D and S in the serine protease domain indicate the position of the three catalytic residues histidine, aspartate and serine, respectively.

Multidomain structure of human TTSPs. Human TTSPs were grouped into four subfamilies based on similarity in domain structure and phylogenetic analysis of the serine protease domains, namely the matriptase, corin, hepsin/TMPRSS and HAT/DESC subfamilies. Consensus domains are shown below. Each diagram was drawn using the web-based SMART software (http://smart.embl-heidelberg.de) with TTSP amino acid sequences obtained from GenBank. Abbreviations: CUB, Cls/Clr, urchin embryonic growth factor and bone morphogenic protein-1 domain; DESC1, differentially expressed squamous cell carcinoma gene 1; FRZ, frizzled domain; HAT, human airway trypsin-like protease; LDLA, low-density lipoprotein receptor domain class A; MAM, a meprin, A5 antigen and receptor protein phosphatase m domain; MSPL, mosaic serine protease long-form; Polyserase-1, polyserine protease-1; SEA, a single sea urchin sperm protein, enteropeptidase, agrin domain; SR, scavenger receptor cysteine-rich domain; TM, transmembrane domain. Letters H, D and S in the serine protease domain (active) indicate the position of the three catalytic residues histidine, aspartate and serine, respectively. Letter A in the serine protease domain (inactive) indicates a serine to alanine exchange.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1167

TMPRSS2 (transmembrane protease, serine 2) Park Y

its gene turned out to be expressed mainly in the Implicated in prostate in an androgen dependent manner. Prostate cancer In the prostate adenocarcinoma, TMPRSS2-EGR fusion mRNAs is highly expressed. Because of its Prognosis location on the surface of prostatic cells, TMPRSS2 TMPRSS2 was originally reported to be a small is a potential new diagnostic marker for prostate intestine-associated serine protease. Later, however, cancer. Breakpoints

References Paoloni-Giacobino A, Chen H, Peitsch MC, Rossier C, Bugge TH, Antalis TM, Wu Q. Type II transmembrane Antonarakis SE. Cloning of the TMPRSS2 gene, which serine proteases. J Biol Chem. 2009 Aug encodes a novel serine protease with transmembrane, 28;284(35):23177-81 LDLRA, and SRCR domains and maps to 21q22.3. Genomics. 1997 Sep 15;44(3):309-20 Choi SY, Bertram S, Glowacka I, Park YW, Pöhlmann S. Type II transmembrane serine proteases in cancer and Vaarala MH, Porvari K, Kyllönen A, Lukkarinen O, Vihko P. viral infections. Trends Mol Med. 2009 Jul;15(7):303-12 The TMPRSS2 gene encoding transmembrane serine protease is overexpressed in a majority of prostate cancer Barwick BG, Abramovitz M, Kodani M, Moreno CS, Nam patients: detection of mutated TMPRSS2 form in a case of R, Tang W, Bouzyk M, Seth A, Leyland-Jones B. Prostate aggressive disease. Int J Cancer. 2001 Dec 1;94(5):705- cancer genes associated with TMPRSS2-ERG gene fusion 10 and prognostic of biochemical recurrence in multiple cohorts. Br J Cancer. 2010 Feb 2;102(3):570-6 Vaarala MH, Porvari KS, Kellokumpu S, Kyllönen AP, Vihko PT. Expression of transmembrane serine protease This article should be referenced as such: TMPRSS2 in mouse and human tissues. J Pathol. 2001 Jan;193(1):134-40 Park Y. TMPRSS2 (transmembrane protease, serine 2). Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12):1166-1168.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1168

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

TMSB10 (thymosin beta 10) Xueshan Qiu Department of Pathology, the First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, China (XQ)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/TMSB10ID42595ch2p11.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI TMSB10ID42595ch2p11.txt

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 Localisation TMSB10 is expressed in cytoplasm. Other names: MIG12, TB10, THYB10 HGNC (Hugo): TMSB10 Function Location: 2p11.2 Overview: TMSB10 is a highly conserved small Note: TMSB10 is a member of the beta-thymosin acid protein. It is present in many tissues and cell family, which is an actin-sequestering protein types. It can sequester actin monomers and bind to involved in cell motility. TMSB10 may be G-actin in a 1:1 complex. correlated with tumor cells proliferation, apoptosis, Actin monomer sequestering protein: TMSB10 is metastasis and angiogenesis. one of G-actin binding proteins, being expressed in all mammalian species. It can sequester monomeric DNA/RNA actin and inhibit actin polymerization. It participates in the regulation of cancer cell motility. Description Development: The expression of TMSB10 is The cDNA sequence for human TMSB10 is 482 associated with the development of several tissues. nucleotides long comprising 3 exons. The open It is involved in the development of the oral cavity reading frame of the coding region is 135 bp. and its annexes. TMSB10 plays an important role in early neuroembryogenesis. It is present in the Protein developing nervous, and has a specific physiological function during cerebellum Description development. TMSB10 is only present at very low Protein length (NP_066926.1): 44 amino acids. levels in a very small subpopulation of glia in the (madkpdmgeiasfdkaklkktetqekntlptketieqekrseis) adult cerebellum. In young animals, most of the Molecular weight: 4.9 kDa. TMSB10 is a small G- TMSB10 is localized in granule cells, Golgi actin binding protein, and it induces neurons and Purkinje cells. In old animals, depolymerization of intracellular F-actin pools by TMSB10 signal is detected faintly in a few Purkinje sequestering actin monomers. cells. Expression Apoptosis: TMSB10 regulates apoptosis. For example, upregulation of TMSB10 in M. bovis- TMSB10 is found in human, rat, mouse, cat, and infected macrophages is linked with increased cell rabbit. TMSB10 is one of the most abundant beta- death due to apoptosis. .

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1169

TMSB10 (thymosin beta 10) Qiu X

TMSB10 can disrupt F-actin stress fibers, markedly actin as well as in breast cancer cell motility. decrease ovarian cancer cells growth, and a high Ovarian cancer rate of apoptosis. TMSB10 plays a significant role in cell apoptosis possibly by acting as an actin- Note mediated tumor suppressor, perhaps functions as a TMSB10 inhibits angiogenesis and tumor growth neoapoptotic influence during embryogenesis, and by interfering Ras signal transduction and may mediate some of the pro-apoptotic anticancer expression of VEGF. actions of retinoids. Thyroid neoplasias Angiogenesis: TMSB10 may be an effective Note inhibitor of angiogenesis by inhibiting endothelial migration, tube formation, VEGF, VEGFR-1 and TMSB10 overexpression is a general event of integrin alphaV expression in HCAECs. TMSB10 thyroid cell neoplastic transformation. An increased is not only a cytoskeletal regulator, it also acts as a expression of TMSB10 mRNA in thyroid potent inhibitor of angiogenesis and tumor growth carcinomas is found. The evaluation of TMSB10 by interaction with Ras. gene expression may be considered a promising Cancers: Elevated expression of TMSB10 is means of human thyroid hyperproliferative associated with invasion and metastasis of several diagnosis. By decreasing TMSB10 expression, the kinds of tumors. It may be considered a potential thyroid cancer cells growth in soft agar is inhibited. tool for the diagnosis of several human neoplasias. Renal cell carcinoma TMSB10 is detected mainly in the malignant tissue, Note particularly in the cancerous cells, whereas the The TMSB10 gene is deregulated in renal cell normal cell population around the lesions showed carcinoma and it may be a new molecular marker very weak staining. Also, the intensity of staining in for renal-cell carcinoma. the cancerous cells is proportionally increased with the increasing grade of the lesions. Cutaneous melanoma Homology Note TMSB10 can be considered as a new progression Homolog to thymosin beta 4 and thymosin beta 15. marker for human cutaneous melanoma. Implicated in References Pancreatic cancer Erickson-Viitanen S, Ruggieri S, Natalini P, Horecker BL. Note Thymosin beta 10, a new analog of thymosin beta 4 in mammalian tissues. Arch Biochem Biophys. 1983 TSMB10 is expressed in human pancreatic Sep;225(2):407-13 carcinoma, but not in non-neoplastic pancreatic Abiko T, Sekino H. Synthesis of deacetyl-thymosin beta 10 tissue, suggesting a role for TMSB10 in the and examination of its immunological effect on T-cell carcinogenesis of pancreatic carcinoma. It is a subpopulations of a uremic patient with tuberculosis. Chem promising marker and a novel therapeutic target for Pharm Bull (Tokyo). 1986 Nov;34(11):4708-17 pancreatic cancer. Exogenous TMSB10 causes the Goodall GJ, Horecker BL. Molecular cloning of the cDNA phosphorylation of JNK and increases the secretion for rat spleen thymosin beta 10 and the deduced amino of cytokines interleukin (IL)-7 and IL-8 in BxPC-3 acid sequence. Arch Biochem Biophys. 1987 pancreatic cancer cells. Jul;256(1):402-5 McCreary V, Kartha S, Bell GI, Toback FG. Sequence of a Non-small cell lung cancer human kidney cDNA clone encoding thymosin beta 10. Note Biochem Biophys Res Commun. 1988 Apr 29;152(2):862-6 TMSB10 might induce microvascular and Hall AK, Hempstead J, Morgan JI. Thymosin beta 10 levels lymphatic vessel formation by up-regulating in developing human brain and its regulation by retinoic vascular endothelial growth factor and vascular acid in the HTB-10 neuroblastoma. Brain Res Mol Brain Res. 1990 Jul;8(2):129-35 endothelial growth factor-C in lung cancer tissues, thus promoting the distant and lymph node Lin SC, Morrison-Bogorad M. Developmental expression of mRNAs encoding thymosins beta 4 and beta 10 in rat metastases and being implicated in the progression brain and other tissues. J Mol Neurosci. 1990;2(1):35-44 of non-small cell lung cancer. Hall AK. Developmental regulation of thymosin beta 10 Breast cancer mRNA in the human brain. Brain Res Mol Brain Res. 1991 Jan;9(1-2):175-7 Note TMSB10 plays a key role in sequestration of G- Hall AK. Differential expression of thymosin genes in

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1170

TMSB10 (thymosin beta 10) Qiu X

human tumors and in the developing human kidney. Int J Califano D, Monaco C, Santelli G, Giuliano A, Veronese Cancer. 1991 Jul 9;48(5):672-7 ML, Berlingieri MT, de Franciscis V, Berger N, Trapasso F, Santoro M, Viglietto G, Fusco A. Thymosin beta-10 gene Hall AK. Retinoic acid and serum modulation of thymosin overexpression correlated with the highly malignant beta-10 gene expression in rat neuroblastoma cells. J Mol neoplastic phenotype of transformed thyroid cells in vivo Neurosci. 1991;2(4):229-37 and in vitro. Cancer Res. 1998 Feb 15;58(4):823-8 Hall AK, Chen SC, Hempstead JL, Morgan JI. Retinoic Carpintero P, Anadón R, Gómez-Márquez J. Expression of acid regulates thymosin beta 10 levels in rat the thymosin beta10 gene in normal and kainic acid- neuroblastoma cells. J Neurochem. 1991 Feb;56(2):462-8 treated rat forebrain. Brain Res Mol Brain Res. 1999 Jun Lin SC, Morrison-Bogorad M. Cloning and characterization 18;70(1):141-6 of a testis-specific thymosin beta 10 cDNA. Expression in Santelli G, Califano D, Chiappetta G, Vento MT, Bartoli post-meiotic male germ cells. J Biol Chem. 1991 Dec PC, Zullo F, Trapasso F, Viglietto G, Fusco A. Thymosin 5;266(34):23347-53 beta-10 gene overexpression is a general event in human Lugo DI, Chen SC, Hall AK, Ziai R, Hempstead JL, Morgan carcinogenesis. Am J Pathol. 1999 Sep;155(3):799-804 JI. Developmental regulation of beta-thymosins in the rat Vassiliadou I, Leondiadis L, Ferderigos N, Ithakissios DS, central nervous system. J Neurochem. 1991 Evangelatos GP, Livaniou E. Investigation of the epitopic Feb;56(2):457-61 structure of thymosin beta10 by epitope mapping Condon MR, Hall AK. Expression of thymosin beta-4 and experiments. . 1999;20(3):411-4 related genes in developing human brain. J Mol Neurosci. Viglietto G, Califano D, Bruni P, Baldassarre G, Vento MT, 1992;3(3):165-70 Belletti B, Fedele M, Santelli G, Boccia A, Manzo G, Hall AK. Retinoids and a retinoic acid receptor differentially Santoro M, Fusco A. Regulation of thymosin beta10 modulate thymosin beta 10 gene expression in transfected expression by TSH and other mitogenic signals in the neuroblastoma cells. Cell Mol Neurobiol. 1992 thyroid gland and in cultured thyrocytes. Eur J Endocrinol. Feb;12(1):45-58 1999 Jun;140(6):597-607 Border BG, Lin SC, Griffin WS, Pardue S, Morrison- Anadón R, Rodríguez Moldes I, Carpintero P, Evangelatos Bogorad M. Alterations in actin-binding beta-thymosin G, Livianou E, Leondiadis L, Quintela I, Cerviño MC, expression accompany neuronal differentiation and Gómez-Márquez J. Differential expression of thymosins migration in rat cerebellum. J Neurochem. 1993 beta(4) and beta(10) during rat cerebellum postnatal Dec;61(6):2104-14 development. Brain Res. 2001 Mar 16;894(2):255-65 Nachmias VT. Small actin-binding proteins: the beta- Bani-Yaghoub M, Felker JM, Ozog MA, Bechberger JF, thymosin family. Curr Opin Cell Biol. 1993 Feb;5(1):56-62 Naus CC. Array analysis of the genes regulated during neuronal differentiation of human embryonal cells. Weterman MA, van Muijen GN, Ruiter DJ, Bloemers HP. Biochem Cell Biol. 2001;79(4):387-98 Thymosin beta-10 expression in melanoma cell lines and melanocytic lesions: a new progression marker for human Koutrafouri V, Leondiadis L, Avgoustakis K, Livaniou E, cutaneous melanoma. Int J Cancer. 1993 Jan Czarnecki J, Ithakissios DS, Evangelatos GP. Effect of 21;53(2):278-84 thymosin peptides on the chick chorioallantoic membrane angiogenesis model. Biochim Biophys Acta. 2001 Nov Yu FX, Lin SC, Morrison-Bogorad M, Atkinson MA, Yin HL. 7;1568(1):60-6 Thymosin beta 10 and thymosin beta 4 are both actin monomer sequestering proteins. J Biol Chem. 1993 Jan Lee SH, Zhang W, Choi JJ, Cho YS, Oh SH, Kim JW, Hu 5;268(1):502-9 L, Xu J, Liu J, Lee JH. Overexpression of the thymosin beta-10 gene in human ovarian cancer cells disrupts F- Hall AK. Amplification-independent overexpression of actin stress fiber and leads to apoptosis. Oncogene. 2001 thymosin beta-10 mRNA in human renal cell carcinoma. Oct 11;20(46):6700-6 Ren Fail. 1994;16(2):243-54 Vasile E, Tomita Y, Brown LF, Kocher O, Dvorak HF. Yu FX, Lin SC, Morrison-Bogorad M, Yin HL. Effects of Differential expression of thymosin beta-10 by early thymosin beta 4 and thymosin beta 10 on actin structures passage and senescent vascular endothelium is in living cells. Cell Motil Cytoskeleton. 1994;27(1):13-25 modulated by VPF/VEGF: evidence for senescent endothelial cells in vivo at sites of atherosclerosis. FASEB Hall AK. Thymosin beta-10 accelerates apoptosis. Cell Mol J. 2001 Feb;15(2):458-66 Biol Res. 1995;41(3):167-80 Gómez-Márquez J, Anadón R. The beta-thymosins, small Voisin PJ, Pardue S, Morrison-Bogorad M. Developmental actin-binding peptides widely expressed in the developing characterization of thymosin beta 4 and beta 10 and adult cerebellum. Cerebellum. 2002 Apr;1(2):95-102 expression in enriched neuronal cultures from rat cerebella. J Neurochem. 1995 Jan;64(1):109-20 Gutiérrez-Pabello JA, McMurray DN, Adams LG. Upregulation of thymosin beta-10 by Mycobacterium bovis Carpintero P, Franco del Amo F, Anadón R, Gómez- infection of bovine macrophages is associated with Márquez J. Thymosin beta10 mRNA expression during apoptosis. Infect Immun. 2002 Apr;70(4):2121-7 early postimplantation mouse development. FEBS Lett. 1996 Sep 23;394(1):103-6 Otto AM, Müller CS, Huff T, Hannappel E. Chemotherapeutic drugs change actin skeleton Verghese-Nikolakaki S, Apostolikas N, Livaniou E, organization and the expression of beta-thymosins in Ithakissios DS, Evangelatos GP. Preliminary findings on human breast cancer cells. J Cancer Res Clin Oncol. 2002 the expression of thymosin beta-10 in human breast May;128(5):247-56 cancer. Br J Cancer. 1996 Nov;74(9):1441-4 Santelli G, Bartoli PC, Giuliano A, Porcellini A, Mineo A, Huff T, Müller CS, Hannappel E. C-terminal truncation of Barone MV, Busiello I, Trapasso F, Califano D, Fusco A. thymosin beta10 by an intracellular protease and its Thymosin beta-10 protein synthesis suppression reduces influence on the interaction with G-actin studied by the growth of human thyroid carcinoma cells in semisolid ultrafiltration. FEBS Lett. 1997 Sep 1;414(1):39-44 medium. Thyroid. 2002 Sep;12(9):765-72

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1171

TMSB10 (thymosin beta 10) Qiu X

Takano T, Hasegawa Y, Miyauchi A, Matsuzuka F, 7;280(40):34003-7 Yoshida H, Kuma K, Amino N. Quantitative analysis of thymosin beta-10 messenger RNA in thyroid carcinomas. Mu H, Ohashi R, Yang H, Wang X, Li M, Lin P, Yao Q, Jpn J Clin Oncol. 2002 Jul;32(7):229-32 Chen C. Thymosin beta10 inhibits cell migration and capillary-like tube formation of human coronary artery Tokuriki M, Noda I, Saito T, Narita N, Sunaga H, Tsuzuki endothelial cells. Cell Motil Cytoskeleton. 2006 H, Ohtsubo T, Fujieda S, Saito H. Gene expression Apr;63(4):222-30 analysis of human middle ear cholesteatoma using complementary DNA arrays. Laryngoscope. 2003 Maelan AE, Rasmussen TK, Larsson LI. Localization of May;113(5):808-14 thymosin beta10 in breast cancer cells: relationship to actin cytoskeletal remodeling and cell motility. Histochem Chiappetta G, Pentimalli F, Monaco M, Fedele M, Cell Biol. 2007 Jan;127(1):109-13 Pasquinelli R, Pierantoni GM, Ribecco MT, Santelli G, Califano D, Pezzullo L, Fusco A. Thymosin beta-10 gene Gu Y, Wang C, Wang Y, Qiu X, Wang E. Expression of expression as a possible tool in diagnosis of thyroid thymosin beta10 and its role in non-small cell lung cancer. neoplasias. Oncol Rep. 2004 Aug;12(2):239-43 Hum Pathol. 2009 Jan;40(1):117-24 Liu CR, Ma CS, Ning JY, You JF, Liao SL, Zheng J. Li M, Zhang Y, Zhai Q, Feurino LW, Fisher WE, Chen C, Differential thymosin beta 10 expression levels and actin Yao Q. Thymosin beta-10 is aberrantly expressed in filament organization in tumor cell lines with different pancreatic cancer and induces JNK activation. Cancer metastatic potential. Chin Med J (Engl). 2004 Invest. 2009 Mar;27(3):251-6 Feb;117(2):213-8 Nemolato S, Messana I, Cabras T, Manconi B, Inzitari R, Rho SB, Chun T, Lee SH, Park K, Lee JH. The interaction Fanali C, Vento G, Tirone C, Romagnoli C, Riva A, Fanni between E-tropomodulin and thymosin beta-10 rescues D, Di Felice E, Faa G, Castagnola M. Thymosin beta(4) tumor cells from thymosin beta-10 mediated apoptosis by and beta(10) levels in pre-term newborn oral cavity and restoring actin architecture. FEBS Lett. 2004 Jan 16;557(1- foetal salivary glands evidence a switch of secretion during 3):57-63 foetal development. PLoS One. 2009;4(4):e5109 Alldinger I, Dittert D, Peiper M, Fusco A, Chiappetta G, Sribenja S, Li M, Wongkham S, Wongkham C, Yao Q, Staub E, Lohr M, Jesnowski R, Baretton G, Ockert D, Chen C. Advances in thymosin beta10 research: Saeger HD, Grützmann R, Pilarsky C. Gene expression differential expression, molecular mechanisms, and clinical analysis of pancreatic cell lines reveals genes implications in cancer and other conditions. Cancer Invest. overexpressed in pancreatic cancer. Pancreatology. 2009 Dec;27(10):1016-22 2005;5(4-5):370-9 Zhang T, Li X, Yu W, Yan Z, Zou H, He X. Overexpression Lee SH, Son MJ, Oh SH, Rho SB, Park K, Kim YJ, Park of thymosin beta-10 inhibits VEGF mRNA expression, MS, Lee JH. Thymosin {beta}(10) inhibits angiogenesis autocrine VEGF protein production, and tube formation in and tumor growth by interfering with Ras function. Cancer hypoxia-induced monkey choroid-retinal endothelial cells. Res. 2005 Jan 1;65(1):137-48 Ophthalmic Res. 2009;41(1):36-43 Rho SB, Lee KW, Chun T, Lee SH, Park K, Lee JH. The This article should be referenced as such: identification of apoptosis-related residues in human thymosin beta-10 by mutational analysis and Qiu X. TMSB10 (thymosin beta 10). Atlas Genet Cytogenet computational modeling. J Biol Chem. 2005 Oct Oncol Haematol. 2010; 14(12):1169-1172.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1172

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Gene Section Review

TYMP (thymidine phosphorylase) Irene V Bijnsdorp, Godefridus J Peters Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands (IVB, GJP)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/TYMPID40397ch22q13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI TYMPID40397ch22q13.txt

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

is involved in nucleotide synthesis and thymidine Identity phosphorolysis. Other names: ECGF1, hPD-ECGF, MNGIE, PDECGF, TP Description HGNC (Hugo): TYMP Thymidine phosphorylase is located at chromosome 22 in the region of q13.33. cDNA is approximately Location: 22q13.33 1.8 kb long, consisting of 10 exons in a 4.3 kb region (Hagiwara et al., 1991; Stenman et al., DNA/RNA 1992). TP was first cloned and sequenced in 1989 Note (Ishikawa et al., 1989). The nucleic acid sequence The TP gene encodes an angiogenic factor which of TP is highly conserved, the human TP shares promotes angiogenesis both in vitro and in vivo and 39% sequence identity with that of E. coli (Barton et al., 1992).

TYMP is located on chromosome 22 of which 3 transcripts have been identified.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1173

TYMP (thymidine phosphorylase) Bijnsdorp IV, Peters GJ

Transcription deoxyribosyl transferase activity by which the deoxyribosyl moiety is transferred from a The promoter region of the TP gene has no TATA pyrimidine nucleoside to another pyrimidine base. box or CCAAT box, but has a high G-C content Subsequently a new pyrimidine nucleoside is and seven copies of the SP-1 binding site upstream formed. from the transcription start site. The sugars that are formed by degradation of Exact TP gene regulation is unknown, but has been thymidine are thought to play a role in the induction described to be (indirectly) regulated by NFkB, of angiogenesis. Deoxyribose-1-P can be converted TNF-alpha and IFN-gamma (Waguri et al., 1997; to deoxyribose-5-phosphate or degraded to Zhu et al., 2002; Zhu et al., 2003; Eda et al., 1993; deoxyribose. Deoxyribose can be secreted, and de Bruin et al., 2004). possibly attract endothelial cells to form new blood vessels (reviewed in de Bruin et al., 2006; Liekens Protein et al., 2007; Bronckaers et al., 2009). TP in some Note cancer cells can also increase their invasive Thymidine phosphorylase was first identified as the potential, although the exact mechanism remains platelet-derived endothelial cell growth factor, unclear. because it was related to endothelial cell growth TP can also activate or inactivate several (Miyazono et al., 1987; Ishikawa et al., 1989). Later pyrimidines or pyrimidine nucleoside analogs with on, it was found that it was identical to thymidine antiviral and antitumoral activity, such as phosphorylase (Furukawa et al., 1992). Thymidine inactivation of trifluorothymidine (TFT) phosphorylase (TP) is the most correct name to (Heidelberger et al., 1964) and 5-fluoro-2'- refer to this protein, since it catalyzes the deoxyruidine (van Laar et al., 1998), or activation phopshorolysis of thymidine to thymine. TP of 5-fluorouracil (5-FU) (Schwartz et al., 1995) and undergoes limited post-translational modification 5-fluoro-5'-deoxyuridine (5'DFUR). and is not glycosylated. Covalent linkage between Homology serine residues of TP and phosphate groups of The TYMP gene is conserved in chimpanzee, nucleotides has been observed, which may facilitate mouse, rat, and zebrafish. secretion of the protein (Usuki et al., 1991). However, TP does not contain a classical secretion signal (Ishikawa et al., 1989). TP is a dimer, Mutations consisting of two identical subunits that are non- Note covalently associated (Desgranges et al., 1981) with Mutations in this gene have been associated with its dimeric molecular mass ranging from 90 kD in mitochondrial neurogastrointestinal Escherichia coli to 110 kD in mammals (Schwartz, encephalomyopathy (MNGIE). Multiple 1978; Desgranges et al., 1981). alternatively spliced variants, encoding the same Description protein, have been identified. TP protein does not contain a known receptor Implicated in binding region or a secretion signal (Ishikawa et al., 1989). It is implicated in nucleotide synthesis and Various cancer degradation of thymidine. TP is also implicated in Note angiogenesis (reviewed in de Bruin et al., 2006; TP in tumor sites can be expressed in the cancer Liekens et al., 2007; Bronckaers et al., 2009). cells, in the most malignant cells, tumor stromal Expression cells (such as macrophages) or in the invasive part of the tumor (van Triest et al., 1999). A high TP TP is highly expressed in liver tissues. Furthermore, expression and activity have been related to a poor TP is often overexpressed in tumor sites and is outcome and increased angiogenesis. The TP gene involved in inflammatory diseases, such as is regulated by many other factors that are rheumatoid arthritis. implicated in cancer, such as NFkB (de Bruin et al., Localisation 2004). TP regulates the expression of IL-8, and TP is expressed in the cytoplasm and the nucleus possibly also that of other genes, although the exact (Fox et al., 1995). mechanism of this regulation is still unclear (Brown et al., 2000; Bijnsdorp et al., 2008). The high TP Function activity in the tumor can selectively activate the TP catalyzes the phosphorolysis of thymidine 5FU prodrug 5'-deoxy-5-fluorouridine to 5FU. (TdR) to thymine and 2-deoxy-alpha-D-ribose 1- 5'deoxy-5-fluorouridine is an intermediate of the phosphate (dR-1-P). TP can also catalyze the oral 5FU prodrug Capecitabine (Xeloda) (de Bruin formation of thymidine from thymine and dR-1-P. et al., 2006). On the other hand TP can inactivate TP also catalyzes the phosphorolysis of the fluoropyrimidine trifluorothymidine (TFT), deoxyuridine to uracil and dR-1-P. TP also has which is registered as the antiviral drug Viroptic®

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1174

TYMP (thymidine phosphorylase) Bijnsdorp IV, Peters GJ

(De Clercq, 2004). An inhibitor of TP, TPI, will Chronic glomerulonephritis prevent inactivation of TFT. TAS-102 is a Note combination of TFT and TPI (in a molar ratio of TP is upregulated in chronic glomerulonephritis (a 1:0.5) which is developed as an anticancer drug renal disease characterized by inflammation of the (Temmink et al., 2007). glomeruli) where it probably plays a critical role in Disease the progression of interstitial fibrosis (Wang et al., Gastrointestinal tumors (Fox et al., 1995; 2006). Yoshikawa et al., 1999; Kimura et al., 2002; Takebayashi et al., 1996), breast cancer Mitochondrial neurogastrointestinal (Moghaddam et al., 1995), bladder cancer (O'Brien encephalomyopathy (MNGIE) et al., 1996). Note Prognosis An autosomal recessive disorder involving DNA High expression is often related to a poor alterations (Bardosi et al., 1987). Gene mutations in prognosis, an increased microvessel density and the TP gene include missense, splice sites increased metastasis. microdeletions and single nucleotide insertions Abnormal protein (Spinazzola et al., 2002; Nishino et al., 2000). These mutations are associated with a severe No fusion protein has been described. reduction in TP activity. This leads to increased Rheumatoid arthritis thymidine plasma levels, and increased Note deoxyuridine levels (which is also a substrate for Elevated levels of (circulating) PD-ECGF (TP) TP). were found in rheumatoid arthritis patients (Asai et Prognosis al., 1993). In the sera and synovial fluids of patients Not determined. suffering from rheumatoid arthritis PD-ECGF (TP) was detected at high levels (Asai et al., 1993). In References addition, there was a significant positive correlation HEIDELBERGER C, ANDERSON SW. FLUORINATED between PD-ECGF (TP) levels in synovial fluid PYRIMIDINES. XXI. THE TUMOR-INHIBITORY ACTIVITY and in serum (Asai et al., 1993). The elevated PD- OF 5-TRIFLUOROMETHYL-2'-DEOXYURIDINE. Cancer ECGF (TP) levels presumably arise through Res. 1964 Dec;24:1979-85 induction of PD-ECGF (TP) in synoviocytes, Schwartz M. Thymidine phosphorylase from Escherichia resulting from aberrant production of cytokines like coli. Methods Enzymol. 1978;51:442-5 TNF-alpha and IL-1 (Waguri et al., 1997). Desgranges C, Razaka G, Rabaud M, Bricaud H. Atherosclerosis Catabolism of thymidine in human blood platelets: purification and properties of thymidine phosphorylase. Note Biochim Biophys Acta. 1981 Jul 27;654(2):211-8 TP is expressed in atherosclerosis. Macrophages, Bardosi A, Creutzfeldt W, DiMauro S, Felgenhauer K, foam cells and giant cells from both aortic and Friede RL, Goebel HH, Kohlschütter A, Mayer G, Rahlf G, coronary plaques expressed TP, suggesting that TP Servidei S. Myo-, neuro-, gastrointestinal encephalopathy may play a role in the pathogenesis of (MNGIE syndrome) due to partial deficiency of cytochrome-c-oxidase. A new mitochondrial multisystem atherosclerosis (Boyle et al., 2000). disorder. Acta Neuropathol. 1987;74(3):248-58 Psoriasis Miyazono K, Okabe T, Urabe A, Takaku F, Heldin CH. Note Purification and properties of an endothelial cell growth factor from human platelets. J Biol Chem. 1987 Mar Increased PD-ECGF (TP) mRNA and 25;262(9):4098-103 immunoreactivity were found in lesional psoriasis Ishikawa F, Miyazono K, Hellman U, Drexler H, Wernstedt compared to the non-lesional skin (Creamer et al., C, Hagiwara K, Usuki K, Takaku F, Risau W, Heldin CH. 1997). In another study it was reported that the Identification of angiogenic activity and the cloning and thymidine phosphorylase activity was twenty-fold expression of platelet-derived endothelial cell growth higher in psoriatic lesions than in normal skin factor. Nature. 1989 Apr 13;338(6216):557-62 (Hammerberg et al., 1991). Hagiwara K, Stenman G, Honda H, Sahlin P, Andersson A, Miyazono K, Heldin CH, Ishikawa F, Takaku F. Inflammatory bowel disease Organization and chromosomal localization of the human Note platelet-derived endothelial cell growth factor gene. Mol Cell Biol. 1991 Apr;11(4):2125-32 In inflammatory bowel disease, TP has been found to be overexpressed, predominantly in macrophages Hammerberg C, Fisher GJ, Voorhees JJ, Cooper KD. Elevated thymidine phosphorylase activity in psoriatic and fibroblasts of the inflamed colonic mucosa. The lesions accounts for the apparent presence of an grade of expression augmented with an increasing epidermal "growth inhibitor," but is not in itself growth grade of inflammation. In addition, TP was found in inhibitory. J Invest Dermatol. 1991 Aug;97(2):286-90 the endothelial cells of the inflamed colonic mucosa Usuki K, Miyazono K, Heldin CH. Covalent linkage (Giatromanolaki et al., 2003; Saito et al., 2003). between nucleotides and platelet-derived endothelial cell

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1175

TYMP (thymidine phosphorylase) Bijnsdorp IV, Peters GJ

growth factor. J Biol Chem. 1991 Oct 25;266(30):20525-31 Yoshikawa T, Suzuki K, Kobayashi O, Sairenji M, Motohashi H, Tsuburaya A, Nakamura Y, Shimizu A, Barton GJ, Ponting CP, Spraggon G, Finnis C, Sleep D. Yanoma S, Noguchi Y. Thymidine phosphorylase/platelet- Human platelet-derived endothelial cell growth factor is derived endothelial cell growth factor is upregulated in homologous to Escherichia coli thymidine phosphorylase. advanced solid types of gastric cancer. Br J Cancer. 1999 Protein Sci. 1992 May;1(5):688-90 Mar;79(7-8):1145-50 Furukawa T, Yoshimura A, Sumizawa T, Haraguchi M, Boyle JJ, Wilson B, Bicknell R, Harrower S, Weissberg PL, Akiyama S, Fukui K, Ishizawa M, Yamada Y. Angiogenic Fan TP. Expression of angiogenic factor thymidine factor. Nature. 1992 Apr 23;356(6371):668 phosphorylase and angiogenesis in human Stenman G, Sahlin P, Dumanski JP, Hagiwara K, Ishikawa atherosclerosis. J Pathol. 2000 Oct;192(2):234-42 F, Miyazono K, Collins VP, Heldin CH. Regional Brown NS, Jones A, Fujiyama C, Harris AL, Bicknell R. localization of the human platelet-derived endothelial cell Thymidine phosphorylase induces carcinoma cell oxidative growth factor (ECGF1) gene to chromosome 22q13. stress and promotes secretion of angiogenic factors. Cytogenet Cell Genet. 1992;59(1):22-3 Cancer Res. 2000 Nov 15;60(22):6298-302 Asai K, Hirano T, Matsukawa K, Kusada J, Takeuchi M, Nishino I, Spinazzola A, Papadimitriou A, Hammans S, Otsuka T, Matsui N, Kato T. High concentrations of Steiner I, Hahn CD, Connolly AM, Verloes A, Guimarães J, immunoreactive gliostatin/platelet-derived endothelial cell Maillard I, Hamano H, Donati MA, Semrad CE, Russell JA, growth factor in synovial fluid and serum of rheumatoid Andreu AL, Hadjigeorgiou GM, Vu TH, Tadesse S, arthritis. Clin Chim Acta. 1993 Sep 17;218(1):1-4 Nygaard TG, Nonaka I, Hirano I, Bonilla E, Rowland LP, Eda H, Fujimoto K, Watanabe S, Ura M, Hino A, Tanaka Y, DiMauro S, Hirano M. Mitochondrial neurogastrointestinal Wada K, Ishitsuka H. Cytokines induce thymidine encephalomyopathy: an autosomal recessive disorder due phosphorylase expression in tumor cells and make them to thymidine phosphorylase mutations. Ann Neurol. 2000 more susceptible to 5'-deoxy-5-fluorouridine. Cancer Jun;47(6):792-800 Chemother Pharmacol. 1993;32(5):333-8 van Triest B, Pinedo HM, Blaauwgeers JL, van Diest PJ, Takeuchi M, Otsuka T, Matsui N, Asai K, Hirano T, Schoenmakers PS, Voorn DA, Smid K, Hoekman K, Moriyama A, Isobe I, Eksioglu YZ, Matsukawa K, Kato T. Hoitsma HF, Peters GJ. Prognostic role of thymidylate Aberrant production of gliostatin/platelet-derived synthase, thymidine phosphorylase/platelet-derived endothelial cell growth factor in rheumatoid synovium. endothelial cell growth factor, and proliferation markers in Arthritis Rheum. 1994 May;37(5):662-72 colorectal cancer. Clin Cancer Res. 2000 Mar;6(3):1063- 72 Fox SB, Moghaddam A, Westwood M, Turley H, Bicknell R, Gatter KC, Harris AL. Platelet-derived endothelial cell Kimura H, Konishi K, Kaji M, Maeda K, Yabushita K, Miwa growth factor/thymidine phosphorylase expression in A. Correlation between expression levels of thymidine normal tissues: an immunohistochemical study. J Pathol. phosphorylase (dThdPase) and clinical features in human 1995 Jun;176(2):183-90 gastric carcinoma. Hepatogastroenterology. 2002 May- Jun;49(45):882-6 Moghaddam A, Zhang HT, Fan TP, Hu DE, Lees VC, Turley H, Fox SB, Gatter KC, Harris AL, Bicknell R. Spinazzola A, Marti R, Nishino I, Andreu AL, Naini A, Thymidine phosphorylase is angiogenic and promotes Tadesse S, Pela I, Zammarchi E, Donati MA, Oliver JA, tumor growth. Proc Natl Acad Sci U S A. 1995 Feb Hirano M. Altered thymidine metabolism due to defects of 14;92(4):998-1002 thymidine phosphorylase. J Biol Chem. 2002 Feb 8;277(6):4128-33 Schwartz EL, Baptiste N, Wadler S, Makower D. Thymidine phosphorylase mediates the sensitivity of Zhu GH, Lenzi M, Schwartz EL. The Sp1 transcription human colon carcinoma cells to 5-fluorouracil. J Biol factor contributes to the tumor necrosis factor-induced Chem. 1995 Aug 11;270(32):19073-7 expression of the angiogenic factor thymidine phosphorylase in human colon carcinoma cells. O'Brien TS, Fox SB, Dickinson AJ, Turley H, Westwood M, Oncogene. 2002 Dec 5;21(55):8477-85 Moghaddam A, Gatter KC, Bicknell R, Harris AL. Expression of the angiogenic factor thymidine Giatromanolaki A, Sivridis E, Maltezos E, Papazoglou D, phosphorylase/platelet-derived endothelial cell growth Simopoulos C, Gatter KC, Harris AL, Koukourakis MI. factor in primary bladder cancers. Cancer Res. 1996 Oct Hypoxia inducible factor 1alpha and 2alpha 15;56(20):4799-804 overexpression in inflammatory bowel disease. J Clin Pathol. 2003 Mar;56(3):209-13 Takebayashi Y, Yamada K, Miyadera K, Sumizawa T, Furukawa T, Kinoshita F, Aoki D, Okumura H, Yamada Y, Saito S, Tsuno NH, Sunami E, Hori N, Kitayama J, Akiyama S, Aikou T. The activity and expression of Kazama S, Okaji Y, Kawai K, Kanazawa T, Watanabe T, thymidine phosphorylase in human solid tumours. Eur J Shibata Y, Nagawa H. Expression of platelet-derived Cancer. 1996 Jun;32A(7):1227-32 endothelial cell growth factor in inflammatory bowel disease. J Gastroenterol. 2003;38(3):229-37 Creamer D, Jaggar R, Allen M, Bicknell R, Barker J. Overexpression of the angiogenic factor platelet-derived Zhu GH, Schwartz EL. Expression of the angiogenic factor endothelial cell growth factor/thymidine phosphorylase in thymidine phosphorylase in THP-1 monocytes: induction psoriatic epidermis. Br J Dermatol. 1997 Dec;137(6):851-5 by autocrine tumor necrosis factor-alpha and inhibition by aspirin. Mol Pharmacol. 2003 Nov;64(5):1251-8 Waguri Y, Otsuka T, Sugimura I, Matsui N, Asai K, Moriyama A, Kato T. Gliostatin/platelet-derived endothelial de Bruin M, Peters GJ, Oerlemans R, Assaraf YG, cell growth factor as a clinical marker of rheumatoid Masterson AJ, Adema AD, Dijkmans BA, Pinedo HM, arthritis and its regulation in fibroblast-like synoviocytes. Br Jansen G. Sulfasalazine down-regulates the expression of J Rheumatol. 1997 Mar;36(3):315-21 the angiogenic factors platelet-derived endothelial cell growth factor/thymidine phosphorylase and interleukin-8 in van Laar JA, Rustum YM, Ackland SP, van Groeningen human monocytic-macrophage THP1 and U937 cells. Mol CJ, Peters GJ. Comparison of 5-fluoro-2'-deoxyuridine with Pharmacol. 2004 Oct;66(4):1054-60 5-fluorouracil and their role in the treatment of colorectal cancer. Eur J Cancer. 1998 Feb;34(3):296-306 De Clercq E. Antiviral drugs in current clinical use. J Clin Virol. 2004 Jun;30(2):115-33

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1176

TYMP (thymidine phosphorylase) Bijnsdorp IV, Peters GJ

de Bruin M, Temmink OH, Hoekman K, Pinedo H, Peters formulation in the treatment of gastrointestinal GJ.. Role of platelet derived endothelial cell growth factor/ malignancies. Cancer Sci. 2007 Jun;98(6):779-89 thymidine phosphorylase in health and disease. Cancer Therapy. 2006;4:99-124. (REVIEW) Bijnsdorp IV, de Bruin M, Laan AC, Fukushima M, Peters GJ. The role of platelet-derived endothelial cell growth Wang EH, Goh YB, Moon IS, Park CH, Lee KH, Kang SH, factor/thymidine phosphorylase in tumor behavior. Kang CS, Choi YJ. Upregulation of thymidine Nucleosides Nucleotides Nucleic Acids. 2008 phosphorylase in chronic glomerulonephritis and its role in Jun;27(6):681-91 tubulointerstitial injury. Nephron Clin Pract. 2006;102(3- 4):c133-42 Bronckaers A, Gago F, Balzarini J, Liekens S. The dual role of thymidine phosphorylase in cancer development Liekens S, Bronckaers A, Pérez-Pérez MJ, Balzarini J. and chemotherapy. Med Res Rev. 2009 Nov;29(6):903-53 Targeting platelet-derived endothelial cell growth factor/thymidine phosphorylase for cancer therapy. This article should be referenced as such: Biochem Pharmacol. 2007 Dec 3;74(11):1555-67 Bijnsdorp IV, Peters GJ. TYMP (thymidine phosphorylase). Temmink OH, Emura T, de Bruin M, Fukushima M, Peters Atlas Genet Cytogenet Oncol Haematol. 2010; GJ. Therapeutic potential of the dual-targeted TAS-102 14(12):1173-1177.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1177

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Leukaemia Section Mini Review der(6)t(1;6)(q21-23;p21) Adriana Zamecnikova Kuwait Cancer Control Center, Laboratory of Cancer Genetics, Department of Hematology, Shuwaikh, 70653, Kuwait (AZ)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/der6t0106q21p21ID1546.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI der6t0106q21p21ID1546.txt

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

Partial karyotypes showing the chromosomal translocation der(6)t(1;6)(q21-23;p21) identified by G-banding.

years old male, progressed to AML. From the Clinics and pathology known data of 14 patients with myelofibrosis, Disease median age was 65.5 years (range, 38-72 years). Most frequently observed in chronic Clinics myeloproliferative disorders, occurs with higher In the largest study, the anomaly was associated frequency in patients with chronic idiopathic with splenomegaly, elevated WBC count, elevated myelofibrosis, polycythemia vera and post- levels of alkaline phosphatase and lactate polycythemic myelofibrosis; may be present either dehydrogenase; median overall survival was 7.8 at diagnosis or during transformation to advanced years: five patients have died (one transformed to stages of the disease. acute myeloid leukemia and the others died because Epidemiology of sepsis or thrombosis). Described in 20 cases (11 males, 9 females): 1 Cytogenetics biphenotypic leukemia (16 years old male); 1 B-cell lymphoma (73 years old female); 2 acute myeloid Cytogenetics morphological leukemia (AML) patients (1 male 71 years old, 1 Breakpoints may be controversial and difficult to female 28 years old); and in 16 patients with ascertain in poor quality preparations. Recently, the myelofibrosis with myeloid metaplasia (9 males; 7 same breakpoint on 6p21.3 and clustering of females): eleven patients had myelofibrosis with breakpoints near the paracentric region 1q21-23 myeloid metaplasia, three post-polycythemic was described in 14 patients with myelofibrosis myeloid metaplasia, and one post-thrombocythemic with myelocytic metaplasia. myeloid metaplasia; one of these patients, a 47

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1178 der(6)t(1;6)(q21-23;p21) Zamecnikova A

Additional anomalies cytotoxic treatments and the underlying molecular consequences of the rearrangement remain to be Sole anomaly in 9 cases (2 AML and 7 cases with determined. myelofibrosis); no recurrent additional anomaly observed in patients with complex karyotypes. 4 patients had two or more different clones (1 patient To be noted with biphenotypic leukemia and 3 myelofibrosis Case Report cases); among them 2 patients had 1q21-23 der(6)t(1;6)(q21;p21) in myelofibrosis following rearrangements involving the homologous polycythemia vera. chromosome 1. References Result of the chromosomal Mertens F, Johansson B, Heim S, Kristoffersson U, anomaly Mitelman F. Karyotypic patterns in chronic myeloproliferative disorders: report on 74 cases and Fusion protein review of the literature. Leukemia. 1991 Mar;5(3):214-20 Oncogenesis Reilly JT, Snowden JA, Spearing RL, Fitzgerald PM, Jones N, Watmore A, Potter A. Cytogenetic abnormalities and The presence of the der(6)t(1;6) results in partial their prognostic significance in idiopathic myelofibrosis: a trisomy for 1q21-23 to 1qter and in loss of 6p21 to study of 106 cases. Br J Haematol. 1997 Jul;98(1):96-102 6pter. The pathogenetic significance may be the Andrieux J, Demory JL, Caulier MT, Agape P, Wetterwald consequence of gain of gene(s) on 1q and/or haplo- M, Bauters F, Laï JL. Karyotypic abnormalities in insufficiency of gene(s) from 6p and alternatively, myelofibrosis following polycythemia vera. Cancer Genet rearrangements of one or more genes at the Cytogenet. 2003 Jan 15;140(2):118-23 breakpoints. The significance of the 6p21 Dingli D, Grand FH, Mahaffey V, Spurbeck J, Ross FM, breakpoint is unclear; however a number of Watmore AE, Reilly JT, Cross NC, Dewald GW, Tefferi A. published reports of myelofibrosis with Der(6)t(1;6)(q21-23;p21.3): a specific cytogenetic abnormality in myelofibrosis with myeloid metaplasia. Br J chromosome 6p breakpoints in the region raise the Haematol. 2005 Jul;130(2):229-32 possibility of a gene involved in the pathogenesis of this hematologic disorder. The inability to identify Hussein K, Van Dyke DL, Tefferi A. Conventional cytogenetics in myelofibrosis: literature review and common breakpoints on 1q, suggests that an discussion. Eur J Haematol. 2009 May;82(5):329-38 increase in gene copy number is a pathogenetic event. Whether trisomy 1q is a secondary event to a This article should be referenced as such: primary (cryptic? e.g. JAK2 V617F mutation) Zamecnikova A. der(6)t(1;6)(q21-23;p21). Atlas Genet anomaly as well as the roles of methylation, Cytogenet Oncol Haematol. 2010; 14(12):1178-1179.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1179

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Leukaemia Section Short Communication ins(9;4)(q33;q12q25) Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/ins0904q33q12q25ID1450.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI ins0904q33q12q25ID1450.txt

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 and pathology Protein Centrosomal protein; regulates CDK5; binds EB1. Disease The CDK5RAP2-EB1 complex stimulates Chronic eosinophilic leukemia microtubule assembly (Fong et al., 2009); critical for centrosomal localization of dynein throughout Epidemiology the cell cycle (Lee and Rhee, 2010). CDK5RAP2- One case to date, a 71-year-old female patient with knockdown cells have increased resistance to chronic eosinophilic leukemia in accelerated phase paclitaxel and doxorubicin (Zhang et al., 2009). (Walz et al., 2006). Homozygous mutations in CDK5RAP2 can cause Prognosis microcephaly (Bond et al., 2005). Remission was obtained with imatinib, but the Result of the chromosomal patient relapsed with imatinib-resistant acute myeloid leukemia that was characterized by a anomaly normal karyotype, absence of detectable CDK5RAP2-PDGFRA mRNA, and a newly Hybrid gene acquired G12D NRAS mutation. Description In-frame fusion between exon 13 of the Genes involved and CDK5RAP2, a 40 bp insert from an inverted sequence of PDGFRA intron 9, and a truncated proteins PDGFRA exon 12. No reciprocal PDGFRA- PDGFRA CDK5RAP2 transcript. Location Fusion protein 4q25 Description Protein N-term CDK5RAP2 - C-term PDGFRA; 1003 Receptor tyrosine kinase. Gain-of-function amino acids; contains 494 amino acids, including mutations of PDGFRA are implicated in a subset of several potential dimerization domains, of gastrointestinal stromal tumors (Heinrich et al., CDK5RAP2 and 509 amino acids from PDGFRA 2003). PDGFRA has also been involved in tyrosine kinase domains. translocations, making hybrid genes with STRN Oncogenesis (2p22), FIP1L1 (4q12), KIF5B (10p11), ETV6 Constitutive tyrosine kinase activity is likely. (12p13) and BCR (22q11). CDK5RAP2 References Location Heinrich MC, Corless CL, Duensing A, McGreevey L, 9q33 Chen CJ, Joseph N, Singer S, Griffith DJ, Haley A, Town A, Demetri GD, Fletcher CD, Fletcher JA. PDGFRA

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1180 ins(9;4)(q33;q12q25) Huret JL

activating mutations in gastrointestinal stromal tumors. 2008 Nov;22(11):1999-2010 Science. 2003 Jan 31;299(5607):708-10 Fong KW, Hau SY, Kho YS, Jia Y, He L, Qi RZ. Interaction Bond J, Roberts E, Springell K, Lizarraga SB, Scott S, of CDK5RAP2 with EB1 to track growing microtubule tips Higgins J, Hampshire DJ, Morrison EE, Leal GF, Silva EO, and to regulate microtubule dynamics. Mol Biol Cell. 2009 Costa SM, Baralle D, Raponi M, Karbani G, Rashid Y, Jafri Aug;20(16):3660-70 H, Bennett C, Corry P, Walsh CA, Woods CG. A centrosomal mechanism involving CDK5RAP2 and CENPJ Zhang X, Liu D, Lv S, Wang H, Zhong X, Liu B, Wang B, controls brain size. Nat Genet. 2005 Apr;37(4):353-5 Liao J, Li J, Pfeifer GP, Xu X. CDK5RAP2 is required for spindle checkpoint function. Cell Cycle. 2009 Apr Walz C, Curtis C, Schnittger S, Schultheis B, Metzgeroth 15;8(8):1206-16 G, Schoch C, Lengfelder E, Erben P, Müller MC, Haferlach T, Hochhaus A, Hehlmann R, Cross NC, Reiter A. Lee S, Rhee K. CEP215 is involved in the dynein- Transient response to imatinib in a chronic eosinophilic dependent accumulation of pericentriolar matrix proteins leukemia associated with ins(9;4)(q33;q12q25) and a for spindle pole formation. Cell Cycle. 2010 Feb CDK5RAP2-PDGFRA fusion gene. Genes Chromosomes 15;9(4):774-83 Cancer. 2006 Oct;45(10):950-6 This article should be referenced as such: Gotlib J, Cools J. Five years since the discovery of FIP1L1-PDGFRA: what we have learned about the fusion Huret JL. ins(9;4)(q33;q12q25). Atlas Genet Cytogenet and other molecularly defined eosinophilias. Leukemia. Oncol Haematol. 2010; 14(12):1180-1181.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1181

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Solid Tumour Section Short Communication t(19;22)(q13;q12) in myoepithelial carcinoma Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Tumors/t1922q13q12MyoCarcID6287.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI t1922q13q12MyoCarcID6287.txt

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 and pathology Genes involved and Disease proteins Myoepithelioma tumours of soft tissue cover a wide ZNF444 range of tumours of various behaviour. While most are of intermediate aggressivity, some metastasize. Location There is no sex ratio predominance. Mean age at 19q13 diagnosis is 38 years; with a range of 3-83 years. Of Protein a hundred of cases reviewed by Hornick and Possess a SCAN domain and 4 C2H2-type zinc Fletcher (2003), 60% were benign and classified as fingers. Transcription factor. myoepitheliomas or mixed tumors, and 40% were EWSR1 classified as myoepithelial carcinomas or malignant mixed tumours. Location Amongst cases with benign or low-grade cytology, 22q12 with a mean follow-up of 3 years, 20% recurred Protein locally and none metastasized. Amongst From N-term to C-term: a transactivation domain cytologically malignant cases, with a mean follow- (TAD) containing multiple degenerate hexapeptide up of 4 years, 40% recurred locally, 1/3 repeats, 3 arginine/glycine rich domains (RGG metastasized, and 4 out of 31 patients died. regions), a RNA recognition motif, and a RanBP2 Tumours are positive for epithelial markers, and for type Zinc finger. Role in transcriptional regulation S100 or GFAP, or myogenic markers (Gleason and for specific genes and in mRNA splicing. Fletcher, 2007). Epidemiology Result of the chromosomal One case to date, a 40-year-old female patient. anomaly After surgical removal, recurrences occured during 2 years, and metastases appeared 3 years later. The Hybrid Gene patient finally died 9.5 years after initial diagnosis Description (Brandal et al., 2009). 5' EWSR1 - 3' ZNF444; fuses EWSR1 exon 8 to the very near end of ZNF444 (at nucleotide 967, Cytogenetics while the full transcript of ZNF444 is 984 nt long!). Cytogenetics Morphological Fusion Protein The t(19;22)(q13;q12) was found within a complex Description karyotype. Truncated EWSR1 with 6 amino acids added from

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1182 t(19;22)(q13;q12) in myoepithelial carcinoma Huret JL

ZNF444. This does not fit with the usual model of series of 29 cases. Am J Surg Pathol. 2007 carcinogenesis found with other EWSR1 Dec;31(12):1813-24 translocations, were there is fusion of the N Brandal P, Panagopoulos I, Bjerkehagen B, Heim S. terminal transactivation domain of EWSR1 to the t(19;22)(q13;q12) Translocation leading to the novel fusion gene EWSR1-ZNF444 in soft tissue myoepithelial DNA binding domain of the partner (e.g. FLI1). carcinoma. Genes Chromosomes Cancer. 2009 References Dec;48(12):1051-6 This article should be referenced as such: Hornick JL, Fletcher CD. Myoepithelial tumors of soft tissue: a clinicopathologic and immunohistochemical study Huret JL. t(19;22)(q13;q12) in myoepithelial carcinoma. of 101 cases with evaluation of prognostic parameters. Am Atlas Genet Cytogenet Oncol Haematol. 2010; J Surg Pathol. 2003 Sep;27(9):1183-96 14(12):1182-1183. Gleason BC, Fletcher CD. Myoepithelial carcinoma of soft tissue in children: an aggressive neoplasm analyzed in a

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1183

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Deep Insight Section

Glutathione S-Transferase pi (GSTP1) Isabelle Meiers Maidstone and Tunbridge Wells NHS Trust, Preston Hall Hospital, Royal British Legion Village, Aylesford, Kent, ME20 7NJ, UK (IM)

Published in Atlas Database: February 2010 Online updated version : http://AtlasGeneticsOncology.org/Deep/GSTP1inCancerID20084.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI GSTP1inCancerID20084.txt

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

Running title: GSTP1 review suppressor genes. The cytosolic isoenzyme Key words: Glutathione S-transferase pi (GSTP1), glutathione S-transferase pi (GSTP1) is an cancer, methylation analysis, antioxidants. important multifunctional detoxifying enzyme within the glutathione S-transferase family enzymes Pi-class glutathione-S-transferase (GSTP1) located that inactivates electrophilic carcinogens by on chromosome 11q13 encodes a phase II conjugation with glutathione (Toffoli et al., 1992; metabolic enzyme that detoxifies reactive Jerónimo et al., 2001). The regulatory sequence electrophilic intermediates. GSTP1 plays an near the GST gene is commonly affected by important role in protecting cells from cytotoxic hypermethylation during the early stages of and carcinogenic agents and is expressed in normal carcinogenesis (Lee et al., 1994; Brooks et al., tissues at variable levels in different cell types. 1998; Cairns et al., 2001; Jerónimo et al., 2002; Altered GSTP1 activity and expression have been Henrique and Jerónimo, 2004). reported in many tumors and this is largely due to Several classes of GST, including alpha, mu, pi, GSTP1 DNA hypermethylation at the CpG island and theta, were previously found in human tissue. in the promoter-5'. For example, compared with benign tissue, there is increased expression of GST pi in cancers of the We review the potential novel role of glutathione S- breast, colon, stomach, pancreas, bladder, lung, transferase pi (GSTP1) and its related expression in head and neck, ovary, and cervix, as well as soft miscellaneous cancers. We focus on the rationale tissue sarcoma, testicular embryonal carcinoma, for use of molecular assays for the detection of meningioma, and glioma (Niitsu et al., 1989; cancer, emphasizing the role of the identification of Randall et al., 1990; Kantor et al., 1991; Satta et al., epigenetic alterations. Finally, we focus on the 1992; Toffoli et al., 1992; Green et al., 1993; Inoue potential role of GSTP1 in the pathway of prostate et al., 1995; Bentz et al., 2000; Tratche et al., 2002; cancer, the most GSTP1 DNA hypermethylation- Simic et al., 2005; Arai et al., 2006). related neoplasm studied to date. However, hypermethylation of the GSTP1 promoter Advances in the epigenetic characterization of has been associated with gene silencing in prostate cancers enabled the development of DNA cancer and kidney cancer (Lee et al., 1994; Brooks methylation assays that may soon be used in et al., 1998; Cairns et al., 2001; Jerónimo et al., diagnostic testing of serum and tissue for cancers. 2002; Dulaimi et al., 2004). Similarly, expression Inhibition of aberrant promoter methylation could of GSTP1 is lower in invasive pituitary tumors than theoretically prevent carcinogenesis. in noninvasive pituitary tumors and methylation Reactive oxygen species that are generated by status correlates with significant downregulation of physiologic processes such as cellular respiration, GSTP1 expression; the frequency of GSTP1 exposure to chemical agents, or exposure to methylation being higher in invasive pituitary ionizing radiation may overcome cellular tumors with reduced-GSTP1 expression than in antioxidant defense and cause DNA damage pituitary adenomas with normal or high GSTP1 (Bostwick et al., 2000). Such damage may result in expression. These data indicate that GSTP1 mutations and alteration of oncogenes or tumor inactivation through CpG hypermethylation is

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1184

Glutathione S-Transferase pi (GSTP1) Meiers I

common in pituitary adenomas and may contribute Due to its relative simplicity, safety, and sensitivity, to aggressive pituitary tumor behavior (Yuan et al., methylation-specific PCR is the most commonly 2008). More recently, a study showed a trend of employed method for methylation analysis increasing GSTP1 methylation frequency with (Herman et al., 1996). increasing grade of mammary phyllodes tumors. The conventional methylation-specific PCR The authors reported that GSTP1 promoter (CMSP) assay uses two sets of primers specifically hypermethylation was associated with loss of designed to amplify the methylated or unmethylated GSTP1 expression. These results suggest that sequence, and the PCR products are run in a gel phyllodes tumors segregate into only two groups on (Herman et al., 1996). The results of CMSP at a the basis of their methylation profiles: the benign particular DNA region are simply reported as group and the combined borderline/malignant methylated or unmethylated, not allowing group (Kim et al., 2009). Other investigators quantitation or identification of partial methylation. studied the role of hypermethylation of the GSTP1 The CMSP assay is mainly used for GSTP1 gene promoter region in endometrial carcinoma and methylation detection in fluids. For instance, found that reduced GSTP1 expression was GSTP1 methylation in serum of men with localized associated with myometrial invasion potential prostate cancer prior to treatment carries a 4.4 fold (Chan et al., 2005). increased risk of biochemical recurrence following Epigenetic alterations: emerging surgery (Bastian et al., 2005). The use of fluorescence-based real-time molecular markers for cancer quantitative methylation-specific PCR (QMSP) detection assay improved the sensitivity of tumor detection. Cancer is a process fuelled both by genetic Continuous monitoring of fluorescent signals alterations and epigenetic mechanisms. Epigenetics during the PCR process enabled quantification of refer to changes in gene expression that can be methylated alleles of a single region amongst mitotically inherited, but are not associated with the unmethylated DNA because the fluorescence changes in the coding sequence of the affected emission of the reporter represents the number of genes. In other words, epigenetics refer to the generated DNA fragments (Heid et al., 1996). inheritance of information based on gene GSTP1 hypermethylation: expression levels, in contrast to genetics that refer significance and incidence related to to transmission of information based on gene sequence (Esteller et al., 2000). DNA methylation, prostate cancer the best understood mechanism in epigenetics, is an Epigenetic silencing of gluthathione-S-transferase enzyme-mediated chemical modification that adds pi (GSTP1) is recognized as being a molecular methyl (-CH3) groups at selected sites on DNA. In hallmark of human prostate cancer. Methylation of humans and most mammals, DNA methylation only CpG islands in the promoter of the pi class of affects the cytosine base (C), when it is followed by glutathione S-transferase occurs in prostatic a guanosine (G). Methylation of the cytosine intraepithelial neoplasia (PIN) and cancer nucleotide residue located within the dinucleotide (Gonzalgo et al., 2004). Other hypermethylated 5'-CpG-3' is the most frequent epigenetic alteration regions relevant to prostate cancer include the in humans. These CpG dinucleotides are not retinoic acid receptor beta 2 (Bastian et al., 2007). randomly distributed in the genome. Indeed, there These findings in prostate cancer suggest that DNA are CpG-rich regions called "CpG islands" methylation is among the early events in frequently associated with the 5' regulatory regions tumorigenesis, but it remains to be seen whether of genes, including the promoter. DNA methylation DNA methylation is a necessary or permissive in the promoter regions is a powerful mechanism event in tumorigenesis. for the suppression of gene activity. The extensive methylation of deoxycytidine DNA methylation analysis: currently nucleotides distributed throughout the 5' "CG island" region of GSTP1 is not detected in benign available methods prostatic epithelium, but has been detected in Methylation of CpG islands is of interest for intraepithelial neoplasia, prostatic adenocarcinoma, diagnostic and prognostic reasons. Methylation of and fluids (plasma, serum, ejaculate, and urine) of one or both alleles of a region can serve as a patients with prostate cancer by methylation- biomarker of cancer or silence gene expression specific polymerase chain reaction assay, and may when they are in a promoter region (Verma and be useful as a cancer-specific molecular biomarker Srivastava, 2002). Assays for methylation are (Lee et al., 1994; Cairns et al., 2001; Henrique and appealing for translational research since they can Jerónimo, 2004; Crocitto et al., 2004; Perry et al., utilize amplification techniques, such as 2006; Hopkins et al., 2007; Cao and Yao, 2010). methylation-specific polymerase chain reaction Quantitative methylation-specific PCR (QMSP) (PCR), and thereby utilize small amounts of reveals that the epigenetic silencing (loss of samples. expression) of the GSTP1 gene is in fact the most

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1185

Glutathione S-Transferase pi (GSTP1) Meiers I

common genetic alteration in prostate cancer hypermethylation detection (Kollermann et al., (>90%) and high-grade prostatic intraepithelial 2006) and the authors suggested that the change neoplasia (PIN) (70%) (Lee et al., 1994; Brooks et from positive to negative GSTP1 hypermethylation al., 1998; Cairns et al., 2001; Harden et al., 2003; status in two patients may point to partial androgen Henrique and Jerónimo, 2004) and this somatic dependency (Kollermann et al., 2006). In addition inactivation ("silencing") of GSTP1 is directly to the supposed hormonal interaction, other associated with promoter methylation (Cairns et al., possible explanations may be speculated to explain 2001; Jerónimo et al., 2002; Henrique and why prostate cancer loses GSTP1 hypermethylation Jerónimo, 2004). Higher levels of GSTP1 promoter after prolonged neoadjuvant hormonal therapy. methylation is associated with the transition from First, the lack of GSTP1 hypermethylation may be prostatic intraepithelial neoplasia (PIN) to attributable to technical problems (false negative carcinoma (Henrique et al., 2006). results). Furthermore, the possibility that both During cancer development, GSTP1 does not tumors primarily lacked GSTP1 hypermethylation appear to function either as an oncogene or as a might be raised. However, further studies are tumor suppressor gene, since induced GSTP1 necessary to assess the frequency and extent of expression in prostate cancer cell lines failed to hormonal interaction with GSTP1 suppress cell growth. Instead, GSTP1 was proposed hypermethylation. to act as a "caretaker" gene. When GSTP1 is Anti-cancer effect of GSTP1 and inactivated, prostate cells appear to become more vulnerable to somatic alterations upon chronic future prospects exposure to genome-damaging stresses as oxidants Increased levels of GST pi may protect human and electrophiles, that are contributed by cancer cells against cytotoxic drugs. Several environment and lifestyle (Kinzler and Vogelstein, antineoplastic drugs, particularly reactive 1997; Cairns et al., 2001). electrophilic alkylating agents, form conjugates The significance of absent GSTP1 (GSTP1 with glutathione spontaneously and in GST- silencing) in high grade PIN and carcinoma is catalyzed reactions (Awasthi et al., 1996). The unclear. It may be an epiphenomenon, simply expression of particular subclasses of GST protects reflecting disruption of the basal cell layer with cells from the cytotoxicities of these cancer drugs, neoplastic progression. However, Lee et al. and overexpression of GST has been implicated in considered the likelihood of a more fundamental antineoplastic drug resistance (Morrow et al., role (Lee et al., 1994). Two studies found that a 1998). Induction of the enzymes is thought to small proportion (3.5-5%) of cases retained modest represent an adaptive response to stress, and may be GSTP1 expression in carcinoma (Cookson et al., triggered by exogenous chemical agents and 1997; Moskaluk et al., 1997). Cookson et al. also probably also by reactive oxygen metabolites recorded positivity in 1 of 17 cases of high grade (Hayes and Pulford, 1995). GST enzymes have a PIN (Cookson et al., 1997) . Unlike genetic broad substrate specificity that includes substances alterations that permanently and definitively change with known mutagenic properties. Elevated serum DNA sequence, promoter methylation is a GST pi has been exploited as a serum tumor marker potentially reversible modification. Hence, for gastrointestinal cancer (Niitsu et al., 1989) and promoter methylation may be amenable to non-Hodgkin's lymphoma (Katahira et al., 2004) as therapeutic intervention aimed at reactivating a method of predicting sensitivity to chemotherapy. silenced cancer genes. This has important Inactivation of GSTP1 in prostate cancer occurs implications for chemoprevention because, as early during carcinogenesis, leaving prostate cells mentioned above, up to 70% of cases of high-grade with inadequate defenses against oxidant and PIN display GSTP1 promoter methylation (Brooks electrophile carcinogens. Epigenetic mechanisms et al., 1998; Cairns et al., 2001; Jerónimo et al., (see above) are strongly implicated in progression 2002). Indeed, recently some authors have (Rennie and Nelson, 1998). Unlike genetic investigated the effects of green tea polyphenols alterations, changes in DNA methylation are (GTPs) on GSTP1 re-expression. They potentially reversible. Thus, therapeutic demonstrated that promoter demethylation by green interventions involving reversal of the methylation tea polyphenols leads to re-expression of GSTP1 in process of several key genes in prostate human prostate cancer cells, therefore making green carcinogenesis might improve current therapeutic tea polyphenols excellent candidates for the options, thereby enhancing the anti-cancer effect of chemoprevention of prostate cancer (Pandey et al., GSTP1 gene in patients "at risk" with high-grade 2009). PIN or in men with established prostate cancer. Some investigators evaluated the impact of Nucleoside-analogue inhibitors of DNA androgen deprivation therapy on the detection of methyltransferases, such as 5-aza-2'-deoxycytidine, GSTP1 hypermethylation in prostate cancer are able to demethylate DNA and restore silenced (Kollermann et al., 2006). In 87% (13/15) of the gene expression. Unfortunately, the clinical utility patients, there was no alteration in GSTP1 of these compounds has not yet been fully realized,

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1186

Glutathione S-Transferase pi (GSTP1) Meiers I

mainly because of their side effects. The anti- SJ, Jones RA, Horne CH. Glutathione S-transferase arrhythmia drug procainamide, a nonnucleoside (placental) as a marker of transformation in the human cervix uteri: an immunohistochemical study. Br J Cancer. inhibitor of DNA methyltransferases (category of 1990 Oct;62(4):614-8 enzymes that catalyse DNA methylation during cell Kantor RR, Giardina SL, Bartolazzi A, Townsend AJ, replication), reversed GSTP1 DNA Myers CE, Cowan KH, Longo DL, Natali PG. Monoclonal hypermethylation and restorted GSTP1 expression antibodies to glutathione S-transferase pi- in LNCaP human prostate cancer cells propagated immunohistochemical analysis of human tissues and in vitro or in vivo as xenograft tumors in athymic cancers. Int J Cancer. 1991 Jan 21;47(2):193-201 nude mice (Cairns et al., 2001). Some investigators Satta T, Isobe K, Yamauchi M, Nakashima I, Takagi H. tested the potential use of procaine, an anesthetic Expression of MDR1 and glutathione S transferase-pi drug related to procainamide. Using the MCF-7 genes and chemosensitivities in human gastrointestinal cancer. Cancer. 1992 Feb 15;69(4):941-6 breast cancer cell line, they have found that procaine produced a 40% reduction in 5- Toffoli G, Frustaci S, Tumiotto L, Talamini R, Gherlinzoni methylcytosine DNA content as determined by F, Picci P, Boiocchi M. Expression of MDR1 and GST-pi in human soft tissue sarcomas: relation to drug resistance high-performance capillary electrophoresis and and biological aggressiveness. Ann Oncol. 1992 total DNA enzyme . Procaine can also Jan;3(1):63-9 demethylate densely hypermethylated CpG islands Green JA, Robertson LJ, Clark AH. Glutathione S- such as those located in the promoter region of the transferase expression in benign and malignant ovarian RAR beta 2 gene, restoring gene expression of tumours. Br J Cancer. 1993 Aug;68(2):235-9 epigenetically silenced genes. This property may be Lee WH, Morton RA, Epstein JI, Brooks JD, Campbell PA, explained by binding of procaine to CpG-enriched Bova GS, Hsieh WS, Isaacs WB, Nelson WG. Cytidine DNA. Finally, procaine also has growth-inhibitory methylation of regulatory sequences near the pi-class effects in these cancer cells, causing mitotic arrest glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A. 1994 (Villar-Garea et al., 2003). Thus, procaine and Nov 22;91(24):11733-7 procainamide are promising candidate agents for Hayes JD, Pulford DJ. The glutathione S-transferase future cancer therapies based on epigenetics. Li et supergene family: regulation of GST and the contribution al. reported that GSTP1 was upregulated in the of the isoenzymes to cancer chemoprotection and drug stromal compartment of hormone-independent resistance. Crit Rev Biochem Mol Biol. 1995;30(6):445-600 prostate cancer, which may contribute to Inoue T, Ishida T, Sugio K, Maehara Y, Sugimachi K. chemoresistance of advanced prostate cancer Glutathione S transferase Pi is a powerful indicator in (Morrow et al., 1998). chemotherapy of human lung squamous-cell carcinoma. Epidemiologic evidence has shown a reduced risk Respiration. 1995;62(4):223-7 of prostate cancer in men consuming selenium, Awasthi S, Bajpai KK, Piper JT, Singhal SS, Ballatore A, suggesting a role for antioxidants in protection Seifert WE Jr, Awasthi YC, Ansari GA. Interactions of against prostate carcinogenesis. A systematic melphalan with glutathione and the role of glutathione S- transferase. Drug Metab Dispos. 1996 Mar;24(3):371-4 review and meta-analysis of the literature confirm that selenium intake may reduce the risk of prostate Heid CA, Stevens J, Livak KJ, Williams PM. Real time cancer (Etminan et al., 2005). Vitamin E intake also quantitative PCR. Genome Res. 1996 Oct;6(10):986-94 may decrease DNA damage and inhibit Herman JG, Graff JR, Myöhänen S, Nelkin BD, Baylin SB. transformation through its antioxidant function. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S Long-term supplementation with alpha-tocopherol A. 1996 Sep 3;93(18):9821-6 substantially reduced prostate cancer incidence and mortality in male smokers (Heinonen et al., 1998). Cookson MS, Reuter VE, Linkov I, Fair WR. Glutathione S- transferase PI (GST-pi) class expression by Therapy directed at the induction or preservation of immunohistochemistry in benign and malignant prostate GSTP1 activity in benign prostatic epithelium may tissue. J Urol. 1997 Feb;157(2):673-6 prevent or delay progression of prostatic cancer. Kinzler KW, Vogelstein B. Cancer-susceptibility genes. GSTP1 has a protective role as an antioxidant agent Gatekeepers and caretakers. Nature. 1997 Apr in transformation and progression of prostate 24;386(6627):761, 763 cancer. The interplay between altered or impaired Moskaluk CA, Duray PH, Cowan KH, Linehan M, Merino expression of GST appears to play a significant role MJ. Immunohistochemical expression of pi-class in carcinogenesis in the prostate. Inhibition of glutathione S-transferase is down-regulated in aberrant promoter methylation could be an effective adenocarcinoma of the prostate. Cancer. 1997 Apr 15;79(8):1595-9 method of chemoprevention. Brooks JD, Weinstein M, Lin X, Sun Y, Pin SS, Bova GS, Epstein JI, Isaacs WB, Nelson WG. CG island methylation References changes near the GSTP1 gene in prostatic intraepithelial Niitsu Y, Takahashi Y, Saito T, Hirata Y, Arisato N, neoplasia. Cancer Epidemiol Biomarkers Prev. 1998 Maruyama H, Kohgo Y, Listowsky I. Serum glutathione-S- Jun;7(6):531-6 transferase-pi as a tumor marker for gastrointestinal Heinonen OP, Albanes D, Virtamo J, Taylor PR, Huttunen malignancies. Cancer. 1989 Jan 15;63(2):317-23 JK, Hartman AM, Haapakoski J, Malila N, Rautalahti M, Randall BJ, Angus B, Akiba R, Hall A, Cattan AR, Proctor Ripatti S, Mäenpää H, Teerenhovi L, Koss L, Virolainen M, Edwards BK. Prostate cancer and supplementation with

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1187

Glutathione S-Transferase pi (GSTP1) Meiers I

alpha-tocopherol and beta-carotene: incidence and secretions using combinatorial MSP analysis. Urology. mortality in a controlled trial. J Natl Cancer Inst. 1998 Mar 2004 Feb;63(2):414-8 18;90(6):440-6 Henrique R, Jerónimo C. Molecular detection of prostate Morrow CS, Smitherman PK, Diah SK, Schneider E, cancer: a role for GSTP1 hypermethylation. Eur Urol. 2004 Townsend AJ. Coordinated action of glutathione S- Nov;46(5):660-9; discussion 669 transferases (GSTs) and multidrug resistance protein 1 (MRP1) in antineoplastic drug detoxification. Mechanism of Katahira T, Takayama T, Miyanishi K, Hayashi T, Ikeda T, GST A1-1- and MRP1-associated resistance to Takahashi Y, Takimoto R, Matsunaga T, Kato J, Niitsu Y. chlorambucil in MCF7 breast carcinoma cells. J Biol Chem. Plasma glutathione S-Transferase P1-1 as a prognostic 1998 Aug 7;273(32):20114-20 factor in patients with advanced non-Hodgkin's lymphoma (stages III and IV). Clin Cancer Res. 2004 Dec Rennie PS, Nelson CC. Epigenetic mechanisms for 1;10(23):7934-40 progression of prostate cancer. Cancer Metastasis Rev. 1998-1999;17(4):401-9 Bastian PJ, Palapattu GS, Lin X, Yegnasubramanian S, Mangold LA, Trock B, Eisenberger MA, Partin AW, Nelson Bentz BG, Haines GK 3rd, Radosevich JA. Glutathione S- WG. Preoperative serum DNA GSTP1 CpG island transferase pi in squamous cell carcinoma of the head and hypermethylation and the risk of early prostate-specific neck. Laryngoscope. 2000 Oct;110(10 Pt 1):1642-7 antigen recurrence following radical prostatectomy. Clin Cancer Res. 2005 Jun 1;11(11):4037-43 Bostwick DG, Alexander EE, Singh R, Shan A, Qian J, Santella RM, Oberley LW, Yan T, Zhong W, Jiang X, Chan QK, Khoo US, Chan KY, Ngan HY, Li SS, Chiu PM, Oberley TD. Antioxidant enzyme expression and reactive Man LS, Ip PP, Xue WC, Cheung AN. Promoter oxygen species damage in prostatic intraepithelial methylation and differential expression of pi-class neoplasia and cancer. Cancer. 2000 Jul 1;89(1):123-34 glutathione S-transferase in endometrial carcinoma. J Mol Diagn. 2005 Feb;7(1):8-16 Esteller M. Epigenetic lesions causing genetic lesions in human cancer: promoter hypermethylation of DNA repair Etminan M, FitzGerald JM, Gleave M, Chambers K. Intake genes. Eur J Cancer. 2000 Dec;36(18):2294-300 of selenium in the prevention of prostate cancer: a systematic review and meta-analysis. Cancer Causes Cairns P, Esteller M, Herman JG, Schoenberg M, Control. 2005 Nov;16(9):1125-31 Jeronimo C, Sanchez-Cespedes M, Chow NH, Grasso M, Wu L, Westra WB, Sidransky D. Molecular detection of Simic T, Mimic-Oka J, Savic-Radojevic A, Opacic M, prostate cancer in urine by GSTP1 hypermethylation. Clin Pljesa M, Dragicevic D, Djokic M, Radosavljevic R. Cancer Res. 2001 Sep;7(9):2727-30 Glutathione S-transferase T1-1 activity upregulated in transitional cell carcinoma of urinary bladder. Urology. Jerónimo C, Usadel H, Henrique R, Oliveira J, Lopes C, 2005 May;65(5):1035-40 Nelson WG, Sidransky D. Quantitation of GSTP1 methylation in non-neoplastic prostatic tissue and organ- Arai T, Miyoshi Y, Kim SJ, Taguchi T, Tamaki Y, Noguchi confined prostate adenocarcinoma. J Natl Cancer Inst. S. Association of GSTP1 CpG islands hypermethylation 2001 Nov 21;93(22):1747-52 with poor prognosis in human breast cancers. Breast Cancer Res Treat. 2006 Nov;100(2):169-76 Jerónimo C, Usadel H, Henrique R, Silva C, Oliveira J, Lopes C, Sidransky D. Quantitative GSTP1 Henrique R, Jerónimo C, Teixeira MR, Hoque MO, hypermethylation in bodily fluids of patients with prostate Carvalho AL, Pais I, Ribeiro FR, Oliveira J, Lopes C, cancer. Urology. 2002 Dec;60(6):1131-5 Sidransky D. Epigenetic heterogeneity of high-grade prostatic intraepithelial neoplasia: clues for clonal Trachte AL, Suthers SE, Lerner MR, Hanas JS, Jupe ER, progression in prostate carcinogenesis. Mol Cancer Res. Sienko AE, Adesina AM, Lightfoot SA, Brackett DJ, Postier 2006 Jan;4(1):1-8 RG. Increased expression of alpha-1-antitrypsin, glutathione S-transferase pi and vascular endothelial Kollermann J, Kempkensteffen C, Helpap B, Schrader M, growth factor in human pancreatic adenocarcinoma. Am J Krause H, Muller M, Miller K, Schostak M. Impact of Surg. 2002 Dec;184(6):642-7; discussion 647-8 hormonal therapy on the detection of promoter hypermethylation of the detoxifying glutathione-S- Verma M, Srivastava S. Epigenetics in cancer: implications transferase P1 gene (GSTP1) in prostate cancer. BMC for early detection and prevention. Lancet Oncol. 2002 Urol. 2006 Jun 27;6:15 Dec;3(12):755-63 Perry AS, Foley R, Woodson K, Lawler M. The emerging Harden SV, Guo Z, Epstein JI, Sidransky D. Quantitative roles of DNA methylation in the clinical management of GSTP1 methylation clearly distinguishes benign prostatic prostate cancer. Endocr Relat Cancer. 2006 tissue and limited prostate adenocarcinoma. J Urol. 2003 Jun;13(2):357-77 Mar;169(3):1138-42 Bastian PJ, Ellinger J, Heukamp LC, Kahl P, Müller SC, Villar-Garea A, Fraga MF, Espada J, Esteller M. Procaine von Rücker A. Prognostic value of CpG island is a DNA-demethylating agent with growth-inhibitory hypermethylation at PTGS2, RAR-beta, EDNRB, and other effects in human cancer cells. Cancer Res. 2003 Aug gene loci in patients undergoing radical prostatectomy. Eur 15;63(16):4984-9 Urol. 2007 Mar;51(3):665-74; discussion 674 Crocitto LE, Korns D, Kretzner L, Shevchuk T, Blair SL, Hopkins TG, Burns PA, Routledge MN. DNA methylation Wilson TG, Ramin SA, Kawachi MH, Smith SS. Prostate of GSTP1 as biomarker in diagnosis of prostate cancer. cancer molecular markers GSTP1 and hTERT in Urology. 2007 Jan;69(1):11-6 expressed prostatic secretions as predictors of biopsy results. Urology. 2004 Oct;64(4):821-5 Yuan Y, Qian ZR, Sano T, Asa SL, Yamada S, Kagawa N, Kudo E. Reduction of GSTP1 expression by DNA Dulaimi E, Ibanez de Caceres I, Uzzo RG, Al-Saleem T, methylation correlates with clinicopathological features in Greenberg RE, Polascik TJ, Babb JS, Grizzle WE, Cairns pituitary adenomas. Mod Pathol. 2008 Jul;21(7):856-65 P. Promoter hypermethylation profile of kidney cancer. Clin Cancer Res. 2004 Jun 15;10(12 Pt 1):3972-9 Kim JH, Choi YD, Lee JS, Lee JH, Nam JH, Choi C, Park MH, Yoon JH. Borderline and malignant phyllodes tumors Gonzalgo ML, Nakayama M, Lee SM, De Marzo AM, display similar promoter methylation profiles. Virchows Nelson WG. Detection of GSTP1 methylation in prostatic

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1188

Glutathione S-Transferase pi (GSTP1) Meiers I

Arch. 2009 Dec;455(6):469-75 This article should be referenced as such: Cao DL, Yao XD. Advances in biomarkers for the early Meiers I. Glutathione S-Transferase pi (GSTP1). Atlas diagnosis of prostate cancer. Chin J Cancer. 2010 Genet Cytogenet Oncol Haematol. 2010; 14(12):1184- Feb;29(2):229-33 1189. Pandey M, Shukla S, Gupta S. Promoter demethylation and chromatin remodeling by green tea polyphenols leads to re-expression of GSTP1 in human prostate cancer cells. Int J Cancer. 2010 Jun 1;126(11):2520-33

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1189

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

Deep Insight Section

The roles of SRA1 gene in breast cancer Yi Yan, Charlton Cooper, Etienne Leygue Manitoba Institute of Cell Biology, University of Manitoba, 770 Bannatyne Avenue, R3E0W3, Winnipeg, Manitoba, Canada (YY, EL); Department of Biochemistry and Medical Genetics, University of Manitoba, 770 Bannatyne Avenue, R3E0W3, Winnipeg, Manitoba, Canada (YY, CC, EL)

Published in Atlas Database: March 2010 Online updated version : http://AtlasGeneticsOncology.org/Deep/SRA1inCancerID20085.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SRA1inCancerID20085.txt

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

Abstract The Steroid receptor RNA activator (SRA) gene has been implicated in estrogen receptor signaling pathway. First identified as a RNA coregulator, SRA had been shown to increase steroid receptor activity. SRA RNA expression is altered during breast tumorigenesis and its molecular role in underscoring these events has been suggested. The subsequent identification of molecules capable of binding SRA, including RNA helicase p68, SRA stem-loop interacting RNA binding protein (SLIRP), and steroidogenic factor 1 (SF1) indicates SRA function is not exclusively limited to modulate steroid receptor activity. A recent genome-wide expression analysis by depleting SRA in cancer cells has further expanded our understanding of a broader biological role played by SRA. In addition, several RNA isoforms have been found to encode an endogenous protein (SRAP), which is well conserved among Chordata. Interestingly, SRAP also modulates steroid receptor activity and functions as a co-regulator in estrogen receptor signaling. The recent observation that a higher expression of SRAP protein is associated with poorer survival in breast cancer patients treated with tamoxifen, highlights the potential relevance of this protein in cancer. Together, the SRA1 gene encodes both functional RNA and protein (SRAP) products, making it a unique member amongst the growing population of steroid receptor co-regulators.

1. Introduction Since the characterization of the first co-regulator, It is now quite apparent that the end results of the steroid receptor co-activator 1 (SRC-1), this list Estrogen Receptor (ER) mediated signaling is not of factors has grown significantly to now include simply limited to ER status and/or the presence of over 300 co-regulators (Lonard and O'Malley, its naturally occurring ligand estridiol. In addition 2007). One particularly interesting member within to the two known estrogen receptors, ERα and ERβ, this family was identified by Lanz et al. in 1999, as ERs-mediated gene transcription also requires it was found not to act as a protein molecule but as transcription co-regulators, which form complexes a functional RNA. This nuclear co-regulator was with estrogen receptors through protein-protein therefore named Steroid receptor RNA activator interactions followed by dynamic recruitment to (SRA) (Lanz et al., 1999). specific gene promoters. Based on the outcomes of 2. Steroid Receptor RNA Activator their regulations, co-regulators are categorized as either co-activators or co-repressors if they either (SRA) promote or prevent gene transcription respectively. 2.1 Discovery of SRA, an RNA Co-activator These complexes regulate the assembly and activity In order to identify new potential co-regulators of the transcription initiation complex through interacting with AF-1 domain of the progesterone chromatin remodeling (McKenna et al., 1999; receptor (PR), Lanz screened a human B- Jenuwein and Allis, 2001). lymphocyte library using AF-1 domain as bait in a

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1190

The roles of SRA1 gene in breast cancer Yan Y, et al.

yeast-two-hybrid assay (Lanz et al., 1999). They 2.3 Effect of SRA RNA on ERα and ERβ identified a new clone, they called SRA, for steroid signaling receptor RNA activator. This cDNA was unable to Several research groups have now confirmed that encode a protein, but was required for the growth of SRA is able to increase induced gene the yeast colony. Further experiment confirmed that transcription by both full length ERs subtypes the potential co-activation role of SRA on PR was (Watanabe et al., 2001; Deblois and Giguère, 2003; mediated through a RNA transcript rather than any Coleman et al., 2004; Klinge et al., 2004). SRA protein product. RNA has been shown to co-activate the action of 2.2 Core sequence of SRA and predicted the AF-2 domain of both ERα and ERβ in a ligand- functional region dependent manner on some, but not all estrogen In the incipient SRA study, a core sequence receptor element (ERE) as measure through spanning SRA exon 2 to exon 5 was found to be luciferase reporters assay (Deblois and Giguère, necessary and sufficient for co-activation function 2003; Coleman et al., 2004). of SRA RNA (Figure 1, Lanz et al., 1999). Several Interestingly, SRA can also enhance AF-1 domain predicted secondary RNA structural motifs are of ERα but not ERβ in a ligand-independent distributed throughout this core sequence, and are manner (Coleman et al., 2004; Deblois and believed to form the functional structures that Giguère, 2003). Overall, data suggest that the action impart SRA activities. Site-directed mutagenesis of the two estrogen receptors are differentially experiment revealed six secondary structural motifs regulated by SRA and SRA regulation of a given (STR1, 7, 9, 10, 11, 12) that independently receptor is also specific of a given ERE sequence participate in PR co-activation by SRA (Lanz et al., (Leygue, 2007). 2002). It was found that silent mutations in both 2.4 Emerging mechanisms of SRA RNA action SRT1 and STR7 of SRA could decrease by more Several studies have been published discussing the than 80% co-activation SRA's function (Lanz et al., mechanism of SRA RNA action (Leygue, 2007). 1999).

Figure 1. SRA1 genomic structure and core sequence. A) SRA sequences were originally described, differing in their 5' and 3' extremities, but sharing a central core sequence depicted in light blue (Lanz et al., 1999). One sequence has been registered with the NCBI nucleotide database (AF092038). Alignment with chromosome 5q31.3 genomic sequence is provided. Introns and exons are represented by black lines and blue boxes, respectively. B) Schematic profile of the predicted secondary structure of human core SRA RNA. The secondary structure profile of SRA core sequence has been modeled using Mfold software (Zuker, 2003). Detailed structure of STR1, 10, 7 (Lanz et al., 2002) is provided. C) By doing site-directed mutagenesis experiment, six secondary structural motifs (STR1, 9, 10, 7, 11, 12) have been identified to participate in co-activation respectively. Especially, silent mutations in both SRT1 and STR7 of SRA could nullify above 80% SRA co-activation function (Lanz et al., 2002).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1191

The roles of SRA1 gene in breast cancer Yan Y, et al.

Firstly, SRA's coactivation function is activated by receptor activity. Indeed it was also confirmed that two pseudouridylases, Pus1p and Pus3p, which SRA enhance the activity of other nuclear receptor have also been characterized as co-activators (Zhao (NRs), such as retinoic acid receptors, thyroid et al., 2007). This modification alters the secondary receptors (Zhao et al., 2004; Xu and Koenig, 2004) structure and rigidity of the target SRA RNA as well as the activity of MyoD, a transcription molecules to promote proper folding, resulting in factor involved in skeletal myogenesis (Caretti et synergized co-activation function (Charette and al., 2006; Caretti et al., 2007); SF-1 and DAX-1, Gray, 2000). The other positive regulators include orphan NRs that plays critical roles in the the receptor co-activator 1 (SRC-1) (Lanz et al., regulation of sex determination, adrenal 1999) and the RNA helicases P68/72 (Watanabe et development steroidogenesis (Xu et al., 2009). al., 2001). SRC-1 belongs to p160 family co- Recently, Foulds et al. investigated the global activators (SRC1, SRC2/TIF2 and SRC3/AIB1), changes in gene expression by microarray analyses which can recruit other co-regulators to steroid in two human cancer cell lines when SRA RNA receptors as well as promote a functional synergy was depleted by small interfering RNAs (Foulds et between AF-1 and AF-2 domains (Louet and al., 2010). Unexpectedly, only a small subset of O'Malley, 2007; McKenna et al., 1999; Smith and direct estrogen receptor-target genes was affected in O'Malley, 2004). Using co-immunoprecipitation estradiol-treated MCF-7 cells. However, they found from an expression system consisting of Xenopus many target genes involved in diverse biological oocytes programmed with in vitro generated RNA, roles such as glucose uptake, cellular signaling, T3 SRA was found to associate with SRC-1 (Lanz et hormone generation were altered upon SRA al., 1999). The p72/p68 proteins are DEAD-box depletion. This suggests SRA has a much broader RNA binding helicases that can physically interact upstream biological impact within the cell than with p160 family proteins and with ERα. AF-1 simply a corgualtor of ER-signaling. region (Caretti et al., 2007). The p72/p68 is able to 2.6 SRA RNA expression and relevance to breast bind to SRA through a well conserved motif in the cancer DEAD box and synergizes with SRA and Different SRA transcripts, detected by Northern SRC2/TIF2 to co-activate ERα activity in the blot, have been observed in normal human tissues presence of estradiol (Caretti et al., 2006). (Lanz et al., 1999). SRA seems highly expressed in On the other side, SRA may also serve as a liver, skeletal muscle, adrenal gland and the platform to recruit some negative regulators pituitary gland, whereas intermediate expression consisting of the SMRT/HDAC1 associated levels are seen in the placenta, lung, kidney and repressor protein (SHARP) (Shi et al., 2001) and pancreas. Interestingly, brain and other typical the SRA stem-loop interacting RNA binding steroid-responsive tissues such as prostate, breast, protein (SLIRP) (Hatchell et al., 2006). SHARP uterus and ovary contained low levels of SRA RNA was found to physically interact with corepressors (Lanz et al., 1999). However, SRA RNA through its repression domain (RD) whereas it expression, assessed by RT-PCR amplification, is interacts with SRA through a RNA recognition increased during breast and ovarian tumorigenesis motif (RRM) (Shi et al., 2001). Similarly, SLIRP (Lanz et al., 2003; Leygue et al., 1999; Hussein- specifically binds to SRA STR-7 and attenuates Fikret and Fuller, 2005). Interestingly, SRA over- SRA-mediated transactivation of endogenous ER expression might characterize particular subtypes of (Hatchell et al., 2006). lesions among different tumors. Indeed, serous The emerging model of SRA action on ERα ovarian tumors expressed higher levels of SRA than signaling had been summarized: Pus1p granulosa cell tumors (Hussein-Fikret and Fuller, pseudouridylates specific SRA RNA uridine 2005). residues, leading to an optimum configuration of The involvement of SRA in ER action suggests this RNA. possible SRA role in breast tumor pathology. The resulting active form of SRA, could stabilize Indeed, ER-α-positive/PR-negative breast tumors complexes with p68 and SRC-1. In this case, expressed more SRA than ER-α-positive/PR- transcription of target genes with suitable ERE will positive breast tumors (Leygue et al., 1999), occur. In contrast, interaction with the negative whereas Tamoxifen-sensitive and resistant breast regulators SLIRP and SHARP with SRA RNA may tumors express similar levels (Murphy et al., 2002). result in the inhibition of ER-mediated However, generation of transgenic mice has transcription. It has been proposed that they might however demonstrated that over-expression of the act by sequestrating SRA by destabilizing the core SRA sequence in the mammary gland only led complex SRA/SRC-1 or by recruiting the nuclear to pre-neoplastic lesions but was not sufficient per receptor corepressor N-CoR at the promoter region se to induce tumorigenesis (Lanz et al., 2003). of silenced genes (Leygue, 2007). Notably, SRA gene depleted MDA-MB-231 cells 2.5 A broader biological role played by SRA are less invasive than control cells, indicating this It has been previously established that SRA action gene might be also critical for invasion (Foulds et is not exclusively limited to increasing steroid al., 2010).

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1192

The roles of SRA1 gene in breast cancer Yan Y, et al.

3. Coding SRA and SRAP both groups showed that SRAP is able to interact with transcriptional regulators. In Chooniedass- 3.1 Discovery of SRAP Kothari's unpublished results using mass Kawashima et al. reported in 2003 the cloning of a spectrometry, MBD3 (methyl-CpG binding domain new rat SRA cDNA mostly identical to the core protein 3, a member of the nucleosome remodeling SRA sequence from exon 2 to exon 5. This cDNA and histone deacetylase complex, Nurd), BAF 57 (a was, however translatable in vitro encoding a core subunit of SWI/SNF chromatin remodeling putative 16 kD protein starting at the third complex) and YB-1 (Y-box bindng protein, a methionine codon of the rat SRA cDNA sequence general transcription factor) have been found to (Kawashima et al., 2003). It should be stressed that interact with SRAP (Jung et al., 2005; the existence of a corresponding endogenous 16 kD Chooniedass-Kothari et al., 2010). By using a SRAP has never been proved. similar approach, Jung et al. also found that the In the nucleotide database of the National Center transcription regulators, such as, BAF 170 (BRG1 for Biotechnology Information (NCBI), most associated factor 170, also belonging to SWI/SNF human SRA sequences contain an intact core chromatin remodeling complex) and YB-1 are sequence (exon-2 to exon-5) but differ in their 5'- associated with SRAP (Jung et al., 2005). It is extremity. Interestingly, some variants having 5' necessary to point out that different cell line models end extention contain two start codons with a large and antibodies were used in these two groups. Jung open reading frame potentially encoding a 236/237 et al. used Hela cell lines and 743 antibody amino acid peptide. These cDNAs, as opposed to (commercial available rabbit polyclonal antibody) the original SRA, were translatable in vitro, as well whereas Chooniedass-Kothari used MCF7 cells as in vivo, leading to the production of a protein stably over-expressed V5 tagged SRAP and V5 localized both in the cytoplasm and the nucleus antibody. (Emberley et al., 2003). In addition, sequence of Interestingly, nobody has confirmed any protein- SRAP is highly conserved among chordate and the protein interaction between SRAP and those presence of endogenous SRAP had been found in potential partners by co-immunoprecipitation the testes, uterus, ovary and prostate, as well as experiments. mammary gland, lung and heart (Chooniedass- The observation that SRAP forms complexes with Kothari et al., 2004). Altogether, accumulated data transcription factors by mass spectrometric analysis has demonstrated that SRA1 gene products consist led us to investigate its direct association with of two characteristic entities: a functional RNA, transcription factors. which through its core sequence, can co-activate By using recombinant SRAP and protein arrays, transcription factor and a protein whose function Chooniedass-Kothari found that SRAP interact with remains yet to be fully understood. different transcription factors including ERα and 3.2 Function of SRAP ERβ with different binding affinities (Chooniedass- Chooniedass-Kothari et al. reported the existence of Kothari et al., 2010). To further validate the putative endogenous human SRA protein in breast interaction between ER and SRAP, we performed cancer cells (Chooniedass-Kothari et al., 2006). A GST pull down assay and a direct interaction decreased response to ERα activity was observed in between GST-SRAP and both full length radio- MCF-7 cells stably transfected with SRAP labeled estrogen receptor α and β was observed suggested that this protein might repress estrogen (Chooniedass-Kothari et al., 2010). Interestingly, receptor activities (Chooniedass-Kothari et al., Kawashima showed that SRAP is also able to 2006). This result contrasts with Kawashima's directly interact with the AF-2 domain of AR in results, who found that the transient transfection of vitro by doing GST pull down assay (Kawashima et full length rat SRA coding sequence and led to an al., 2003). activation of the response to androgen (Kurisu et 3.3 Alternative RNA splicing of SRA gene in al., 2006). It should be pointed out that, both coding breast cancer sequence of SRA used by these two groups also The balance between co-activators and co- contains the functional core sequence of SRA RNA repressors may ultimately controls estrogen action proven to co-activate ERα. Therefore, it is difficult in a given tissue (Lonard and O'Malley, 2006). A to draw any conclusions regarding the individual direct participation of this balance during breast function of SRAP on estrogen receptor activities tumorigenesis and cancer progression is now when functional SRA RNA and SRAP protein are suspected, and a search for possible means to co-expressed. control it has started worldwide (Perissi and In order to understand the functional role of SRAP Rosenfeld, 2005; Hall and McDonnell, 2005). independently of SRA RNA, two different groups Alternative splicing of SRA gene might control the have investigated physical protein properties by balance between the coding and non-coding SRA, tandem mass spectrometric analysis of SRAP co- and ultimately might function as the potential immunoprecipitation samples (Jung et al., 2005; mechanism to regulate the balance between co- Chooniedass-Kothari et al., 2010). Interestingly, activators and co-repressors.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1193

The roles of SRA1 gene in breast cancer Yan Y, et al.

Figure 2: Coding and non-coding SRA transcripts in human breast cancer cells. SRA1 gene, located on chromosome 5q31.3, consists of 5 exons (boxes) and 4 introns (plain lines). The originally described SRA sequence (AF092038) contains a core sequence (light blue), necessary and sufficient for SRA RNAs to act as co-activators (Lanz et al., 1999). Three coding isoforms have now been identified (SRA1, SRA2, SRA3), which mainly differ from AF092038 by an extended 5'-extremity containing AUG initiating codons (vertical white bar in exon 1). The stop codon of the resulting open reading frame is depicted by a black vertical bar in exon5. Black stars in exon 2 and 3 correspond to a point mutation (position 98 of the core: U to C) and a point mutation followed by a full codon (position 271 of the core: G to CGAC), respectively. Three non-coding SRA isoforms containing a differentially-spliced intron-1 have been characterized: FI, full intron-1 retention; PI, partial intron-1 retention; AD, alternative 5' donor and partial intron retention. Thick straight line, 60 bp of intron 1 retained in PI; triangulated lines represent splicing events (Modified from Cooper et al. 2009).

Both non-coding and coding SRA transcripts co- splicing-switching oligonucleotide strategy exist in breast cells (Figure 2, Hube et al., 2006). (Mercatante et al., 2001; Mercatante and Kole, Using a previously validated triple-primer PCR 2002). This approach resulted in an increase in the (TP-PCR) assay (Leygue et al., 1996), which allows production of intron retained transcripts, decrease in co-amplification and relative quantification of two the expression of SRAP, resulting in an observed transcripts sharing a common region but differing significant increase in the expression of the in another, we found that breast cancer cell lines co- urokinase plasminogen activator (uPA, PLAU), expressed normally spliced coding SRA RNA as gene intimately linked to invasion mechanisms well as SRA RNA containing intron-1 (Hube et al., (Harbeck et al., 2004) as well as of ERβ, involved, 2006). Interestingly, breast cancer cell lines differ as highlighted earlier, in breast cancer cell growth in their relative levels of coding/non-coding SRA (Han et al., 2005). transcripts. In particular, the three most invasive 3.4 SRAP expression and relevance to breast cell lines (MDA-MB-231, 468, and BT-20) cancer expressed the highest, whereas the "closest to SRAP expression was assessed by Tissue normal" MCF-10A1 breast cells expressed the Microarray (TMA) analysis of 372 breast tumors lowest relative levels of SRA intron-1 RNA. This (Yan et al., 2009). SRAP levels were significantly suggests that a balance changed toward the higher in estrogen receptor-alpha positive, in production of non-coding SRA1 RNA in breast progesterone receptor positive and in older patients cells might be associated with growth and/or (age > 64). When considering ER+ tumors, PR+ invasion properties (Hube et al., 2006). tumors, or young patients (≤ 64 years), patients Alternative splicing events result from the relative with high SRAP expression had a significantly local concentration of RNA binding proteins within worse breast cancer specific survival (BCSS) than the microenvironnement surrounding the nascent patients with low SRAP levels. SRAP also pre-mRNA (Mercatante et al., 2001). We were appeared as a very powerful indicator of poor recently able to artificially alter the balance prognostic for BCSS in the subset of ER+, node between coding and non-coding SRA1 RNAs in T5 negative and young breast cancer patients. breast cancer cells using a previously described Altogether suggest that SRAP levels might provide

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1194

The roles of SRA1 gene in breast cancer Yan Y, et al.

additional information on potential risk of truncated mRNA expression. Am J Pathol. 1996 recurrence and negative outcome in a specific set of Apr;148(4):1097-103 patients. Lanz RB, McKenna NJ, Onate SA, Albrecht U, Wong J, Tsai SY, Tsai MJ, O'Malley BW. A steroid receptor 4. Conclusion coactivator, SRA, functions as an RNA and is present in an Accumulated data suggest that the bi-faceted SRC-1 complex. Cell. 1999 Apr 2;97(1):17-27 SRA/SRAP system, including SRA non-coding Leygue E, Dotzlaw H, Watson PH, Murphy LC. Expression RNA and SRA protein, regulates estrogen receptor of the steroid receptor RNA activator in human breast signaling pathways and plays a critical role in tumors. Cancer Res. 1999 Sep 1;59(17):4190-3 breast tumorigenesis and tumor progression. SRA is McKenna NJ, Lanz RB, O'Malley BW. Nuclear receptor the first example of a new kind of molecules active coregulators: cellular and molecular biology. Endocr Rev. 1999 Jun;20(3):321-44 at both RNA as well as at the protein levels. Investigating and understanding this bi-faceted Charette M, Gray MW. Pseudouridine in RNA: what, system might open a new era of novel preventive or where, how, and why. IUBMB Life. 2000 May;49(5):341-51 therapeutic strategies for breast cancer patients. Jenuwein T, Allis CD. Translating the histone code. Science. 2001 Aug 10;293(5532):1074-80 Acknowledgements Mercatante DR, Bortner CD, Cidlowski JA, Kole R. This work is supported by the Canadian Institute of Modification of alternative splicing of Bcl-x pre-mRNA in Health Research / the Canadian Breast Cancer prostate and breast cancer cells. analysis of apoptosis and Research Alliance (MOP-129794) and the cell death. J Biol Chem. 2001 May 11;276(19):16411-7 CancerCare Manitoba Foundation (761017028). Y Shi Y, Downes M, Xie W, Kao HY, Ordentlich P, Tsai CC, Yan has been supported by the MHRC (Manitoba Hon M, Evans RM. Sharp, an inducible cofactor that Health Research Council) Studentship. integrates nuclear receptor repression and activation. Genes Dev. 2001 May 1;15(9):1140-51 Abbreviations Watanabe M, Yanagisawa J, Kitagawa H, Takeyama K, AF-1: activation function 1 Ogawa S, Arao Y, Suzawa M, Kobayashi Y, Yano T, Yoshikawa H, Masuhiro Y, Kato S. A subfamily of RNA- AF-2: activation function 2 binding DEAD-box proteins acts as an estrogen receptor AR: androgen receptor alpha coactivator through the N-terminal activation domain DAX-1: dosage-sensitive sex reversal-adrenal (AF-1) with an RNA coactivator, SRA. EMBO J. 2001 Mar hypoplasia congenital critical region on X 15;20(6):1341-52 chromosome gene 1; NR0B1 Lanz RB, Razani B, Goldberg AD, O'Malley BW. Distinct DBD: DNA binding domain RNA motifs are important for coactivation of receptors by steroid receptor RNA activator ER: estrogen receptor (SRA). Proc Natl Acad Sci U S A. 2002 Dec ERE: estrogen receptor 10;99(25):16081-6 GR: glucocorticoid receptor Mercatante DR, Kole R. Control of alternative splicing by LBD: ligand binding domain antisense oligonucleotides as a potential chemotherapy: MBD3: methyl-CpG binding domain protein 3 effects on gene expression. Biochim Biophys Acta. 2002 NRs: nuclear receptors Jul 18;1587(2-3):126-32 NCoR: nuclear co-repressor Murphy LC, Leygue E, Niu Y, Snell L, Ho SM, Watson PH. PLAU: urokinase plasminogen activator Relationship of coregulator and oestrogen receptor isoform PR: progesterone receptor expression to de novo tamoxifen resistance in human Pus1p: pseudouridine synthase 1 breast cancer. Br J Cancer. 2002 Dec 2;87(12):1411-6 Pus3p: pseudouridine syntheses 3 Deblois G, Giguère V. Ligand-independent coactivation of SERM: selective estrogen receptor modulators ERalpha AF-1 by steroid receptor RNA activator (SRA) via MAPK activation. J Steroid Biochem Mol Biol. 2003 SF-1: nuclear receptor steroidogenic factor 1 Jun;85(2-5):123-31 SDM: site-directed mutatagenesis SHARP: SMRT/HDAC1 associated repressor Emberley E, Huang GJ, Hamedani MK, Czosnek A, Ali D, Grolla A, Lu B, Watson PH, Murphy LC, Leygue E. protein Identification of new human coding steroid receptor RNA SLIRP: SRA stem-loop interacting RNA binding activator isoforms. Biochem Biophys Res Commun. 2003 protein Feb 7;301(2):509-15 SRA: steroid receptor RNA activator Kawashima H, Takano H, Sugita S, Takahara Y, Sugimura SRAP: steroid receptor RNA activator protein K, Nakatani T. A novel steroid receptor co-activator protein SR: serine/arginine-rich proteins (SRAP) as an alternative form of steroid receptor RNA- activator gene: expression in prostate cancer cells and SRC-1: steroid receptor co-activator 1 enhancement of androgen receptor activity. Biochem J. STR: secondary structural motif 2003 Jan 1;369(Pt 1):163-71 YB-1: Y-box bindng protein Lanz RB, Chua SS, Barron N, Söder BM, DeMayo F, O'Malley BW. Steroid receptor RNA activator stimulates References proliferation as well as apoptosis in vivo. Mol Cell Biol. 2003 Oct;23(20):7163-76 Leygue E, Murphy L, Kuttenn F, Watson P. Triple primer polymerase chain reaction. A new way to quantify Chooniedass-Kothari S, Emberley E, Hamedani MK, Troup S, Wang X, Czosnek A, Hube F, Mutawe M, Watson PH,

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1195

The roles of SRA1 gene in breast cancer Yan Y, et al.

Leygue E. The steroid receptor RNA activator is the first Hube F, Guo J, Chooniedass-Kothari S, Cooper C, functional RNA encoding a protein. FEBS Lett. 2004 May Hamedani MK, Dibrov AA, Blanchard AA, Wang X, Deng 21;566(1-3):43-7 G, Myal Y, Leygue E. Alternative splicing of the first intron of the steroid receptor RNA activator (SRA) participates in Coleman KM, Lam V, Jaber BM, Lanz RB, Smith CL. SRA the generation of coding and noncoding RNA isoforms in coactivation of estrogen receptor-alpha is phosphorylation- breast cancer cell lines. DNA Cell Biol. 2006 Jul;25(7):418- independent, and enhances 4-hydroxytamoxifen agonist 28 activity. Biochem Biophys Res Commun. 2004 Oct 8;323(1):332-8 Kurisu T, Tanaka T, Ishii J, Matsumura K, Sugimura K, Nakatani T, Kawashima H. Expression and function of Harbeck N, Kates RE, Gauger K, Willems A, Kiechle M, human steroid receptor RNA activator in prostate cancer Magdolen V, Schmitt M. Urokinase-type plasminogen cells: role of endogenous hSRA protein in androgen activator (uPA) and its inhibitor PAI-I: novel tumor-derived receptor-mediated transcription. Prostate Cancer Prostatic factors with a high prognostic and predictive impact in Dis. 2006;9(2):173-8 breast cancer. Thromb Haemost. 2004 Mar;91(3):450-6 Lonard DM, O'Malley BW. The expanding cosmos of Klinge CM, Jernigan SC, Mattingly KA, Risinger KE, Zhang nuclear receptor coactivators. Cell. 2006 May J. Estrogen response element-dependent regulation of 5;125(3):411-4 transcriptional activation of estrogen receptors alpha and beta by coactivators and corepressors. J Mol Endocrinol. Caretti G, Lei EP, Sartorelli V. The DEAD-box p68/p72 2004 Oct;33(2):387-410 proteins and the noncoding RNA steroid receptor activator SRA: eclectic regulators of disparate biological functions. Smith CL, O'Malley BW. Coregulator function: a key to Cell Cycle. 2007 May 15;6(10):1172-6 understanding tissue specificity of selective receptor modulators. Endocr Rev. 2004 Feb;25(1):45-71 Leygue E. Steroid receptor RNA activator (SRA1): unusual bifaceted gene products with suspected relevance to Xu B, Koenig RJ. An RNA-binding domain in the thyroid breast cancer. Nucl Recept Signal. 2007 Aug 3;5:e006 hormone receptor enhances transcriptional activation. J Biol Chem. 2004 Aug 6;279(32):33051-6 Lonard DM, O'malley BW. Nuclear receptor coregulators: judges, juries, and executioners of cellular regulation. Mol Zhao X, Patton JR, Davis SL, Florence B, Ames SJ, Cell. 2007 Sep 7;27(5):691-700 Spanjaard RA. Regulation of nuclear receptor activity by a pseudouridine synthase through posttranscriptional Louet JF, O'Malley BW. Coregulators in adipogenesis: modification of steroid receptor RNA activator. Mol Cell. what could we learn from the SRC (p160) coactivator 2004 Aug 27;15(4):549-58 family? Cell Cycle. 2007 Oct 15;6(20):2448-52 Hall JM, McDonnell DP. Coregulators in nuclear estrogen Zhao X, Patton JR, Ghosh SK, Fischel-Ghodsian N, Shen receptor action: from concept to therapeutic targeting. Mol L, Spanjaard RA. Pus3p- and Pus1p-dependent Interv. 2005 Dec;5(6):343-57 pseudouridylation of steroid receptor RNA activator controls a functional switch that regulates nuclear receptor Han B, Nakamura M, Mori I, Nakamura Y, Kakudo K. signaling. Mol Endocrinol. 2007 Mar;21(3):686-99 Urokinase-type plasminogen activator system and breast cancer (Review). Oncol Rep. 2005 Jul;14(1):105-12 Cooper C, Guo J, Yan Y, Chooniedass-Kothari S, Hube F, Hamedani MK, Murphy LC, Myal Y, Leygue E. Increasing Hussein-Fikret S, Fuller PJ. Expression of nuclear receptor the relative expression of endogenous non-coding Steroid coregulators in ovarian stromal and epithelial tumours. Mol Receptor RNA Activator (SRA) in human breast cancer Cell Endocrinol. 2005 Jan 14;229(1-2):149-60 cells using modified oligonucleotides. Nucleic Acids Res. Jung SY, Malovannaya A, Wei J, O'Malley BW, Qin J. 2009 Jul;37(13):4518-31 Proteomic analysis of steady-state nuclear hormone Xu B, Yang WH, Gerin I, Hu CD, Hammer GD, Koenig RJ. receptor coactivator complexes. Mol Endocrinol. 2005 Dax-1 and steroid receptor RNA activator (SRA) function Oct;19(10):2451-65 as transcriptional coactivators for steroidogenic factor 1 in Perissi V, Rosenfeld MG. Controlling nuclear receptors: steroidogenesis. Mol Cell Biol. 2009 Apr;29(7):1719-34 the circular logic of cofactor cycles. Nat Rev Mol Cell Biol. Yan Y, Skliris GP, Penner C, Chooniedass-Kothari S, 2005 Jul;6(7):542-54 Cooper C, Nugent Z, Blanchard A, Watson PH, Myal Y, Caretti G, Schiltz RL, Dilworth FJ, Di Padova M, Zhao P, Murphy LC, Leygue E. Steroid Receptor RNA Activator Ogryzko V, Fuller-Pace FV, Hoffman EP, Tapscott SJ, Protein (SRAP): a potential new prognostic marker for Sartorelli V. The RNA helicases p68/p72 and the estrogen receptor-positive/node-negative/younger breast noncoding RNA SRA are coregulators of MyoD and cancer patients. Breast Cancer Res. 2009;11(5):R67 skeletal muscle differentiation. Dev Cell. 2006 Chooniedass-Kothari S, Hamedani MK, Wang X, Leygue Oct;11(4):547-60 E.. The Steroid receptor RNA activator protein can act as a Chooniedass-Kothari S, Hamedani MK, Troup S, Hubé F, transcriptional repressor. FEBS Letters 2010: submitted. Leygue E. The steroid receptor RNA activator protein is Foulds CE, Tsimelzon A, Long W, Le A, Tsai SY, Tsai MJ, expressed in breast tumor tissues. Int J Cancer. 2006 Feb O'Malley BW. Research resource: expression profiling 15;118(4):1054-9 reveals unexpected targets and functions of the human Hatchell EC, Colley SM, Beveridge DJ, Epis MR, Stuart steroid receptor RNA activator (SRA) gene. Mol LM, Giles KM, Redfern AD, Miles LE, Barker A, Endocrinol. 2010 May;24(5):1090-105 MacDonald LM, Arthur PG, Lui JC, Golding JL, McCulloch RK, Metcalf CB, Wilce JA, Wilce MC, Lanz RB, O'Malley This article should be referenced as such: BW, Leedman PJ. SLIRP, a small SRA binding protein, is Yan Y, Cooper C, Leygue E. The roles of SRA1 gene in a nuclear receptor corepressor. Mol Cell. 2006 Jun breast cancer. Atlas Genet Cytogenet Oncol Haematol. 9;22(5):657-68 2010; 14(12):1190-1196.

Atlas Genet Cytogenet Oncol Haematol. 2010; 14(12) 1196

Atlas of Genetics and Cytogenetics

in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS

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

Subscription: The Atlas is FREE!

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

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

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS