Full issue
Transcription
Full issue
Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Volume 15 - Number 7 July 2011 The PDF version of the Atlas of Genetics and Cytogenetics in Oncology and Haematology is a reissue of the original articles published in collaboration with the Institute for Scientific and Technical Information (INstitut de l’Information Scientifique et Technique - INIST) of the French National Center for Scientific Research (CNRS) on its electronic publishing platform I-Revues. Online and PDF versions of the Atlas of Genetics and Cytogenetics in Oncology and Haematology are hosted by INIST-CNRS. Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Scope The Atlas of Genetics and Cytogenetics in Oncology and Haematology is a peer reviewed on-line journal in open access, devoted to genes, cytogenetics, and clinical entities in cancer, and cancer-prone diseases. It presents structured review articles ("cards") on genes, leukaemias, solid tumours, cancer-prone diseases, more traditional review articles on these and also on surrounding topics ("deep insights"), case reports in hematology, and educational items in the various related topics for students in Medicine and in Sciences. Editorial correspondance Jean-Loup Huret Genetics, Department of Medical Information, University Hospital F-86021 Poitiers, France tel +33 5 49 44 45 46 or +33 5 49 45 47 67 [email protected] or [email protected] Staff Mohammad Ahmad, Mélanie Arsaban, Marie-Christine Jacquemot-Perbal, Maureen Labarussias, Vanessa Le Berre, Anne Malo, Catherine Morel-Pair, Laurent Rassinoux, 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 Alessandro Beghini Anne von Bergh Judith Bovée Vasantha Brito-Babapulle Charles Buys Anne Marie Capodano Fei Chen Antonio Cuneo Paola Dal Cin Louis Dallaire Brigitte Debuire François Desangles Enric Domingo-Villanueva Ayse Erson Richard Gatti Ad Geurts van Kessel Oskar Haas Anne Hagemeijer Nyla Heerema Jim Heighway Sakari Knuutila Lidia Larizza Lisa Lee-Jones Edmond Ma Roderick McLeod Cristina Mecucci Yasmin Mehraein Fredrik Mertens Konstantin Miller Felix Mitelman Hossain Mossafa Stefan Nagel Florence Pedeutour Elizabeth Petty Susana Raimondi Mariano Rocchi Alain Sarasin Albert Schinzel Clelia Storlazzi Sabine Strehl Nancy Uhrhammer Dan Van Dyke Roberta Vanni Franck Viguié José Luis Vizmanos Thomas Wan (Ankara, Turkey) (Milan, Italy) (Rotterdam, The Netherlands) (Leiden, The Netherlands) (London, UK) (Groningen, The Netherlands) (Marseille, France) (Morgantown, West Virginia) (Ferrara, Italy) (Boston, Massachussetts) (Montreal, Canada) (Villejuif, France) (Paris, France) (London, UK) (Ankara, Turkey) (Los Angeles, California) (Nijmegen, The Netherlands) (Vienna, Austria) (Leuven, Belgium) (Colombus, Ohio) (Liverpool, UK) (Helsinki, Finland) (Milano, Italy) (Newcastle, UK) (Hong Kong, China) (Braunschweig, Germany) (Perugia, Italy) (Homburg, Germany) (Lund, Sweden) (Hannover, Germany) (Lund, Sweden) (Cergy Pontoise, France) (Braunschweig, Germany) (Nice, France) (Ann Harbor, Michigan) (Memphis, Tennesse) (Bari, Italy) (Villejuif, France) (Schwerzenbach, Switzerland) (Bari, Italy) (Vienna, Austria) (Clermont Ferrand, France) (Rochester, Minnesota) (Montserrato, Italy) (Paris, France) (Pamplona, Spain) (Hong Kong, China) Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Solid Tumours Section Genes Section Genes / Leukaemia Sections Solid Tumours Section Leukaemia Section Deep Insights Section Solid Tumours Section Genes / Deep Insights Sections Leukaemia Section Genes / Solid Tumours Section Education Section Deep Insights Section Leukaemia / Solid Tumours Sections Solid Tumours Section Solid Tumours Section Cancer-Prone Diseases / Deep Insights Sections Cancer-Prone Diseases Section Genes / Leukaemia Sections Deep Insights Section Leukaemia Section Genes / Deep Insights Sections Deep Insights Section Solid Tumours Section Solid Tumours Section Leukaemia Section Deep Insights / Education Sections Genes / Leukaemia Sections Cancer-Prone Diseases Section Solid Tumours Section Education Section Deep Insights Section Leukaemia Section Deep Insights / Education Sections Genes / Solid Tumours Sections Deep Insights Section Genes / Leukaemia Section Genes Section Cancer-Prone Diseases Section Education Section Genes Section Genes / Leukaemia Sections Genes / Cancer-Prone Diseases Sections Education Section Solid Tumours Section Leukaemia Section Leukaemia Section Genes / Leukaemia Sections Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Volume 15, Number 7, July 2011 Table of contents Gene Section EIF3A (eukaryotic translation initiation factor 3, subunit A) Ji-Ye Yin, Zizheng Dong, Jian-Ting Zhang 544 ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Roopika Menon, Martin Braun, Sven Perner 547 ETV4 (ets variant 4) Yasuyoshi Miyata 554 GPC5 (glypican 5) Khin Thway, Joanna Selfe, Janet Shipley 557 GSDMA (gasdermin A) Norihisa Saeki, Hiroki Sasaki 560 IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) Theoni Trangas, Panayotis Ioannidis 562 MTA3 (metastasis associated 1 family, member 3) Ansgar Brüning, Ioannis Mylonas 567 NMT1 (N-myristoyltransferase 1) Ponniah Selvakumar, Sujeet Kumar, Jonathan R Dimmock, Rajendra K Sharma 570 PAEP (progestagen-associated endometrial protein) Hannu Koistinen, Markku Seppälä 576 SHBG (sex hormone-binding globulin) Nicoletta Fortunati, Maria Graziella Catalano 582 SLC39A6 (solute carrier family 39 (zinc transporter), member 6) Shin Hamada, Kennichi Satoh, Tooru Shimosegawa 586 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) Massimo Nabissi, Giorgio Santoni 588 WRAP53 (WD repeat containing, antisense to TP53) Marianne Farnebo 596 YBX1 (Y box binding protein 1) Valentina Evdokimova, Alexey Sorokin 598 ZBTB33 (zinc finger and BTB domain containing 33) Michael R Dohn, Albert B Reynolds 605 Leukaemia Section t(3;5)(p21;q32) Jean-Loup Huret Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 608 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Deep Insight Section Role of HB-EGF in cancer Rosalyn M Adam Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 610 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review EIF3A (eukaryotic translation initiation factor 3, subunit A) Ji-Ye Yin, Zizheng Dong, Jian-Ting Zhang Department of Pharmacology and Toxicology and IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA (JYY, ZD, JTZ) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/EIF3AID40425ch10q26.html DOI: 10.4267/2042/45982 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology spectrin domain, and a 10-amino acid repeat domain (Pincheira et al., 2001b). It has phosphorylation sites at Ser-881, Ser-1198, Ser-1336 and Ser-1364 (Damoc et al., 2007). The PCI domain spans from amino acid 405 to 495, which contains purely alpha-helix (Pincheira et al., 2001b). Since most of the proteins containing this domain are part of a multi-protein complex, it is tempting to speculate that this domain may be involved in the interaction of eIF3a with other molecules in eIF3 (Hofmann and Bucher, 1998). The spectrin domain, which consists of 112 amino acids, is a sequence almost identical to spectrin, an actin-binding protein (Pascual et al., 1997). Although the exact function of this domain remains unknown, it may be responsible for the binding of eIF3a to actin filaments (Pincheira et al., 2001a). The 10-amino acid repeat domain spanning 925-1172 amino acids is the largest domain of eIF3a. It can be divided into about 25 repeats of DDDRGPRRGA (Johnson et al., 1997; Pincheira et al., 2001b). This domain has been suggested to contribute to interaction of eIF4B and eIF3a (Methot et al., 1996). Regulatory role in gene expression: eIF3a not only functions as a regular translation initiation factor and participates in translation initiation of global mRNAs, it also regulates the translation of a subset of mRNAs which are involved in cell cycle, tumorigenesis and DNA repair (Yin et al., 2010). It has been observed that overexpression of ectopic eIF3a increases the expression of ribonucleotide reductase Identity Other names: EIF3; EIF3S10; KIAA0139; P167; TIF32; eIF3-p170; eIF3-theta; p180; p185 HGNC (Hugo): EIF3A Location: 10q26.11 DNA/RNA Description The eIF3a gene spans over a region of 46 kbp DNA including 22 coding exons and 2 non-coding exons (exon 2 and exon 10). Transcription The eIF3a mRNA consists of about 5256 nucleotides with an open reading frame (ORF) of 4149 bases. Pseudogene No pseudogene has been identified. Protein Description Structure: The eIF3a protein consists of 1382 amino acid residues with an apparent molecular weight of ~170 kDa as determined using SDS-PAGE (Pincheira et al., 2001b). Its primary sequence contains a PCI (Proteasome, COP9, Initiation factor 3) domain, a Schematic presentation of eIF3a domain structure. Human eIF3a consists of 1382 amino acid residues with three putative domains of PCI, spectrin, and 10-amino acid repeat. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 544 EIF3A (eukaryotic translation initiation factor 3, subunit A) Yin JY, et al. be associated with higher risk of breast cancers (Olson et al., 2009). M2 (RRM2) and alpha-tubulin, but decreases that of p27kip without affect their mRNA levels (Dong and Zhang, 2003; Dong et al., 2004). Recently, it has also been found that eIF3a suppresses the synthesis of DNA repair proteins including: XPA, XPC, RPA 14, RPA 32 and RPA 70 KDa (Yin et al., unpublished data). Although the detailed mechanism of eIF3a regulation in translational control is yet to be determined, it is thought that eIF3a may regulate these genes at their 5'and 3'-UTRs (Dong and Zhang, 2003; Dong et al., 2004). Binding with other molecule: Since eIF3a is the largest subunit of the eIF3 complex, the interaction between eIF3a and other subunits of eIF3 were intensively studied. It can bind with eIF3b (Methot et al., 1997), eIF3c (Valasek et al., 2002), eIF3f (Asano et al., 1997), eIF3h (Asano et al., 1997), eIF3j (Valasek et al., 1999) and eIF3k (Mayeur et al., 2003). During the translation initiation, the amino terminal domain of eIF3a can bind with 40S protein RPS0A, while the C terminal domain binds with the 18S rRNA (Valasek et al., 2003). Apart from above molecule, eIF3a has also been shown to interact with eIF4B (Methot et al., 1996), actin (Pincheira et al., 2001a), and cytokeratin 7 (Lin et al., 2001). Implicated in Breast cancer Note eIF3a was overexpressed in breast cancer tissues. Oncogenesis The eIF3a was highly expressed in all tested tissues from breast cancer patients compared with normal control tissues, which indicated that it may contribute to the oncogenesis of breast cancer (Bachmann et al., 1997). Cervical carcinoma Note eIF3a was found to be a molecular parameter of predicting cervical carcinoma progression and prognoses. Prognosis Patients with high eIF3a expression have better prognosis than those with lower ones, thus it will be useful in predicting cervical cancer prognosis (Dellas et al., 1998). Expression Gastric carcinoma eIF3a is ubiquitously expressed in all human tissues (Nagase et al., 1995; Scholler and Kanner, 1997; Pincheira et al., 2001b). However, its expression is higher in proliferating tissues such as bone marrow, thymus and fetal tissues (Pincheira et al., 2001b). Note eIF3a is an early tumor maker of gastric carcinoma. Oncogenesis eIF3a was highly expressed in well differentiated, early invasive stage and no-metastases gastric carcinoma (Chen and Burger, 2004). Localisation eIF3a has been found in both cytoplasmic and membrane fractions and the cytoplasmic eIF3a appears to be phosphorylated at its serine residues (Pincheira et al., 2001a). However, 70-80% of eIF3a is cytoplasmic. Lung cancer Homology Note eIF3a is highly expressed in lung cancer compared with normal tissues. Prognosis eIF3a expression in human lung cancers negatively correlates with patient response to platinum-based chemotherapy, suggesting that lung cancer patients with higher eIF3a expression level respond better to platinum-based chemotherapy (Yin et al., unpublished findings). Oncogenesis eIF3a was over-expressed in all types of human lung cancer. Furthermore, it is ubiquitously highly expressed in proliferating and developing tissues. This suggested eIF3a may be involved in oncogenesis of lung cancer (Pincheira et al., 2001b). Centrosomin A and B have strong homology to eIF3a. The spectrin domain is essentially identical to spectrin. Esophagus squamous-cell carcinoma Function eIF3a has been shown to play important roles in the biological processes: translational initiation (including generation of ribosomal subunit from 80S ribosomes, 43S pre-initiation complex formation and 48S preinitiation complex formation) (Dong and Zhang, 2006), regulation of mRNA translation (Dong and Zhang, 2003; Dong et al., 2004), differentiation and development (Liu et al., 2007), apoptosis (Nakai et al., 2005), cell cycle regulation (Dong et al., 2009), oncogenesis (Dong and Zhang, 2006; Zhang et al., 2007), and drug response (unpublished observations). Note eIF3a may be a biomaker of esophagus squamous-cell carcinoma. Prognosis Patients with higher eIF3a expression have better Mutations Note Two SNPs (rs10787899 and rs3824830) were found to Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 545 EIF3A (eukaryotic translation initiation factor 3, subunit A) Yin JY, et al. Pincheira R, Chen Q, Zhang JT. Identification of a 170-kDa protein over-expressed in lung cancers. Br J Cancer. 2001b Jun 1;84(11):1520-7 overall survival and fewer tumor metastases than those with lower ones (Chen and Burger, 1999). References Valásek L, Nielsen KH, Hinnebusch AG. Direct eIF2-eIF3 contact in the multifactor complex is important for translation initiation in vivo. EMBO J. 2002 Nov 1;21(21):5886-98 Nagase T, Seki N, Tanaka A, Ishikawa K, Nomura N. Prediction of the coding sequences of unidentified human genes. IV. The coding sequences of 40 new genes (KIAA0121KIAA0160) deduced by analysis of cDNA clones from human cell line KG-1. DNA Res. 1995 Aug 31;2(4):167-74, 199-210 Dong Z, Zhang JT. EIF3 p170, a mediator of mimosine effect on protein synthesis and cell cycle progression. Mol Biol Cell. 2003 Sep;14(9):3942-51 Dong Z, Zhang JT. Initiation factor eIF3 and regulation of mRNA translation, cell growth, and cancer. Crit Rev Oncol Hematol. 2006 Sep;59(3):169-80 Méthot N, Song MS, Sonenberg N. A region rich in aspartic acid, arginine, tyrosine, and glycine (DRYG) mediates eukaryotic initiation factor 4B (eIF4B) self-association and interaction with eIF3. Mol Cell Biol. 1996 Oct;16(10):5328-34 Valásek L, Mathew AA, Shin BS, Nielsen KH, Szamecz B, Hinnebusch AG. The yeast eIF3 subunits TIF32/a, NIP1/c, and eIF5 make critical connections with the 40S ribosome in vivo. Genes Dev. 2003 Mar 15;17(6):786-99 Asano K, Vornlocher HP, Richter-Cook NJ, Merrick WC, Hinnebusch AG, Hershey JW. Structure of cDNAs encoding human eukaryotic initiation factor 3 subunits. Possible roles in RNA binding and macromolecular assembly. J Biol Chem. 1997 Oct 24;272(43):27042-52 Chen G, Burger MM. p150 overexpression in gastric carcinoma: the association with p53, apoptosis and cell proliferation. Int J Cancer. 2004 Nov 10;112(3):393-8 Bachmann F, Bänziger R, Burger MM. Cloning of a novel protein overexpressed in human mammary carcinoma. Cancer Res. 1997 Mar 1;57(5):988-94 Dong Z, Liu LH, Han B, Pincheira R, Zhang JT. Role of eIF3 p170 in controlling synthesis of ribonucleotide reductase M2 and cell growth. Oncogene. 2004 May 6;23(21):3790-801 Johnson KR, Merrick WC, Zoll WL, Zhu Y. Identification of cDNA clones for the large subunit of eukaryotic translation initiation factor 3. Comparison of homologues from human, Nicotiana tabacum, Caenorhabditis elegans, and Saccharomyces cerevisiae. J Biol Chem. 1997 Mar 14;272(11):7106-13 Nakai Y, Shiratsuchi A, Manaka J, Nakayama H, Takio K, Zhang JT, Suganuma T, Nakanishi Y. Externalization and recognition by macrophages of large subunit of eukaryotic translation initiation factor 3 in apoptotic cells. Exp Cell Res. 2005 Sep 10;309(1):137-48 Méthot N, Rom E, Olsen H, Sonenberg N. The human homologue of the yeast Prt1 protein is an integral part of the eukaryotic initiation factor 3 complex and interacts with p170. J Biol Chem. 1997 Jan 10;272(2):1110-6 Mayeur GL, Fraser CS, Peiretti F, Block KL, Hershey JW. Characterization of eIF3k: a newly discovered subunit of mammalian translation initiation factor elF3. Eur J Biochem. 2003 Oct;270(20):4133-9 Pascual J, Castresana J, Saraste M. Evolution of the spectrin repeat. Bioessays. 1997 Sep;19(9):811-7 Damoc E, Fraser CS, Zhou M, Videler H, Mayeur GL, Hershey JW, Doudna JA, Robinson CV, Leary JA. Structural characterization of the human eukaryotic initiation factor 3 protein complex by mass spectrometry. Mol Cell Proteomics. 2007 Jul;6(7):1135-46 Scholler JK, Kanner SB. The human p167 gene encodes a unique structural protein that contains centrosomin A homology and associates with a multicomponent complex. DNA Cell Biol. 1997 Apr;16(4):515-31 Liu Z, Dong Z, Yang Z, Chen Q, Pan Y, Yang Y, Cui P, Zhang X, Zhang JT. Role of eIF3a (eIF3 p170) in intestinal cell differentiation and its association with early development. Differentiation. 2007 Sep;75(7):652-61 Dellas A, Torhorst J, Bachmann F, Bänziger R, Schultheiss E, Burger MM. Expression of p150 in cervical neoplasia and its potential value in predicting survival. Cancer. 1998 Oct 1;83(7):1376-83 Hofmann K, Bucher P. The PCI domain: a common theme in three multiprotein complexes. Trends Biochem Sci. 1998 Jun;23(6):204-5 Zhang L, Pan X, Hershey JW. Individual overexpression of five subunits of human translation initiation factor eIF3 promotes malignant transformation of immortal fibroblast cells. J Biol Chem. 2007 Feb 23;282(8):5790-800 Chen G, Burger MM. p150 expression and its prognostic value in squamous-cell carcinoma of the esophagus. Int J Cancer. 1999 Apr 20;84(2):95-100 Dong Z, Liu Z, Cui P, Pincheira R, Yang Y, Liu J, Zhang JT. Role of eIF3a in regulating cell cycle progression. Exp Cell Res. 2009 Jul 1;315(11):1889-94 Valásek L, Hasek J, Trachsel H, Imre EM, Ruis H. The Saccharomyces cerevisiae HCR1 gene encoding a homologue of the p35 subunit of human translation initiation factor 3 (eIF3) is a high copy suppressor of a temperature-sensitive mutation in the Rpg1p subunit of yeast eIF3. J Biol Chem. 1999 Sep 24;274(39):27567-72 Olson JE, Wang X, Goode EL, Pankratz VS, Fredericksen ZS, Vierkant RA, Pharoah PD, Cerhan JR, Couch FJ. Variation in genes required for normal mitosis and risk of breast cancer. Breast Cancer Res Treat. 2010 Jan;119(2):423-30 Yin JY, Dong Z, Liu ZQ, Zhang JT. Translational control gone awry: a new mechanism of tumorigenesis and novel targets of cancer treatments. Biosci Rep. 2010 Oct;31(1):1-15 Lin L, Holbro T, Alonso G, Gerosa D, Burger MM. Molecular interaction between human tumor marker protein p150, the largest subunit of eIF3, and intermediate filament protein K7. J Cell Biochem. 2001;80(4):483-90 This article should be referenced as such: Yin JY, Dong Z, Zhang JT. EIF3A (eukaryotic translation initiation factor 3, subunit A). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):544-546. Pincheira R, Chen Q, Huang Z, Zhang JT. Two subcellular localizations of eIF3 p170 and its interaction with membranebound microfilaments: implications for alternative functions of p170. Eur J Cell Biol. 2001a Jun;80(6):410-8 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 546 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Roopika Menon, Martin Braun, Sven Perner Institute of Pathology, University Hospital Tuebingen, Liebermeisterstr. 8, D-72076 Tuebingen, Germany (RM, MB, SP) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/ERGID53ch21q22.html DOI: 10.4267/2042/45991 This article is an update of : Rainis-Ganon L, Izraeli S. ERG (v-ets erythroblastosis virus E26 oncogene like (avian)). Atlas Genet Cytogenet Oncol Haematol 2007;11(1) This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity DNA/RNA Other names: erg-3; p55 HGNC (Hugo): ERG Location: 21q22.2 Description The ERG gene belongs to the erythroblast transformation-specific (ETS) family of transcriptions factors. The ERG gene (ETS related gene 1) is located on chromosome 21, and consists of 17 exons, approximately 300 kb DNA in length. Transcription The ERG gene forms 20 known transcripts (ranging from 560 to 5034 bp in length), amongst which 15 are coding for proteins, and 5 are non-coding. 8 alternative splice variants are known. Probe(s) - Courtesy Mariano Rocchi, Resources for Molecular Cytogenetics. Pseudogene No observed pseudogenes. ERG gene locus on the q-arm of chromosome 21 (21q22.2) spanning from 39751949 to 40033704 (according to UCSC genome browser, Feb. 2009 GRCh37/hg19, and Ensemble, Aug. 2010). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 547 ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Menon R, et al. Protein Implicated in Description Ewing's sarcoma Amongst the 20 known transcripts of the ERG gene, 15 are protein coding. The 15 proteins range from 171 to 486 amino acids in length, and up to 55 kDa in weight. Prognosis The prognostic relevance of an ERG gene fusion or an ERG overexpression in Ewing's sarcoma (EWS-ETS fusion type) is yet to be determined. So far, no prognostic relevance could be shown. Hybrid/Mutated gene If a gene fusion occurs in Ewing's sarcoma, most frequently it is a fusion of EWS to FLI-1 (in app. 85% of cases) or ERG (in app. 10% of cases). Other ETS genes rarely serve as EWS gene fusion partners (in app. 5% of cases). Abnormal protein The EWS gene fuses with the carboxyl terminal of ERG containing the ETS DNA binding domain of ERG. Therefore, the resulting fusion protein deregulates a large number of genes by so far poorly defined mechanisms. Oncogenesis In a transgenic mouse model expression of the EWSERG in lymphoid progenitors induced T-cell leukemia. Expression On the protein level, ERG is mainly expressed in the nucleus and is rarely seen in the cytoplasm. Basically, in the GNF SymAtlas database, major ERG expression was found to be in CD34+ cells (that include both hematopoietic stem cells and endothelial cells). In detail, ERG is reported to be expressed during early T and B cell development, and down-regulated in later stages of B and T cell differentiation. Also, ERG is expressed in platelets, megakaryoblastic cell lines, primary megakaryoblastic leukemia (AMKL or M7AML) in Down syndrome patients. Furthermore, ERG is strongly expressed in ERG gene rearranged prostate tissue (both in prostatic cancer tissue and adjacent prostatic intraepithelial neoplasia lesions). Of note, using immunohistochemistry, ERG expression is regularly observed in lymphocytes and small blood vessels. Acute myeloid leukemia (AML) Localisation Prognosis Several studies suggest a poorer prognosis for FUSERG gene fusion positive AML as compared to nonfused AML. Moreover, an ERG overexpression, not necessarily due to the FUS-ERG gene fusion, predicts an increased relapse risk and shorter survival in AML patients. However, the exact contribution of ERG overexpression to myeloid leukemiogenesis and progression is still unknown. Hybrid/Mutated gene In the FUS-ERG gene fusion, the FUS gene fuses with the carboxyl terminal of ERG containing the ETS DNA binding domain of ERG. Of note, in a single case, a gene fusion of ERG with the myeloid ELF-like factor 1 (ELF4) was detected. Oncogenesis The FUS-ERG fusion protein helps in activating the oncogenic activity of transcription factors. Predominantely nuclear and rarely cytoplasmic. Function The ERG protein is a member of the ETS-family and is known to bind to purine-rich sequences. ERG and other members of the same family are downstream regulators of mitogenic signal transduction pathways. They are key regulators of embryonic development, cell proliferation, differentiation, angiogenesis, inflammation, and apoptosis. At the DNA level, isoforms of ERG are known to regulate methylation. Further, ERG is required for platelet adhesion to the subendothelium, inducing vascular cell remodeling. Moreover, hematopoesis, as well as the differentiation and maturation of megakaryocytic cells are regulated by ERG. Overexpression of the ERG protein is suggested to aid in forming solid tumors. However, the exact molecular mechanisms of ERG as a transcription factor are still unknown. Prostate cancer Homology Prognosis The body of literature is controversial about the prognostic relevance of ERG rearrangements in prostate cancer. Some studies reported an association of the ERG rearrangement with adverse clinical parameters (i.e. time to prostate cancer specific death and the development of hormone-refractory metastasis). A member of the ETS transcription factors, most homologous to FLI1. Mutations Note No known mutations. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 548 ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Menon R, et al. Schematic displaying ERG rearrangement status (via FISH) in prostate cancer. The red-labelled centromeric and the green-labelled telomeric probes span the ERG locus on chromosomes 21. If a break-apart occurs, the green signal is either lost (ERG rearrangement through deletion) or translocated (ERG rearrangement through insertion). An ERG break-apart as determined by FISH accounts for a fusion of ERG mainly with TMPRSS2 but also with other 5' fusion partners such as SLC45A3, HERPUD1, or NDRG1. A: Both alleles with wild type (wt) ERG. B: One allele with ERG rearrangement through deletion (single red signal) and the other allele with wt ERG (yellow signal). C: One allele with ERG rearrangement through insertion (separated red and green signal) and the other allele with wt ERG (yellow signal). On the other hand, some studies demonstrated an association of ERG rearrangement with parameters of more favourable outcome, such as lower Gleason score, stage, volume, better overall survival, or late biochemical recurrence. Interestingly, a subset of studies without any such association was reported as well. Hybrid/Mutated gene In approximately 50% of prostate cancers, the ERG gene is rearranged, i.e. fused to another gene. In case of a rearrangement, TMPRSS2 is the ERG 5' fusion partner in the vast majority of cases (app. 85%). Other known, but rarely occurring ERG fusion partners include NDRG1, SLC45A3, and HERPUD1. The ERG gene rearrangement either occurs due to a deletion, or an insertion. Abnormal protein An ERG gene rearrangement in prostate cancer mainly results in an androgen dependant ERG overexpression. Oncogenesis In-vitro models complement that over expression of truncated ERG and various TMPRSS2-ERG isoforms increase cell migration and invasion. In-vivo recapitulation of ETS fusions by prostate specific expression of truncated ERG in mice resulted in the development of PIN but not carcinoma. Subsequent Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) work on transgenic TMPRSS2-ERG mice develop PIN progressing to invasive cancer, but only in the context of PI3-kinase pathway activation. TMPRSS2-ERGpositive human tumors are also enriched for PTEN loss, suggesting cooperation in prostate tumorigenesis. Acute lymphoblastic leukemia (ALL) Prognosis Overexpression of ERG was shown to be a risk factor in adult T-ALL. ALL patients with ERG overexpression were four times more likely to fail longterm recurrence free survival, indicating inferior survival. Oncogenesis Studies assessing ERG overexpression in ALL have shown that due to the involvement of ERG in T-cell development, it may have an oncogenic potential. Acute megakaryoblastic leukemia (AMKL) Prognosis Even though ERG is highly considered to be oncogenic in AMKL, no prognostic relevance has been determined. Oncogenesis ERG was found to be expressed megakaryoblastic leukemic cell lines and in primary leukemic cells from 549 ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Menon R, et al. DS patients. Moreover, in mouse models, expression of ERG drove megakaryopoiesis and lead to a rapid development of aggressive leukemia. ERG involvement in endothelial development Note ERG has been reported to regulate genes involved in chondrogenesis and angiogenesis and functions as a modulator of endothelial cell differentiation. In an invitro study, the decrease of the ERG protein follows a reduction in endothelial cell proliferation and vascular tube formation. In human umbilical vein endothelial cell lines, vascular endothelial growth factor (VEGF) was seen to significantly up-regulate ERG expression. Controversially, on the other hand, ERG expression was shown to inhibit responsiveness to the VEGF receptor in a Down Syndrome mouse model. Alzheimer's disease (AD) Note ERG has been linked to AD, due to an ERG protein overexpression as compared to control patients. This is further supported by experiments conducted on patients suffering from Down syndrome, who gradually develop AD-like symptoms, linked to ERG overexpression. Down syndrome (DS) Note DS is associated with trisomy of the chromosome 21, where the ERG gene is located. The trisomy is considered to be responsible for an ERG overexpression. In a DS mouse model, an induced functional disomy of the ERG allele corrects some pathologic features of the disease, including myeloproliferation and progenitor cell expansion, suggesting a pathogenic effect of trisomy driven ERG overexpression. ERG involvement in lymphoid development Note ERG was reported to be expressed in during early T and B cell development, and to be down-regulated in later stages of B and T cell differentiation. In detail, the ERG protein modulates the maturation of lymphoid cells. Interestingly, ERG overexpression is associated with T-ALL. Breakpoints References Duterque-Coquillaud M, Niel C, Plaza S, Stehelin D. New human erg isoforms generated by alternative splicing are transcriptional activators. Oncogene. 1993 Jul;8(7):1865-73 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Giovannini M, Biegel JA, Serra M, Wang JY, Wei YH, Nycum L, Emanuel BS, Evans GA. EWS-erg and EWS-Fli1 fusion transcripts in Ewing's sarcoma and primitive neuroectodermal 550 ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Menon R, et al. tumors with variant translocations. J Clin Invest. 1994 Aug;94(2):489-96 specific transcription factor expression. J Immunol. 2005 Nov 1;175(9):5912-22 Kong XT, Ida K, Ichikawa H, Shimizu K, Ohki M, Maseki N, Kaneko Y, Sako M, Kobayashi Y, Tojou A, Miura I, Kakuda H, Funabiki T, Horibe K, Hamaguchi H, Akiyama Y, Bessho F, Yanagisawa M, Hayashi Y. Consistent detection of TLS/FUSERG chimeric transcripts in acute myeloid leukemia with t(16;21)(p11;q22) and identification of a novel transcript. Blood. 1997 Aug 1;90(3):1192-9 Anderson MK. At the crossroads: diverse roles of early thymocyte transcriptional regulators. Immunol Rev. 2006 Feb;209:191-211 Baldus CD, Burmeister T, Martus P, Schwartz S, Gökbuget N, Bloomfield CD, Hoelzer D, Thiel E, Hofmann WK. High expression of the ETS transcription factor ERG predicts adverse outcome in acute T-lymphoblastic leukemia in adults. J Clin Oncol. 2006 Oct 10;24(29):4714-20 Hewett PW, Nishi K, Daft EL, Clifford Murray J. Selective expression of erg isoforms in human endothelial cells. Int J Biochem Cell Biol. 2001 Apr;33(4):347-55 Moore SD, Offor O, Ferry JA, Amrein PC, Morton CC, Dal Cin P. ELF4 is fused to ERG in a case of acute myeloid leukemia with a t(X;21)(q25-26;q22). Leuk Res. 2006 Aug;30(8):1037-42 Iwamoto M, Higuchi Y, Enomoto-Iwamoto M, Kurisu K, Koyama E, Yeh H, Rosenbloom J, Pacifici M. The role of ERG (ets related gene) in cartilage development. Osteoarthritis Cartilage. 2001;9 Suppl A:S41-7 Loughran SJ, Kruse EA, Hacking DF, de Graaf CA, Hyland CD, Willson TA, Henley KJ, Ellis S, Voss AK, Metcalf D, Hilton DJ, Alexander WS, Kile BT. The transcription factor Erg is essential for definitive hematopoiesis and the function of adult hematopoietic stem cells. Nat Immunol. 2008 Jul;9(7):810-9 McLaughlin F, Ludbrook VJ, Cox J, von Carlowitz I, Brown S, Randi AM. Combined genomic and antisense analysis reveals that the transcription factor Erg is implicated in endothelial cell differentiation. Blood. 2001 Dec 1;98(12):3332-9 Carver BS, Tran J, Gopalan A, Chen Z, Shaikh S, Carracedo A, Alimonti A, Nardella C, Varmeh S, Scardino PT, CordonCardo C, Gerald W, Pandolfi PP. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nat Genet. 2009 May;41(5):619-24 Yeoh EJ, Ross ME, Shurtleff SA, Williams WK, Patel D, Mahfouz R, Behm FG, Raimondi SC, Relling MV, Patel A, Cheng C, Campana D, Wilkins D, Zhou X, Li J, Liu H, Pui CH, Evans WE, Naeve C, Wong L, Downing JR. Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell. 2002 Mar;1(2):133-43 King JC, Xu J, Wongvipat J, Hieronymus H, Carver BS, Leung DH, Taylor BS, Sander C, Cardiff RD, Couto SS, Gerald WL, Sawyers CL. Cooperativity of TMPRSS2-ERG with PI3-kinase pathway activation in prostate oncogenesis. Nat Genet. 2009 May;41(5):524-6 Oikawa T, Yamada T. Molecular biology of the Ets family of transcription factors. Gene. 2003 Jan 16;303:11-34 Salek-Ardakani S, Smooha G, de Boer J, Sebire NJ, Morrow M, Rainis L, Lee S, Williams O, Izraeli S, Brady HJ. ERG is a megakaryocytic oncogene. Cancer Res. 2009 Jun 1;69(11):4665-73 Baldus CD, Liyanarachchi S, Mrózek K, Auer H, Tanner SM, Guimond M, Ruppert AS, Mohamed N, Davuluri RV, Caligiuri MA, Bloomfield CD, de la Chapelle A. Acute myeloid leukemia with complex karyotypes and abnormal chromosome 21: Amplification discloses overexpression of APP, ETS2, and ERG genes. Proc Natl Acad Sci U S A. 2004 Mar 16;101(11):3915-20 Tomlins SA, Bjartell A, Chinnaiyan AM, Jenster G, Nam RK, Rubin MA, Schalken JA. ETS gene fusions in prostate cancer: from discovery to daily clinical practice. Eur Urol. 2009 Aug;56(2):275-86 Oikawa T. ETS transcription factors: possible targets for cancer therapy. Cancer Sci. 2004 Aug;95(8):626-33 Barwick BG, Abramovitz M, Kodani M, Moreno CS, Nam R, Tang W, Bouzyk M, Seth A, Leyland-Jones B. Prostate cancer genes associated with TMPRSS2-ERG gene fusion and prognostic of biochemical recurrence in multiple cohorts. Br J Cancer. 2010 Feb 2;102(3):570-6 Owczarek CM, Portbury KJ, Hardy MP, O'Leary DA, Kudoh J, Shibuya K, Shimizu N, Kola I, Hertzog PJ. Detailed mapping of the ERG-ETS2 interval of human chromosome 21 and comparison with the region of conserved synteny on mouse chromosome 16. Gene. 2004 Jan 7;324:65-77 Boormans JL, Hermans KG, Made AC, van Leenders GJ, Wildhagen MF, Collette L, Schröder FH, Trapman J, Verhagen PC. Expression of the androgen-regulated fusion gene TMPRSS2-ERG does not predict response to endocrine treatment in hormone-naïve, node-positive prostate cancer. Eur Urol. 2010 May;57(5):830-5 Marcucci G, Baldus CD, Ruppert AS, Radmacher MD, Mrózek K, Whitman SP, Kolitz JE, Edwards CG, Vardiman JW, Powell BL, Baer MR, Moore JO, Perrotti D, Caligiuri MA, Carroll AJ, Larson RA, de la Chapelle A, Bloomfield CD. Overexpression of the ETS-related gene, ERG, predicts a worse outcome in acute myeloid leukemia with normal karyotype: a Cancer and Leukemia Group B study. J Clin Oncol. 2005 Dec 20;23(36):9234-42 Coskun E, von der Heide EK, Schlee C, Kühnl A, Gökbuget N, Hoelzer D, Hofmann WK, Thiel E, Baldus CD. The role of microRNA-196a and microRNA-196b as ERG regulators in acute myeloid leukemia and acute T-lymphoblastic leukemia. Leuk Res. 2011 Feb;35(2):208-13 Rainis L, Toki T, Pimanda JE, Rosenthal E, Machol K, Strehl S, Göttgens B, Ito E, Izraeli S. The proto-oncogene ERG in megakaryoblastic leukemias. Cancer Res. 2005 Sep 1;65(17):7596-602 De Vita S, Canzonetta C, Mulligan C, Delom F, Groet J, Baldo C, Vanes L, Dagna-Bricarelli F, Hoischen A, Veltman J, Fisher EM, Tybulewicz VL, Nizetic D. Trisomic dose of several chromosome 21 genes perturbs haematopoietic stem and progenitor cell differentiation in Down's syndrome. Oncogene. 2010 Nov 18;29(46):6102-14 Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, Varambally S, Cao X, Tchinda J, Kuefer R, Lee C, Montie JE, Shah RB, Pienta KJ, Rubin MA, Chinnaiyan AM. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005 Oct 28;310(5748):644-8 Eid MA, Attia M, Abdou S, El-Shazly SF, Elahwal L, Farrag W, Mahmoud L. BAALC and ERG expression in acute myeloid leukemia with normal karyotype: impact on prognosis. Int J Lab Hematol. 2010 Apr;32(2):197-205 van Zelm MC, van der Burg M, de Ridder D, Barendregt BH, de Haas EF, Reinders MJ, Lankester AC, Révész T, Staal FJ, van Dongen JJ. Ig gene rearrangement steps are initiated in early human precursor B cell subsets and correlate with Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Falzarano SM, Navas M, Simmerman K, Klein EA, Rubin MA, Zhou M, Magi-Galluzzi C. ERG rearrangement is present in a 551 ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Menon R, et al. subset of transition zone prostatic tumors. Mod Pathol. 2010 Nov;23(11):1499-506 prostate cancer of Caucasian, African-American and Japanese patients. Prostate. 2011 Apr;71(5):489-97 Goldstein AS, Huang J, Guo C, Garraway IP, Witte ON. Identification of a cell of origin for human prostate cancer. Science. 2010 Jul 30;329(5991):568-71 Malone VS, Dobin SM, Jones KA, Donner LR. CD99-positive large cell neuroendocrine carcinoma with rearranged EWSR1 gene in an infant: a case of prognostically favorable tumor. Virchows Arch. 2010 Sep;457(3):389-95 Gupta S, Iljin K, Sara H, Mpindi JP, Mirtti T, Vainio P, Rantala J, Alanen K, Nees M, Kallioniemi O. FZD4 as a mediator of ERG oncogene-induced WNT signaling and epithelial-tomesenchymal transition in human prostate cancer cells. Cancer Res. 2010 Sep 1;70(17):6735-45 Mao X, Yu Y, Boyd LK, Ren G, Lin D, Chaplin T, Kudahetti SC, Stankiewicz E, Xue L, Beltran L, Gupta M, Oliver RT, Lemoine NR, Berney DM, Young BD, Lu YJ. Distinct genomic alterations in prostate cancers in Chinese and Western populations suggest alternative pathways of prostate carcinogenesis. Cancer Res. 2010 Jul 1;70(13):5207-12 Haffner MC, Aryee MJ, Toubaji A, Esopi DM, Albadine R, Gurel B, Isaacs WB, Bova GS, Liu W, Xu J, Meeker AK, Netto G, De Marzo AM, Nelson WG, Yegnasubramanian S. Androgen-induced TOP2B-mediated double-strand breaks and prostate cancer gene rearrangements. Nat Genet. 2010 Aug;42(8):668-75 Ng AP, Hyland CD, Metcalf D, Carmichael CL, Loughran SJ, Di Rago L, Kile BT, Alexander WS. Trisomy of Erg is required for myeloproliferation in a mouse model of Down syndrome. Blood. 2010 May 13;115(19):3966-9 Hämäläinen M, Juvonen V, Häikiö S, Lakkala T, Johansson J, Pelliniemi TT, Salmi TT, Remes K, Kairisto V. ETS-related gene ERG expression in AML patients is significantly associated with NPM1 mutation status. Eur J Haematol. 2010 Oct;85(4):361-2 Oh SH, Park TS, Choi JR, Lee S, Cho SY, Kim SY, Kim J, Park JK, Song SA, Lee JY, Shin JH, Kim HR, Lee JN. Two childhood cases of acute leukemia with t(16;21)(p11.2;q22): second case report of infantile acute lymphoblastic leukemia with unusual type of FUS-ERG chimeric transcript. Cancer Genet Cytogenet. 2010 Jul 15;200(2):180-3 Han B, Suleman K, Wang L, Siddiqui J, Sercia L, Magi-Galluzzi C, Palanisamy N, Chinnaiyan AM, Zhou M, Shah RB. ETS gene aberrations in atypical cribriform lesions of the prostate: Implications for the distinction between intraductal carcinoma of the prostate and cribriform high-grade prostatic intraepithelial neoplasia. Am J Surg Pathol. 2010 Apr;34(4):478-85 Oram SH, Thoms JA, Pridans C, Janes ME, Kinston SJ, Anand S, Landry JR, Lock RB, Jayaraman PS, Huntly BJ, Pimanda JE, Göttgens B. A previously unrecognized promoter of LMO2 forms part of a transcriptional regulatory circuit mediating LMO2 expression in a subset of T-acute lymphoblastic leukaemia patients. Oncogene. 2010 Oct 28;29(43):5796-808 Heo SH, Choi YJ, Ryoo HM, Cho JY. Expression profiling of ETS and MMP factors in VEGF-activated endothelial cells: role of MMP-10 in VEGF-induced angiogenesis. J Cell Physiol. 2010 Sep;224(3):734-42 Park K, Tomlins SA, Mudaliar KM, Chiu YL, Esgueva R, Mehra R, Suleman K, Varambally S, Brenner JC, MacDonald T, Srivastava A, Tewari AK, Sathyanarayana U, Nagy D, Pestano G, Kunju LP, Demichelis F, Chinnaiyan AM, Rubin MA. Antibody-based detection of ERG rearrangement-positive prostate cancer. Neoplasia. 2010 Jul;12(7):590-8 Jinawath N, Morsberger L, Norris-Kirby A, Williams LM, Yonescu R, Argani P, Griffin CA, Murphy KM. Complex rearrangement of chromosomes 1, 7, 21, 22 in Ewing sarcoma. Cancer Genet Cytogenet. 2010 Aug;201(1):42-7 Perner S, Svensson MA, Hossain RR, Day JR, Groskopf J, Slaughter RC, Jarleborn AR, Hofer MD, Kuefer R, Demichelis F, Rickman DS, Rubin MA. ERG rearrangement metastasis patterns in locally advanced prostate cancer. Urology. 2010 Apr;75(4):762-7 Kunderfranco P, Mello-Grand M, Cangemi R, Pellini S, Mensah A, Albertini V, Malek A, Chiorino G, Catapano CV, Carbone GM. ETS transcription factors control transcription of EZH2 and epigenetic silencing of the tumor suppressor gene Nkx3.1 in prostate cancer. PLoS One. 2010 May 10;5(5):e10547 Pfeiffer MJ, Mulders PF, Schalken JA. An in vitro model for preclinical testing of endocrine therapy combinations for prostate cancer. Prostate. 2010 Oct 1;70(14):1524-32 Le Bras A, Samson C, Trentini M, Caetano B, Lelievre E, Mattot V, Beermann F, Soncin F. VE-statin/egfl7 expression in endothelial cells is regulated by a distal enhancer and a proximal promoter under the direct control of Erg and GATA-2. PLoS One. 2010 Aug 16;5(8):e12156 Reynolds LE, Watson AR, Baker M, Jones TA, D'Amico G, Robinson SD, Joffre C, Garrido-Urbani S, RodriguezManzaneque JC, Martino-Echarri E, Aurrand-Lions M, Sheer D, Dagna-Bricarelli F, Nizetic D, McCabe CJ, Turnell AS, Kermorgant S, Imhof BA, Adams R, Fisher EM, Tybulewicz VL, Hart IR, Hodivala-Dilke KM. Tumour angiogenesis is reduced in the Tc1 mouse model of Down's syndrome. Nature. 2010 Jun 10;465(7299):813-7 Le Deley MC, Delattre O, Schaefer KL, Burchill SA, Koehler G, Hogendoorn PC, Lion T, Poremba C, Marandet J, Ballet S, Pierron G, Brownhill SC, Nesslböck M, Ranft A, Dirksen U, Oberlin O, Lewis IJ, Craft AW, Jürgens H, Kovar H. Impact of EWS-ETS fusion type on disease progression in Ewing's sarcoma/peripheral primitive neuroectodermal tumor: prospective results from the cooperative Euro-E.W.I.N.G. 99 trial. J Clin Oncol. 2010 Apr 20;28(12):1982-8 Rice KR, Chen Y, Ali A, Whitman EJ, Blase A, Ibrahim M, Elsamanoudi S, Brassell S, Furusato B, Stingle N, Sesterhenn IA, Petrovics G, Miick S, Rittenhouse H, Groskopf J, McLeod DG, Srivastava S. Evaluation of the ETS-related gene mRNA in urine for the detection of prostate cancer. Clin Cancer Res. 2010 Mar 1;16(5):1572-6 Lee K, Chae JY, Kwak C, Ku JH, Moon KC. TMPRSS2-ERG gene fusion and clinicopathologic characteristics of Korean prostate cancer patients. Urology. 2010 Nov;76(5):1268.e7-13 Rubio-Briones J, Fernández-Serra A, Calatrava A, GarcíaCasado Z, Rubio L, Bonillo MA, Iborra I, Solsona E, LópezGuerrero JA. Clinical implications of TMPRSS2-ERG gene fusion expression in patients with prostate cancer treated with radical prostatectomy. J Urol. 2010 May;183(5):2054-61 Leinonen KA, Tolonen TT, Bracken H, Stenman UH, Tammela TL, Saramäki OR, Visakorpi T. Association of SPINK1 expression and TMPRSS2:ERG fusion with prognosis in endocrine-treated prostate cancer. Clin Cancer Res. 2010 May 15;16(10):2845-51 Scheble VJ, Braun M, Wilbertz T, Stiedl AC, Petersen K, Schilling D, Reischl M, Seitz G, Fend F, Kristiansen G, Perner S. ERG rearrangement in small cell prostatic and lung cancer. Histopathology. 2010 Jun;56(7):937-43 Magi-Galluzzi C, Tsusuki T, Elson P, Simmerman K, LaFargue C, Esgueva R, Klein E, Rubin MA, Zhou M. TMPRSS2-ERG gene fusion prevalence and class are significantly different in Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 552 ERG (v-ets erythroblastosis virus E26 oncogene like (avian)) Menon R, et al. Schulz WA, Ingenwerth M, Djuidje CE, Hader C, Rahnenführer J, Engers R. Changes in cortical cytoskeletal and extracellular matrix gene expression in prostate cancer are related to oncogenic ERG deregulation. BMC Cancer. 2010 Sep 22;10:505 the growth-promoting TMPRSS2:ERG Endocrinology. 2010 Apr;151(4):1409-17 fusion. Wei GH, Badis G, Berger MF, Kivioja T, Palin K, Enge M, Bonke M, Jolma A, Varjosalo M, Gehrke AR, Yan J, Talukder S, Turunen M, Taipale M, Stunnenberg HG, Ukkonen E, Hughes TR, Bulyk ML, Taipale J. Genome-wide analysis of ETS-family DNA-binding in vitro and in vivo. EMBO J. 2010 Jul 7;29(13):2147-60 Stott SL, Lee RJ, Nagrath S, Yu M, Miyamoto DT, Ulkus L, Inserra EJ, Ulman M, Springer S, Nakamura Z, Moore AL, Tsukrov DI, Kempner ME, Dahl DM, Wu CL, Iafrate AJ, Smith MR, Tompkins RG, Sequist LV, Toner M, Haber DA, Maheswaran S. Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer. Sci Transl Med. 2010 Mar 31;2(25):25ra23 Wilson NK, Foster SD, Wang X, Knezevic K, Schütte J, Kaimakis P, Chilarska PM, Kinston S, Ouwehand WH, Dzierzak E, Pimanda JE, de Bruijn MF, Göttgens B. Combinatorial transcriptional control in blood stem/progenitor cells: genome-wide analysis of ten major transcriptional regulators. Cell Stem Cell. 2010 Oct 8;7(4):532-44 Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E, Reva B, Antipin Y, Mitsiades N, Landers T, Dolgalev I, Major JE, Wilson M, Socci ND, Lash AE, Heguy A, Eastham JA, Scher HI, Reuter VE, Scardino PT, Sander C, Sawyers CL, Gerald WL. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010 Jul 13;18(1):11-22 Yu J, Yu J, Mani RS, Cao Q, Brenner CJ, Cao X, Wang X, Wu L, Li J, Hu M, Gong Y, Cheng H, Laxman B, Vellaichamy A, Shankar S, Li Y, Dhanasekaran SM, Morey R, Barrette T, Lonigro RJ, Tomlins SA, Varambally S, Qin ZS, Chinnaiyan AM. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell. 2010 May 18;17(5):443-54 van Doorninck JA, Ji L, Schaub B, Shimada H, Wing MR, Krailo MD, Lessnick SL, Marina N, Triche TJ, Sposto R, Womer RB, Lawlor ER. Current treatment protocols have eliminated the prognostic advantage of type 1 fusions in Ewing sarcoma: a report from the Children's Oncology Group. J Clin Oncol. 2010 Apr 20;28(12):1989-94 This article should be referenced as such: Menon R, Braun M, Perner S. ERG (v-ets erythroblastosis virus E26 oncogene like (avian)). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):547-553. Washington MN, Weigel NL. 1{alpha},25-Dihydroxyvitamin D3 inhibits growth of VCaP prostate cancer cells despite inducing Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) gene 553 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review ETV4 (ets variant 4) Yasuyoshi Miyata Department of Urology, Nagasaki University School of Medicine, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan (YM) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/ETV4ID133ch17q21.html DOI : 10.4267/2042/45992 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Localisation Identity Nuclear (Monté et al., 1994; Takahashi et al., 2005). Ubiquitinated protein is localized in the dot-like structure in the nuclear (Takahashi et al., 2005). Other names: E1A-F; E1AF; PEA3; PEAS3 HGNC (Hugo): ETV4 Location: 17q21.31 Function ETV4 is capable of regulating transcription by binding to the Ets-binding site in the promoter of its target genes. Biologically, it contributes in a number of processes including neuronal pathfinding, mammary gland development, and male sexual function (Laing et al., 2000; Ladle et al., 2002; Kurpios et al., 2003). In various malignancies, its over-expression has been observed and it was also associated with tumor progression and outcome of patients with these malignancies. As mechanism of such function, regulation of hepatocyte growth factor (HGF)-induced cell migration (Hakuma et al., 2005), HER2-mediated malignant potential (Benz et al., 1997), and other ETV4-related factors including cyclooxygenase (COX)-2 and matrix metalloproteinases (MMPs) have been reported (Higashino et al., 1995; Horiuchi et al., 2003; Shindoh et al., 2004). DNA/RNA Description The gene spans approximately 30 kb and contained 14 exons. The largest exon (901 bp) contains the end of the ETS domain, the carboxy-terminal domain and the 3'-untranslated region. The remaining exons varied from 48 bp (exon 5) to 266 bp (exon 9). Protein Description ETV4 is a member of ets-oncogene family transcription factors that were cloned by the ability to bind to enhancer motifs of the adenovirus E1A gene (Lavia et al., 2003). It is composed of 555 amino acids, which has a molecular weight ranging 61~70 kDa. GeneLoc map region. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 554 ETV4 (ets variant 4) Miyata Y In addition, ETV4 promotes cell cycle progression via upregulation of cyclin D3 transcription in breast cancer cell (MDA231 cell) (Jiang et al., 2007). In contrast, ETV4 function as negative regulator of sonic hedgehog expression (Mao et al., 2009). ETV4 expression was not detected in normal lung tissues. On the other hand, it is expressed in distal lung epithelium during lung development and in human lung cancer cells (Hiroumi et al., 2001; Liu et al., 2003). It was reported to be associated with cell invasion (Hiroumi et al., 2001) and metastasis via regulation of caveolin-1 transcription (Sloan et al., 2009) and Metrelated factors (Hakuma et al., 2005). Implicated in Breast cancer Malignant melanoma Disease Over-expression of ETV4 has been detected in human cancer cell lines (Baert et al., 1997). In animal model and human tissues, its over-expression was also found and it was associated with malignant potential including invasion and metastasis (Trimble et al., 1993; De Launoit et al., 2000; Benz et al., 1997; Bièche et al., 2004). Such ETV4-related functions are controlled via regulation of cyclin D3 transcription (Jiang et al., 2007), HER-2/Neu (Benz et al., 1997), and MMPs (Bièche et al., 2004). Disease In cell lines, ETV4 plays important roles for malignant behavior including invasion and metastasis thorough up-regulation of MT1-MMP (Hata et al., 2008). Prostate cancer Disease In human tissues, its expression in cancer cell was significantly higher than that in normal cells and it was also positively associated with pT stage. This finding was influenced with regulation of MMP-7 and MMP-9, but not of MMP-1, MMP-3, and MMP-14 (MT1MMP) (Maruta et al., 2009). Gastric cancer Disease Correlated with tumor progression via up-regulation of matrilysin in human tissues (Yamamoto et al., 2004). Colorectal cancer Disease In early stage of colorectal carcinogenesis, its overexpression plays important roles through MMPs, COX2, and iNos (Nosho et al., 2005). Lung cancer Disease Breakpoints Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 555 ETV4 (ets variant 4) Miyata Y Bièche I, Tozlu S, Girault I, Onody P, Driouch K, Vidaud M, Lidereau R. Expression of PEA3/E1AF/ETV4, an Ets-related transcription factor, in breast tumors: positive links to MMP2, NRG1 and CGB expression. Carcinogenesis. 2004 Mar;25(3):405-11 References Trimble MS, Xin JH, Guy CT, Muller WJ, Hassell JA. PEA3 is overexpressed in mouse metastatic mammary adenocarcinomas. Oncogene. 1993 Nov;8(11):3037-42 Shindoh M, Higashino F, Kohgo T. E1AF, an ets-oncogene family transcription factor. Cancer Lett. 2004 Dec 8;216(1):1-8 Monté D, Baert JL, Defossez PA, de Launoit Y, Stéhelin D. Molecular cloning and characterization of human ERM, a new member of the Ets family closely related to mouse PEA3 and ER81 transcription factors. Oncogene. 1994 May;9(5):1397406 Yamamoto H, Horiuchi S, Adachi Y, Taniguchi H, Nosho K, Min Y, Imai K. Expression of ets-related transcriptional factor E1AF is associated with tumor progression and overexpression of matrilysin in human gastric cancer. Carcinogenesis. 2004 Mar;25(3):325-32 Higashino F, Yoshida K, Noumi T, Seiki M, Fujinaga K. Etsrelated protein E1A-F can activate three different matrix metalloproteinase gene promoters. Oncogene. 1995 Apr 6;10(7):1461-3 Hakuma N, Kinoshita I, Shimizu Y, Yamazaki Nishimura M, Dosaka-Akita H. E1AF/PEA3 Rho/Rho-associated kinase pathway to malignancy potential of non-small-cell lung Cancer Res. 2005 Dec 1;65(23):10776-82 Baert JL, Monté D, Musgrove EA, Albagli O, Sutherland RL, de Launoit Y. Expression of the PEA3 group of ETS-related transcription factors in human breast-cancer cells. Int J Cancer. 1997 Mar 4;70(5):590-7 Nosho K, Yoshida M, Yamamoto H, Taniguchi H, Adachi Y, Mikami M, Hinoda Y, Imai K. Association of Ets-related transcriptional factor E1AF expression with overexpression of matrix metalloproteinases, COX-2 and iNOS in the early stage of colorectal carcinogenesis. Carcinogenesis. 2005 May;26(5):892-9 Benz CC, O'Hagan RC, Richter B, Scott GK, Chang CH, Xiong X, Chew K, Ljung BM, Edgerton S, Thor A, Hassell JA. HER2/Neu and the Ets transcription activator PEA3 are coordinately upregulated in human breast cancer. Oncogene. 1997 Sep 25;15(13):1513-25 Takahashi A, Higashino F, Aoyagi M, Yoshida K, Itoh M, Kobayashi M, Totsuka Y, Kohgo T, Shindoh M. E1AF degradation by a ubiquitin-proteasome pathway. Biochem Biophys Res Commun. 2005 Feb 11;327(2):575-80 de Launoit Y, Chotteau-Lelievre A, Beaudoin C, Coutte L, Netzer S, Brenner C, Huvent I, Baert JL. The PEA3 group of ETS-related transcription factors. Role in breast cancer metastasis. Adv Exp Med Biol. 2000;480:107-16 Jiang J, Wei Y, Liu D, Zhou J, Shen J, Chen X, Zhang S, Kong X, Gu J. E1AF promotes breast cancer cell cycle progression via upregulation of Cyclin D3 transcription. Biochem Biophys Res Commun. 2007 Jun 22;358(1):53-8 Laing MA, Coonrod S, Hinton BT, Downie JW, Tozer R, Rudnicki MA, Hassell JA. Male sexual dysfunction in mice bearing targeted mutant alleles of the PEA3 ets gene. Mol Cell Biol. 2000 Dec;20(24):9337-45 Hata H, Kitamura T, Higashino F, Hida K, Yoshida K, Ohiro Y, Totsuka Y, Kitagawa Y, Shindoh M. Expression of E1AF, an ets-oncogene transcription factor, highly correlates with malignant phenotype of malignant melanoma through upregulation of the membrane-type-1 matrix metalloproteinase gene. Oncol Rep. 2008 May;19(5):1093-8 Hiroumi H, Dosaka-Akita H, Yoshida K, Shindoh M, Ohbuchi T, Fujinaga K, Nishimura M. Expression of E1AF/PEA3, an Etsrelated transcription factor in human non-small-cell lung cancers: its relevance in cell motility and invasion. Int J Cancer. 2001 Sep;93(6):786-91 Ladle DR, Frank E. The role of the ETS gene PEA3 in the development of motor and sensory neurons. Physiol Behav. 2002 Dec;77(4-5):571-6 Mao J, McGlinn E, Huang P, Tabin CJ, McMahon AP. Fgfdependent Etv4/5 activity is required for posterior restriction of Sonic Hedgehog and promoting outgrowth of the vertebrate limb. Dev Cell. 2009 Apr;16(4):600-6 Horiuchi S, Yamamoto H, Min Y, Adachi Y, Itoh F, Imai K. Association of ets-related transcriptional factor E1AF expression with tumour progression and overexpression of MMP-1 and matrilysin in human colorectal cancer. J Pathol. 2003 Aug;200(5):568-76 Maruta S, Sakai H, Kanda S, Hayashi T, Kanetake H, Miyata Y. E1AF expression is associated with extra-prostatic growth and matrix metalloproteinase-7 expression in prostate cancer. APMIS. 2009 Nov;117(11):791-6 Kurpios NA, Sabolic NA, Shepherd TG, Fidalgo GM, Hassell JA. Function of PEA3 Ets transcription factors in mammary gland development and oncogenesis. J Mammary Gland Biol Neoplasia. 2003 Apr;8(2):177-90 Sloan KA, Marquez HA, Li J, Cao Y, Hinds A, O'Hara CJ, Kathuria S, Ramirez MI, Williams MC, Kathuria H. Increased PEA3/E1AF and decreased Net/Elk-3, both ETS proteins, characterize human NSCLC progression and regulate caveolin-1 transcription in Calu-1 and NCI-H23 NSCLC cell lines. Carcinogenesis. 2009 Aug;30(8):1433-42 Lavia P, Mileo AM, Giordano A, Paggi MG. Emerging roles of DNA tumor viruses in cell proliferation: new insights into genomic instability. Oncogene. 2003 Sep 29;22(42):6508-16 This article should be referenced as such: Liu Y, Jiang H, Crawford HC, Hogan BL. Role for ETS domain transcription factors Pea3/Erm in mouse lung development. Dev Biol. 2003 Sep 1;261(1):10-24 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) K, Yoshida K, activates the increase the cancer cells. Miyata Y. ETV4 (ets variant 4). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):554-556. 556 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review GPC5 (glypican 5) Khin Thway, Joanna Selfe, Janet Shipley Molecular Cytogenetics, Section of Molecular Carcinogenesis, the Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, United Kingdom (KT, JS, JS) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/GPC5ID45705ch13q31.html DOI: 10.4267/2042/45993 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Protein HGNC (Hugo): GPC5 Location: 13q31.3 Local order: Centromere - MIR17HG - GPC5 - GPC6 - DCT - TGDS - GPR180 - SOX21 - telomere. Description 572 amino acids; 64 kDa protein (core protein). GPC5 is a heparan sulfate proteoglycan (HSPG), that is bound to the cell surface by a glycosyl-phosphatidylinositol (GPI) anchor. DNA/RNA Expression GPC5 is expressed mainly in fetal tissues, including brain, lung and liver. In the adult, expression is primarily in brain tissue. Note The gene spans 1.47 Mb of DNA, comprising 8 exons. Transcription Localisation 2.904 kb mRNA. 1718 bp open reading frame. Attached to the cell membrane by a GPI anchor. Schematic of glypican protein structure at the cell surface. The protein is held in the plasma membrane by a GPI anchor at the carboxyl terminus. Numerous glycosoaminoglycan (GAG) attachment sites close to the membrane surface allow heparin and chondroitin sulphate chains to be attached to the core protein (shown in green). The amino terminal end of the protein is a globular structure held together by a conserved set of cysteine residues forming disulphide bridges. (Picture reproduced from Filmus and Selleck, 2001). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 557 GPC5 (glypican 5) Thway K, et al. well as gain of GPC5 copies in both alveolar and embryonal rhabdomyosarcoma (Gordon et al., 2000). GPC5 is overexpressed in the majority of rhabdomyosarcomas compared with normal skeletal muscle and has been shown to modulate responses to FGF2 in rhabdomyosarcoma cells (Williamson et al., 2007). GPC5 may also potentiate hedgehog signalling in these cells as it can bind to both Hedgehog and the Patched receptor (Li et al., 2010a). A recent genome wide association study has linked polymorphisms in GPC5 to risk of lung cancer in never-smokers (Li et al., 2010b). The high-risk allele was coincident with lower expression of GPC5, suggesting that the role of GPC5 is likely to be tumour type-specific in an analogous manner to GPC3, the closest family member to GPC5. Function The precise functions of GPC5 have yet to be fully established. HSPGs are common constituents of cell surfaces and the extracellular matrix (ECM), with essential functions in cell growth and development (Burgess and Macaig, 1989; Andres et al., 1992). Glypicans appear to be expressed predominantly during development, with expression levels changing in a stage- and tissue-specific manner, suggesting their involvement in morphogenesis (Sing and Filmus, 2002). As they can bind numerous ligands and be associated with a variety of receptors, they act as coreceptors for a number of heparin-binding growth factors, modulating their activity. The heparan sulfate modifications of glypicans can mediate interactions with growth factors or ECM proteins, but ligands and ECM proteins can also bind through motifs in the core proteins (Mythreye and Blobe, 2009). Glypicans can be secreted from the cell surface, such soluble forms can also bind growth factors. Evidence to date suggests that glypicans can regulate Wnt, hedgehog, fibroblast growth factor and bone morphogenetic protein pathways. The effect on these pathways may be stimulatory or inhibitory depending on cellular context (Gallet et al., 2008; Capurro et al., 2008; Kreuger et al., 2004; Yan and Lin, 2007; Grisaru et al., 2001; Yan et al., 2010). GPC5 expression has been shown in the developing central nervous system, limbs and kidneys of mice, and its expression in mammalian fetal tissues suggests roles in growth and differentiation during development (Veugelers et al., 1997; Saunders et al., 1997; Luxardi et al., 2007). Its almost exclusive expression in adult brain tissue suggests a possible role in controlling neurotropic factors and maintaining neural function. Developmental disorders Note Studies on the role of GPC5 in disease are still relatively limited. In humans, deletions of the 13q31-32 region are associated with the 13q deletion syndrome, a developmental disorder with a wide phenotypic spectrum including mental and growth retardation, congenital defects and craniofacial dysmorphy, and GPC5 is suggested as a candidate gene for digital malformations in this syndrome (Quelin et al., 2009). Correspondingly, GPC5 is also a candidate gene for postaxial polydactyly type A2, which is associated with duplication of 13q31-32 (van der Zwaag et al., 2010). Multiple sclerosis Note Several genome wide association studies have identified GPC5 as having a potential role in Multiple Sclerosis (MS) (Baranzini et al., 2009; Lorentzen et al., 2010). Several different GPC5 polymorphisms were also highlighted in an independent study designed to determine which genes are associated with efficacy of interferon beta therapy in MS (Byun et al., 2008), this finding has subsequently been confirmed in a separate study (Cenit et al., 2009). HSPGs are found in dense networks in active MS plaques, where they may sequester pro-inflammatory cytokines. Homology GPC5 is a member of the glypican family of HSPGs, of which six members (GPC1, GPC2, GPC3, GPC4, GPC5, GPC6) have been identified in mammals. GPC3 is the most homologous member to GPC5 in humans. There is approximately 20-60% sequence homology between family members, including conservation of a pattern of 14 cysteine residues. Homolog glypican-like genes are also present in Drosophila (dally and dallylike). References Burgess WH, Maciag T. The heparin-binding (fibroblast) growth factor family of proteins. Annu Rev Biochem. 1989;58:575-606 Implicated in Andres JL, DeFalcis D, Noda M, Massagué J. Binding of two growth factor families to separate domains of the proteoglycan betaglycan. J Biol Chem. 1992 Mar 25;267(9):5927-30 Tumourigenesis Note Amplification of 13q31-32 has been shown in poor prognosis liposarcomas, breast cancers and neurologic tumours (Reardon et al., 2000; Ojopi et al., 2001; Ullmann et al., 2001; Schmidt et al., 2005). Amplification of 13q31-32 has also been shown in approximately 20% of alveolar rhabdomyosarcoma, as Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Saunders S, Paine-Saunders S, Lander AD. Expression of the cell surface proteoglycan glypican-5 is developmentally regulated in kidney, limb, and brain. Dev Biol. 1997 Oct 1;190(1):78-93 Veugelers M, Vermeesch J, Reekmans G, Steinfeld R, Marynen P, David G. Characterization of glypican-5 and chromosomal localization of human GPC5, a new member of the glypican gene family. Genomics. 1997 Feb 15;40(1):24-30 558 GPC5 (glypican 5) Thway K, et al. Gordon AT, Brinkschmidt C, Anderson J, Coleman N, Dockhorn-Dworniczak B, Pritchard-Jones K, Shipley J. A novel and consistent amplicon at 13q31 associated with alveolar rhabdomyosarcoma. Genes Chromosomes Cancer. 2000 Jun;28(2):220-6 signaling and wingless May;14(5):712-25 transcytosis. Dev Cell. 2008 Filmus J, Selleck SB. Glypicans: proteoglycans with a surprise. J Clin Invest. 2001 Aug;108(4):497-501 Baranzini SE, Wang J, Gibson RA, Galwey N, Naegelin Y, Barkhof F, Radue EW, Lindberg RL, Uitdehaag BM, Johnson MR, Angelakopoulou A, Hall L, Richardson JC, Prinjha RK, Gass A, Geurts JJ, Kragt J, Sombekke M, Vrenken H, Qualley P, Lincoln RR, Gomez R, Caillier SJ, George MF, Mousavi H, Guerrero R, Okuda DT, Cree BA, Green AJ, Waubant E, Goodin DS, Pelletier D, Matthews PM, Hauser SL, Kappos L, Polman CH, Oksenberg JR. Genome-wide association analysis of susceptibility and clinical phenotype in multiple sclerosis. Hum Mol Genet. 2009 Feb 15;18(4):767-78 Grisaru S, Cano-Gauci D, Tee J, Filmus J, Rosenblum ND. Glypican-3 modulates BMP- and FGF-mediated effects during renal branching morphogenesis. Dev Biol. 2001 Mar 1;231(1):31-46 Cénit MD, Blanco-Kelly F, de las Heras V, Bartolomé M, de la Concha EG, Urcelay E, Arroyo R, Martínez A. Glypican 5 is an interferon-beta response gene: a replication study. Mult Scler. 2009 Aug;15(8):913-7 Ojopi EP, Rogatto SR, Caldeira JR, Barbiéri-Neto J, Squire JA. Comparative genomic hybridization detects novel amplifications in fibroadenomas of the breast. Genes Chromosomes Cancer. 2001 Jan;30(1):25-31 Mythreye K, Blobe GC. Proteoglycan signaling co-receptors: roles in cell adhesion, migration and invasion. Cell Signal. 2009 Nov;21(11):1548-58 Reardon DA, Jenkins JJ, Sublett JE, Burger PC, Kun LK. Multiple genomic alterations including N-myc amplification in a primary large cell medulloblastoma. Pediatr Neurosurg. 2000 Apr;32(4):187-91 Quélin C, Bendavid C, Dubourg C, de la Rochebrochard C, Lucas J, Henry C, Jaillard S, Loget P, Loeuillet L, Lacombe D, Rival JM, David V, Odent S, Pasquier L. Twelve new patients with 13q deletion syndrome: genotype-phenotype analyses in progress. Eur J Med Genet. 2009 Jan-Feb;52(1):41-6 Ullmann R, Petzmann S, Sharma A, Cagle PT, Popper HH. Chromosomal aberrations in a series of large-cell neuroendocrine carcinomas: unexpected divergence from small-cell carcinoma of the lung. Hum Pathol. 2001 Oct;32(10):1059-63 Li FE, Shi W, Capurro M, Filmus J.. Glypican-5 stimulates rhabdomyosarcoma cell proliferation by activating hedgehog signaling. Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; 2010a. Abstract no. 3191. Song HH, Filmus J. The role of glypicans in mammalian development. Biochim Biophys Acta. 2002 Dec 19;1573(3):241-6 Kreuger J, Perez L, Giraldez AJ, Cohen SM. Opposing activities of Dally-like glypican at high and low levels of Wingless morphogen activity. Dev Cell. 2004 Oct;7(4):503-12 Li Y, Sheu CC, Ye Y, de Andrade M, Wang L, Chang SC, Aubry MC, Aakre JA, Allen MS, Chen F, Cunningham JM, Deschamps C, Jiang R, Lin J, Marks RS, Pankratz VS, Su L, Li Y, Sun Z, Tang H, Vasmatzis G, Harris CC, Spitz MR, Jen J, Wang R, Zhang ZF, Christiani DC, Wu X, Yang P.. Genetic variants and risk of lung cancer in never smokers: a genomewide association study. Lancet Oncol. 2010b Apr;11(4):321-30. Epub 2010 Mar 19. Schmidt H, Bartel F, Kappler M, Würl P, Lange H, Bache M, Holzhausen HJ, Taubert H. Gains of 13q are correlated with a poor prognosis in liposarcoma. Mod Pathol. 2005 May;18(5):638-44 Luxardi G, Galli A, Forlani S, Lawson K, Maina F, Dono R. Glypicans are differentially expressed during patterning and neurogenesis of early mouse brain. Biochem Biophys Res Commun. 2007 Jan 5;352(1):55-60 Lorentzen AR, Melum E, Ellinghaus E, Smestad C, Mero IL, Aarseth JH, Myhr KM, Celius EG, Lie BA, Karlsen TH, Franke A, Harbo HF.. Association to the Glypican-5 gene in multiple sclerosis. J Neuroimmunol. 2010 Sep 14;226(1-2):194-7. Epub 2010 Aug 6. Williamson D, Selfe J, Gordon T, Lu YJ, Pritchard-Jones K, Murai K, Jones P, Workman P, Shipley J. Role for amplification and expression of glypican-5 in rhabdomyosarcoma. Cancer Res. 2007 Jan 1;67(1):57-65 van der Zwaag PA, Dijkhuizen T, Gerssen-Schoorl KB, Colijn AW, Broens PM, Flapper BC, van Ravenswaaij-Arts CM.. An interstitial duplication of chromosome 13q31.3q32.1 further delineates the critical region for postaxial polydactyly type A2. Eur J Med Genet. 2010 Jan-Feb;53(1):45-9. Epub 2009 Nov 23. Yan D, Lin X. Drosophila glypican Dally-like acts in FGFreceiving cells to modulate FGF signaling during tracheal morphogenesis. Dev Biol. 2007 Dec 1;312(1):203-16 Byun E, Caillier SJ, Montalban X, Villoslada P, Fernández O, Brassat D, Comabella M, Wang J, Barcellos LF, Baranzini SE, Oksenberg JR. Genome-wide pharmacogenomic analysis of the response to interferon beta therapy in multiple sclerosis. Arch Neurol. 2008 Mar;65(3):337-44 Yan D, Wu Y, Yang Y, Belenkaya TY, Tang X, Lin X.. The cellsurface proteins Dally-like and Ihog differentially regulate Hedgehog signaling strength and range during development. Development. 2010 Jun;137(12):2033-44. Capurro MI, Xu P, Shi W, Li F, Jia A, Filmus J. Glypican-3 inhibits Hedgehog signaling during development by competing with patched for Hedgehog binding. Dev Cell. 2008 May;14(5):700-11 This article should be referenced as such: Thway K, Selfe J, Shipley J. GPC5 (glypican 5). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):557-559. Gallet A, Staccini-Lavenant L, Thérond PP. Cellular trafficking of the glypican Dally-like is required for full-strength Hedgehog Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 559 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review GSDMA (gasdermin A) Norihisa Saeki, Hiroki Sasaki Genetics Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan (NS, HS) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/GSDMAID45650ch17q21.html DOI: 10.4267/2042/45994 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Protein Other names: FLJ39120; GSDM; GSDM1; MGC129596 HGNC (Hugo): GSDMA Location: 17q21.1 Local order: Telomeric to ORMDL3 and GSDMB genes; centromeric to PSMD3 gene. Note GSDMA is the first member of Gasdermin family genes which, with Gadermin-related genes, DFNA5 and DFNB59, form Gasdermin superfamily. DNA/RNA Genomic organization of the GSDMA gene. Description GSDMA is involved in TGF-beta signaling which regulates apoptosis induction in pit cells of the gastric epithelium. Signaling from TGF-beta receptor up-regulates LMO1, a transcription factor. LMO1 binds to the promoter of GSDMA gene and enhances its expression, that results in the apoptosis induction in the pit cells. 12 exons, spans approximately 13 kb of genomic DNA in the centromere-to-telomere orientation. The translation initiation codon is located to exon 2, and the stop codon to exon 12. Transcription Description mRNA of approximately 1.5 kb. The GSDMA gene encodes a 445 amino acid protein with estimated molecular weight of 49377.95 Da. The Gasdermin family proteins have 9 conserved motifs but no known functional motif. Pseudogene Not reported. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 560 GSDMA (gasdermin A) Saeki N, Sasaki H Expression Esophageal cancer GSDMA protein is expressed in pit cells of the gastric epithelium, where it is involved in maintenance of homeostasis by its apoptosis induction ability under TGF-beta signaling. Its expression was also observed in epithelial cells of the esophagus, skin and mammary gland. Note GSDMA gene is frequently silenced in esophageal squamous cell carcinoma (41 in 42 cases examined). References Saeki N, Kuwahara Y, Sasaki H, Satoh H, Shiroishi T. Gasdermin (Gsdm) localizing to mouse Chromosome 11 is predominantly expressed in upper gastrointestinal tract but significantly suppressed in human gastric cancer cells. Mamm Genome. 2000 Sep;11(9):718-24 Localisation Cytoplasm. Function Lunny DP, Weed E, Nolan PM, Marquardt A, Augustin M, Porter RM. Mutations in gasdermin 3 cause aberrant differentiation of the hair follicle and sebaceous gland. J Invest Dermatol. 2005 Mar;124(3):615-21 Apoptosis induction, but detail is unknown. Homology Human genome possesses its three paralogues, GSDMB, GSDMC and GSDMD. Both N- and Cterminal amino acids are conserved among them. Saeki N, Kim DH, Usui T, Aoyagi K, Tatsuta T, Aoki K, Yanagihara K, Tamura M, Mizushima H, Sakamoto H, Ogawa K, Ohki M, Shiroishi T, Yoshida T, Sasaki H. GASDERMIN, suppressed frequently in gastric cancer, is a target of LMO1 in TGF-beta-dependent apoptotic signalling. Oncogene. 2007 Oct 4;26(45):6488-98 Mutations Germinal Tamura M, Tanaka S, Fujii T, Aoki A, Komiyama H, Ezawa K, Sumiyama K, Sagai T, Shiroishi T. Members of a novel gene family, Gsdm, are expressed exclusively in the epithelium of the skin and gastrointestinal tract in a highly tissue-specific manner. Genomics. 2007 May;89(5):618-29 Not reported. Somatic Not reported. Saeki N, Usui T, Aoyagi K, Kim DH, Sato M, Mabuchi T, Yanagihara K, Ogawa K, Sakamoto H, Yoshida T, Sasaki H. Distinctive expression and function of four GSDM family genes (GSDMA-D) in normal and malignant upper gastrointestinal epithelium. Genes Chromosomes Cancer. 2009 Mar;48(3):261-71 Implicated in Gastric cancer Note GSDMA gene is frequently silenced in gastric adenocarcinoma (16 in 18 cases examined including both diffuse and intestinal types), whose relation to prognosis is unknown. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) This article should be referenced as such: Saeki N, Sasaki H. GSDMA (gasdermin A). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):560-561. 561 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) Theoni Trangas, Panayotis Ioannidis Department of Biological Applications and Technologies University of Ioannina, Ioannina, Greece (TT), National Reference Center for Mycobacteria, Sotiria Hospital, Athens, Greece (PI) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/IGF2BP1ID40969ch17q21.html DOI: 10.4267/2042/45995 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology upstream (promoter) region contains binding sites for the following transcription factors: delta CREB, CREB, NF-kappaB1, NF-kappaB, AP-1, HNF4 alpha2, FOXO1a, MZF-1, Max and c-Myc. Beta-catenin/TCF4 binding and activation of transcription has been experimentally confirmed (Gu et al., 2008). Identity Other names: CRD-BP; CRDBP; IMP-1; IMP1; VICKZ1; ZBP1 HGNC (Hugo): IGF2BP1 Location: 17q21.32 Local order: The IGF2BP1 gene is located on the plus strand on chromosome 17, at 17q21.32. This gene starts at 47074774 and ends at 47133507 bp from pter, encompasses 58734 bp and lies 5' of the gene B4GALNT2, encoding beta-1,4-N-acetylgalactosaminyl transferase 2. Note The IGF2BP1 gene encodes a member of the IGF-II mRNA-binding protein (IMP) family (RRM IMP/VICKZ family). Transcription Two protein coding transcripts exist resulting from alternative splicing: Transcript variant 1 (NM_006546). The length of this transcript is 8769 nt and encompasses all 15 exons (exon 1: 509 bp, exon 2: 60 bp, exon 3: 48 bp, exon 4: 51 bp, exon 5: 63 bp, exon 6: 281 bp, exon 7: 134 bp, exon 8: 122 bp, exon 9: 135 bp, exon 10: 122 bp, exon 11: 119 bp, exon 12: 74 bp, exon 13: 131 bp, exon14: 113 bp, exon 15: 6973 bp). Several alternative 3' ends (polyadenylation sites) exist at exon 15 3'-UTR (marked by flags in the figure above). Translation starts at +335 and ends at +2068. Transcript variant 2 (NM_001160423.1). It encompasses 8352 bp and lacks two consecutive inframe exons (6 and 7). Other spliced variants have been reported without corresponding protein product recorded. DNA/RNA Description There are 4 probable alternative promoters driving transcription of IGF2BP1 and two of them have been experimentally confirmed (Gu et al., 2008). The Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 562 IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) Trangas T, Ioannidis P (spermatogonia), in semen (Hammer et al., 2005) and in intestinal crypts (Nielsen et al., 1999; Dimitriadis et al., 2007). It is expressed de novo in kidney, prostate, trachea, testis, ovarian and lung cancer, melanoma, mesenchymal and brain tumors. At protein level, it is expressed in testicular, lung and colon cancer. Protein Description The IGF2BP1 protein translated from the transcript variant 1 consists of 577 aa (63,48 kD) and has 2 highly conserved RRM motifs belonging to the RNA recognition motif (RRM) superfamily and 4 KH domains (NP_006537.3). The third and fourth KH domains constitute both the protein dimerization motif and the RNA binding domain. The four KH domains promote granule formation and stress granule targeting (Stöhr et al., 2006). Two nuclear export signals (NES) exist within the second and fourth KH domains (Nielsen et al., 2003). The KH domains have been implicated in the suppression of HIV-1 infectivity (Zhou et al., 2008). Phosphorylation sites are marked (Bennetzen et al., 2010; Dephoure et al., 2008). Phosphorylation of Tyrosine 396 prevents RNA binding and translation inhibition of beta-actin mRNA (Hüttelmaier et al., 2005). The IGF2BP1 protein translated from transcript 2 variant is predicted to consist of 438 aa (48.597 kD) and contain 2 RRM and 3 KH domains (NP_001153895.1). Localisation IGF2BP1 has been detected in the nucleus, cytoplasm, cytoplasmic mRNPs, granules (Nielsen et al., 2002; Nielsen et al., 2003). In stress granules IGF2BP1 co localizes with G3BP1 and TIAL1 (Stöhr et al., 2006). It has also been detected in lamellipodia (Yaniv et al., 2003), growth cones and the leading edge of developing axons (Eom et al., 2003). Function mRNA translation: IGF2BP1 regulates translation by binding the 5'-UTR of the mRNA of certain genes, including insulin-like growth factor 2 (Nielsen et al., 1999), and beta actin (Hüttelmaier et al., 2005). It has been identified in a HCV IRES-mediated translation complex along with EIF3C and RPS3, enhancing translation of the Hepatitis C virus (HCV) RNAreplicon via the internal ribosome entry site (IRES), without affecting 5'cap-dependent translation (Weinlich et al., 2009). IGF2BP1 binds the adenine-rich autoregulatory sequence (ARS) of the 5'-UTR of the PABPC1 mRNA in collaboration with CSDE1 and PABPC1 proteins and causes translational repression (Patel and Bag, 2006; Patel et al., 2005). Expression IGF2BP1 is widely expressed in fetal tissues (liver, lung, kidney, thymus, etc), placenta and CD34+ cord blood cells (Nielsen et al., 1999; Ioannidis et al., 2001; Ioannidis et al., 2005). Postnatally it is expressed in ovary (oocytes and granulosa cells), in testis IGF2BP1 protein translated from transcript 1 variant. IGF2BP1 protein translated from transcript 2 variant. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 563 IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) Trangas T, Ioannidis P mRNA stabilization: IGF2BP1 binds to the coding region mRNA stability determinant (CRD) of c-myc mRNA and protects it from endonucleolytic cleavage (Doyle et al.,1998; Lemm and Ross, 2002). It protects MDR-1 mRNAs from endonucleolytic cleavage by binding to a coding region element (Sparanese and Lee, 2007). Also binds to the coding region of betaTrCP1 mRNA and stabilizes it by disrupting miRNAdependent interaction with AGO2 (Noubissi et al., 2006; Elcheva et al., 2009). Binds and stabilizes GLI1 mRNA causing an elevation of GLI1 expression and transcriptional activity (Noubissi et al., 2009). IGF2BP1 binds to multiple elements in the 3'-UTR of the CD44 mRNAs and stabilizes this mRNA (Vikesaa et al., 2006). Binds to the 3'-UTR of Micropthalmia associated transcription factor mRNA and prevents the binding of miR-340 to its target sites, resulting in stabilization of the transcript, elevated expression and activity of this transcription factor (Goswami et al., 2010). mRNA transportation: IGF2BP1 binds to the fourth and fifth exons of the oncofetal H19 RNA (Runge et al., 2000) and with ELAVL4 and G3BP to 3'-UTR of the neuron-specific TAU mRNA (Atlas et al., 2004; Atlas et al., 2007) and regulates their localization. In collaboration with IGF2BP2, IGF2BP1 binds to the conserved 54-nucleotide element in the 3'-UTR of the beta actin mRNA, known as the 'zip code'. IGF2BP1 promotes the localization of the beta-actin mRNA to dendrites (Eom et al., 2003). IGF2BP1 may act as a regulator of mRNA transport to activated synapses in response to synaptic activity. Protein binding: IGF2BP1 interacts through the third and fourth KH domains with PABPC1 in a RNAindependent manner (Patel and Bag, 2006) and can form homo- and heterodimers with IGF2BP2 or IGF2BP3 (Nielsen et al., 2004). It interacts with fragile X metal retardation protein isoform 18 (Rackham and Brown, 2004). It interacts with DHX9, ELAVL2, HNRNPA2B1, HNRNPC, HNRNPH1, HNRNPU, IGF2BP2, IGF2BP3, ILF2 and YBX1 (Weindensdorfer et al., 2009). IGF2BP1 was identified in a mRNP granule complex, with hnRNP A1, hnRNP A2/B1, hnRNP D, hnRNP L, hnRNP Q, hnRNP R, hnRNP U, YB1/major core protein, interleukin enhancer-binding factor 2 and 3, PABP1, PABP2, PABP4, nucleolin, RNA helicase A, a series of 40 S ribosomal proteins, and the nuclear cap-binding protein CBP80 (Jønson et al., 2007). IGF2BP1 associates with HIV-1 particles. It interacts (via KH3 and KH4 domains) with HIV-1 GAG protein and diminishes viral RNA packaging, thwarts GAG processing to the cellular membranes, and impedes HIV-1 assembly (Zhou et al., 2008). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Homology The identity of human IGF2BP1 over an aligned region in UniGene is as follows: - Pan troglodytes: 99.83% - Canis lupas familiaris: 90.93% - Bos taurus: 91.05% - Mus musculus: 89.43% - Rattus norvegicus: 89.43% Implicated in Lung cancer Disease IGF2BP1 is expressed in lung cancer and its expression correlates with adverse histological and clinical features and is an indicator of poor prognosis. Suppression of its expression with siRNA suppresses growth of NSCLC cells in vitro (Ioannidis et al., 2004; Kato et al., 2007). Ovarian cancer Disease Increased expression of IGF2BP1 mRNA is associated with an advanced clinical stage and poor prognosis in patients with ovarian cancer (Köbel et al., 2007). Testicular cancer Disease Detected in testicular carcinomas even in early stage carcinoma in situ (Hammer et al., 2005). Melanoma Disease IGF2BP1 is highly expressed in primary human malignant melanomas and melanoma cell line (Elcheva et al., 2008). Breast cancer Disease The IGF2BP1 gene is amplified in breast cancer (Doyle et al., 2000). Significant associations are detected between IGF2BP1 expression and the absence of estrogen receptors. IGF2BP1 collaborates with c-myc amplification to render tumours more aggressive (Ioannidis et al., 2003). Tissue specific induced expression in transgenic mice promotes tumor formation (Tessier et al., 2004). Colon cancer Disease IGF2BP1 is scarce or absent from normal colon but is over expressed in colorectal cancer. IGF2BP1 positive tumours associate with metastasis/recurrence and shorter survival (Ross et al., 2001; Dimitriadis et al., 2007). 564 IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) Trangas T, Ioannidis P Yaniv K, Fainsod A, Kalcheim C, Yisraeli JK. The RNA-binding protein Vg1 RBP is required for cell migration during early neural development. Development. 2003 Dec;130(23):5649-61 Hepatocellular carcinoma Disease IGF2BP1 is detected as an autoantigen hepatocellular carcinoma (Himoto et al., 2005). in Atlas R, Behar L, Elliott E, Ginzburg I. The insulin-like growth factor mRNA binding-protein IMP-1 and the Ras-regulatory protein G3BP associate with tau mRNA and HuD protein in differentiated P19 neuronal cells. J Neurochem. 2004 May;89(3):613-26 Oncogenesis Note The oncogenic action of IGF2BP1 is effected through the stabilization of the mRNA of oncogenes such as cmyc, betaTrCP1, Gli and upregulation of their expression. IGF2BP1 expression may promote metastasis by shuttling requisite RNAs to the lamellipodia of migrating cells (Vikesaa et al., 2006; Vainer et al., 2008). Ioannidis P, Kottaridi C, Dimitriadis E, Courtis N, Mahaira L, Talieri M, Giannopoulos A, Iliadis K, Papaioannou D, Nasioulas G, Trangas T. Expression of the RNA-binding protein CRD-BP in brain and non-small cell lung tumors. Cancer Lett. 2004 Jun 25;209(2):245-50 Nielsen J, Kristensen MA, Willemoës M, Nielsen FC, Christiansen J. Sequential dimerization of human zipcodebinding protein IMP1 on RNA: a cooperative mechanism providing RNP stability. Nucleic Acids Res. 2004;32(14):436876 References Rackham O, Brown CM. Visualization of RNA-protein interactions in living cells: FMRP and IMP1 interact on mRNAs. EMBO J. 2004 Aug 18;23(16):3346-55 Doyle GA, Betz NA, Leeds PF, Fleisig AJ, Prokipcak RD, Ross J. The c-myc coding region determinant-binding protein: a member of a family of KH domain RNA-binding proteins. Nucleic Acids Res. 1998 Nov 15;26(22):5036-44 Tessier CR, Doyle GA, Clark BA, Pitot HC, Ross J. Mammary tumor induction in transgenic mice expressing an RNA-binding protein. Cancer Res. 2004 Jan 1;64(1):209-14 Nielsen J, Christiansen J, Lykke-Andersen J, Johnsen AH, Wewer UM, Nielsen FC. A family of insulin-like growth factor II mRNA-binding proteins represses translation in late development. Mol Cell Biol. 1999 Feb;19(2):1262-70 Hammer NA, Hansen TO, Byskov AG, Rajpert-De Meyts E, Grøndahl ML, Bredkjaer HE, Wewer UM, Christiansen J, Nielsen FC. Expression of IGF-II mRNA-binding proteins (IMPs) in gonads and testicular cancer. Reproduction. 2005 Aug;130(2):203-12 Doyle GA, Bourdeau-Heller JM, Coulthard S, Meisner LF, Ross J. Amplification in human breast cancer of a gene encoding a c-myc mRNA-binding protein. Cancer Res. 2000 Jun 1;60(11):2756-9 Himoto T, Kuriyama S, Zhang JY, Chan EK, Nishioka M, Tan EM. Significance of autoantibodies against insulin-like growth factor II mRNA-binding proteins in patients with hepatocellular carcinoma. Int J Oncol. 2005 Feb;26(2):311-7 Runge S, Nielsen FC, Nielsen J, Lykke-Andersen J, Wewer UM, Christiansen J. H19 RNA binds four molecules of insulinlike growth factor II mRNA-binding protein. J Biol Chem. 2000 Sep 22;275(38):29562-9 Hüttelmaier S, Zenklusen D, Lederer M, Dictenberg J, Lorenz M, Meng X, Bassell GJ, Condeelis J, Singer RH. Spatial regulation of beta-actin translation by Src-dependent phosphorylation of ZBP1. Nature. 2005 Nov 24;438(7067):5125 Ioannidis P, Trangas T, Dimitriadis E, Samiotaki M, Kyriazoglou I, Tsiapalis CM, Kittas C, Agnantis N, Nielsen FC, Nielsen J, Christiansen J, Pandis N. C-MYC and IGF-II mRNAbinding protein (CRD-BP/IMP-1) in benign and malignant mesenchymal tumors. Int J Cancer. 2001 Nov;94(4):480-4 Ioannidis P, Mahaira LG, Perez SA, Gritzapis AD, Sotiropoulou PA, Kavalakis GJ, Antsaklis AI, Baxevanis CN, Papamichail M. CRD-BP/IMP1 expression characterizes cord blood CD34+ stem cells and affects c-myc and IGF-II expression in MCF-7 cancer cells. J Biol Chem. 2005 May 20;280(20):20086-93 Ross J, Lemm I, Berberet B. Overexpression of an mRNAbinding protein in human colorectal cancer. Oncogene. 2001 Oct 4;20(45):6544-50 Lemm I, Ross J. Regulation of c-myc mRNA decay by translational pausing in a coding region instability determinant. Mol Cell Biol. 2002 Jun;22(12):3959-69 Patel GP, Ma S, Bag J. The autoregulatory translational control element of poly(A)-binding protein mRNA forms a heteromeric ribonucleoprotein complex. Nucleic Acids Res. 2005;33(22):7074-89 Nielsen FC, Nielsen J, Kristensen MA, Koch G, Christiansen J. Cytoplasmic trafficking of IGF-II mRNA-binding protein by conserved KH domains. J Cell Sci. 2002 May 15;115(Pt 10):2087-97 Noubissi FK, Elcheva I, Bhatia N, Shakoori A, Ougolkov A, Liu J, Minamoto T, Ross J, Fuchs SY, Spiegelman VS. CRD-BP mediates stabilization of betaTrCP1 and c-myc mRNA in response to beta-catenin signalling. Nature. 2006 Jun 15;441(7095):898-901 Eom T, Antar LN, Singer RH, Bassell GJ. Localization of a beta-actin messenger ribonucleoprotein complex with zipcodebinding protein modulates the density of dendritic filopodia and filopodial synapses. J Neurosci. 2003 Nov 12;23(32):10433-44 Patel GP, Bag J. IMP1 interacts with poly(A)-binding protein (PABP) and the autoregulatory translational control element of PABP-mRNA through the KH III-IV domain. FEBS J. 2006 Dec;273(24):5678-90 Ioannidis P, Mahaira L, Papadopoulou A, Teixeira MR, Heim S, Andersen JA, Evangelou E, Dafni U, Pandis N, Trangas T. 8q24 Copy number gains and expression of the c-myc mRNA stabilizing protein CRD-BP in primary breast carcinomas. Int J Cancer. 2003 Mar 10;104(1):54-9 Stöhr N, Lederer M, Reinke C, Meyer S, Hatzfeld M, Singer RH, Hüttelmaier S. ZBP1 regulates mRNA stability during cellular stress. J Cell Biol. 2006 Nov 20;175(4):527-34 Nielsen J, Adolph SK, Rajpert-De Meyts E, Lykke-Andersen J, Koch G, Christiansen J, Nielsen FC. Nuclear transit of human zipcode-binding protein IMP1. Biochem J. 2003 Dec 1;376(Pt 2):383-91 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Vikesaa J, Hansen TV, Jønson L, Borup R, Wewer UM, Christiansen J, Nielsen FC. RNA-binding IMPs promote cell adhesion and invadopodia formation. EMBO J. 2006 Apr 5;25(7):1456-68 565 IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1) Trangas T, Ioannidis P Atlas R, Behar L, Sapoznik S, Ginzburg I. Dynamic association with polysomes during P19 neuronal differentiation and an untranslated-region-dependent translation regulation of the tau mRNA by the tau mRNA-associated proteins IMP1, HuD, and G3BP1. J Neurosci Res. 2007 Jan;85(1):173-83 Vainer G, Vainer-Mosse E, Pikarsky A, Shenoy SM, Oberman F, Yeffet A, Singer RH, Pikarsky E, Yisraeli JK. A role for VICKZ proteins in the progression of colorectal carcinomas: regulating lamellipodia formation. J Pathol. 2008 Aug;215(4):445-56 Dimitriadis E, Trangas T, Milatos S, Foukas PG, Gioulbasanis I, Courtis N, Nielsen FC, Pandis N, Dafni U, Bardi G, Ioannidis P. Expression of oncofetal RNA-binding protein CRD-BP/IMP1 predicts clinical outcome in colon cancer. Int J Cancer. 2007 Aug 1;121(3):486-94 Zhou Y, Rong L, Lu J, Pan Q, Liang C. Insulin-like growth factor II mRNA binding protein 1 associates with Gag protein of human immunodeficiency virus type 1, and its overexpression affects virus assembly. J Virol. 2008 Jun;82(12):5683-92 Elcheva I, Goswami S, Noubissi FK, Spiegelman VS. CRD-BP protects the coding region of betaTrCP1 mRNA from miR-183mediated degradation. Mol Cell. 2009 Jul 31;35(2):240-6 Kato T, Hayama S, Yamabuki T, Ishikawa N, Miyamoto M, Ito T, Tsuchiya E, Kondo S, Nakamura Y, Daigo Y. Increased expression of insulin-like growth factor-II messenger RNAbinding protein 1 is associated with tumor progression in patients with lung cancer. Clin Cancer Res. 2007 Jan 15;13(2 Pt 1):434-42 Noubissi FK, Goswami S, Sanek NA, Kawakami K, Minamoto T, Moser A, Grinblat Y, Spiegelman VS. Wnt signaling stimulates transcriptional outcome of the Hedgehog pathway by stabilizing GLI1 mRNA. Cancer Res. 2009 Nov 15;69(22):8572-8 Jønson L, Vikesaa J, Krogh A, Nielsen LK, Hansen T, Borup R, Johnsen AH, Christiansen J, Nielsen FC. Molecular composition of IMP1 ribonucleoprotein granules. Mol Cell Proteomics. 2007 May;6(5):798-811 Weidensdorfer D, Stöhr N, Baude A, Lederer M, Köhn M, Schierhorn A, Buchmeier S, Wahle E, Hüttelmaier S. Control of c-myc mRNA stability by IGF2BP1-associated cytoplasmic RNPs. RNA. 2009 Jan;15(1):104-15 Köbel M, Weidensdorfer D, Reinke C, Lederer M, Schmitt WD, Zeng K, Thomssen C, Hauptmann S, Hüttelmaier S. Expression of the RNA-binding protein IMP1 correlates with poor prognosis in ovarian carcinoma. Oncogene. 2007 Nov 29;26(54):7584-9 Weinlich S, Hüttelmaier S, Schierhorn A, Behrens SE, Ostareck-Lederer A, Ostareck DH. IGF2BP1 enhances HCV IRES-mediated translation initiation via the 3'UTR. RNA. 2009 Aug;15(8):1528-42 Sparanese D, Lee CH. CRD-BP shields c-myc and MDR-1 RNA from endonucleolytic attack by a mammalian endoribonuclease. Nucleic Acids Res. 2007;35(4):1209-21 Bennetzen MV, Larsen DH, Bunkenborg J, Bartek J, Lukas J, Andersen JS. Site-specific phosphorylation dynamics of the nuclear proteome during the DNA damage response. Mol Cell Proteomics. 2010 Jun;9(6):1314-23 Dephoure N, Zhou C, Villén J, Beausoleil SA, Bakalarski CE, Elledge SJ, Gygi SP. A quantitative atlas of mitotic phosphorylation. Proc Natl Acad Sci U S A. 2008 Aug 5;105(31):10762-7 Goswami S, Tarapore RS, Teslaa JJ, Grinblat Y, Setaluri V, Spiegelman VS. MicroRNA-340-mediated degradation of microphthalmia-associated transcription factor mRNA is inhibited by the coding region determinant-binding protein. J Biol Chem. 2010 Jul 2;285(27):20532-40 Elcheva I, Tarapore RS, Bhatia N, Spiegelman VS. Overexpression of mRNA-binding protein CRD-BP in malignant melanomas. Oncogene. 2008 Aug 28;27(37):506974 This article should be referenced as such: Gu W, Wells AL, Pan F, Singer RH. Feedback regulation between zipcode binding protein 1 and beta-catenin mRNAs in breast cancer cells. Mol Cell Biol. 2008 Aug;28(16):4963-74 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Trangas T, Ioannidis P. IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):562-566. 566 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review MTA3 (metastasis associated 1 family, member 3) Ansgar Brüning, Ioannis Mylonas University Hospital Munich, Department of Obstetrics/Gynaecology, Molecular Biology Laboratory, Marchioninistrasse 15, 81377 Munchen, Germany (AB, IM) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/MTA3ID41445ch2p21.html DOI: 10.4267/2042/45996 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology The human MTA3 gene is composed of 14 exons. The MTA3 promoter sequence contains SP1, AP1, and oestrogen receptor binding sites (ER half sites). Identity Other names: KIAA1266 HGNC (Hugo): MTA3 Location: 2p21 Transcription Two open reading frames of 1785 bp (isoform 1; 594 AA; MTA3L) and 1548 bp (isoform 2; 515 AA; MTA3S, MTA3) were identified and predicted to be transcribed. The smaller isoform (MTA3S = MTA3) appears to be the most abundantly expressed isoform at the RNA and protein level. DNA/RNA Description The human MTA3 gene was identified through sequence homologies to other members of the MTA gene family (human MTA1, human MTA2, murine MTA3). Pseudogene PGO.9606.51655; PGO.9606.72237. Genomic organization of the human MTA3 gene. The intron/exon structure of MTA3 with start (ATG) and stop (TAA) codons indicated. All 14 exons are depicted; the intron sequences shortened for better graphical visualization. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 567 MTA3 (metastasis associated 1 family, member 3) Brüning A, Mylonas I Domain structure of the MTA3 protein. BAH (bromo-adjacent homology) domain: putative protein-protein interaction domain, involved in gene silencing; ELM (Egl-27 and MTA1 homology) domain: unknown function; SANT (SWI3, ADA2, N-CoR and TFIIIB B) domain: putative DNA binding domain; ZnF (GATA-type zinc finger) domain: direct DNA binding domain. of divers signalling pathways, including the Wnt signalling pathway. Secretion of Wnt factors and their binding by mammary epithelial cells is necessary for correct gland development and its deregulation has been described to be involved in tumorigenesis. MTA3 has been shown to inhibit Wnt4 expression by its transcriptional repression function, causing reduced Wnt4 secretion and subsequent lower beta-catenin levels. Therefore, based on the observations made with transgenic mouse models, expression of MTA3 in mammary epithelial cells has been associated with the inhibition of ductal branching in virgin and pregnant murine mammary glands. Epithelial cancer Deregulation of MTA3 expression in epithelial breast cancer, endometrial cancer, and ovarian cancer is associated with cancer progression by promoting the epithelial-mesenchymal transition (EMT). It is principally believed that reduced expression of MTA3 allows higher expression levels of SNAIL and SLUG, two repressors of metastasis-associated cell adhesion proteins such as E-cadherin and occludin. Haemangiogenesis and lymphomagenesis A high expression level of MTA3 was found in germinal centre B lymphocytes, suggesting an involvement in B cell maturation by direct interaction with BCL6. BCL6 (B-cell lymphoma-6) is a transcriptional repressor that is co-expressed with MTA3 in the germinal centre, where normal B cells proliferate and undergo maturation. BCL6 functions as a transcriptional repressor and suppresses, in cooperation with MTA3, the expression of PRDM1 (Pr domain-containing protein 1), a master regulator of plasma cell differentiation. Overexpression of BCL6 is often observed in lymphomas, especially in large B-cell lymphomas. Thus, the cooperative action of BCL6 together with MTA3 is believed to block differentiation of large B-cell lymphomas, facilitating lymphomagenesis. Placenta development A high expression level of MTA3 in trophoblast cells and trophoblast tumour cells suggests Protein Description MTA3 functions as a transcriptional repressor by interacting with histone deacetylases and nucleosome remodelling complexes such as Mi-2/NuRD. Expression MTA3 expression has been found in normal human breast, ovarian, and endometrial epithelial cells, in malignant breast, ovarian, and endometrial cancer cells and cancer cell lines, in trophoblast cells and chorionic cancer cell lines, in germinal centre B cells, and in B cell-derived lymphomas. A tissue distribution analysis of MTA3 expression in mice revealed an even more widespread distribution of MTA3 in the developing embryo and in adult tissues (heart, brain, spleen, lung, liver and kidney). In humans, MTA3 expression appears to be absent from fibroblasts. Localisation MTA3 exhibits primarily a nuclear localisation, although additional cytoplasmic localisation has been described. Function In epithelial cells, MTA3 maintains the expression of E-cadherin through the suppression of the E-cadherin inhibitor SNAIL. Expression of MTA3 is regulated by oestrogens via direct binding of the oestrogen receptor to the MTA3 promoter and is thus involved in the generation and maintenance of oestrogen-dependent epithelia such as the breast ductal epithelium and the ovarian surface epithelium. Mammary gland development Animal experiments revealed involvement of MTA3 expression in mammary gland morphogenesis mediated by the suppression of the Wnt4 signalling pathway and upregulation of epithelial cell adhesion proteins such as E-cadherin. Normal mammary gland development, as confirmed and studied by several knock out and knock in mouse models, relies on the concerted and correct integration Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 568 MTA3 (metastasis associated 1 family, member 3) Brüning A, Mylonas I The MTA3 regulation network. A. Breast ductal epithelia cells; epithelial cancer cells. B. Germinal center B lymphocytes; B cell-derived lymphomas. The regulation of MTA3 expression and its target genes by transcriptional activators (green) and transcriptional repressors (red) is shown. ER: oestrogen receptor. Fujita N, Jaye DL, Geigerman C, Akyildiz A, Mooney MR, Boss JM, Wade PA. MTA3 and the Mi-2/NuRD complex regulate cell fate during B lymphocyte differentiation. Cell. 2004 Oct 1;119(1):75-86 involvement of MTA3 in placenta development and homeostasis. However, the exact role of MTA3 expression for placenta development and the downstream targets of MTA3 in trophoblast cells are unknown and have to be elucidated. Fujita N, Kajita M, Taysavang P, Wade PA. Hormonal regulation of metastasis-associated protein 3 transcription in breast cancer cells. Mol Endocrinol. 2004 Dec;18(12):2937-49 Homology Mishra SK, Talukder AH, Gururaj AE, Yang Z, Singh RR, et al. Upstream determinants of estrogen receptor-alpha regulation of metastatic tumor antigen 3 pathway. J Biol Chem. 2004 Jul 30;279(31):32709-15 MTA3 exhibits a high homology to human MTA1, MTA2, and murine MTA3. Implicated in Zhang H, Singh RR, Talukder AH, Kumar R. Metastatic tumor antigen 3 is a direct corepressor of the Wnt4 pathway. Genes Dev. 2006 Nov 1;20(21):2943-8 Endometrial cancer Note MTA3 expression is significantly reduced in endometrioid adenocarcinomas of poor differentiation, although not associated with patients' survival. Zhang H, Stephens LC, Kumar R. Metastasis tumor antigen family proteins during breast cancer progression and metastasis in a reliable mouse model for human breast cancer. Clin Cancer Res. 2006 Mar 1;12(5):1479-86 Jaye DL, Iqbal J, Fujita N, Geigerman CM, Li S, et al. The BCL6-associated transcriptional co-repressor, MTA3, is selectively expressed by germinal centre B cells and lymphomas of putative germinal centre derivation. J Pathol. 2007 Sep;213(1):106-15 Ovarian cancer Note MTA3 expression is reduced in ovarian cancer with poor differentiation, although not at significant levels. Brüning A, Makovitzky J, Gingelmaier A, Friese K, Mylonas I. The metastasis-associated genes MTA1 and MTA3 are abundantly expressed in human placenta and chorionic carcinoma cells. Histochem Cell Biol. 2009 Jul;132(1):33-8 Breast cancer Note Although extensively studied on breast cancer cells and tissues, revealing a close correlation of MTA3 expression with oestrogen receptor expression, no studies have yet shown a direct association of MTA3 expression with clinicopathological parameters in breast cancer. Li X, Jia S, Wang S, Wang Y, Meng A. Mta3-NuRD complex is a master regulator for initiation of primitive hematopoiesis in vertebrate embryos. Blood. 2009 Dec 24;114(27):5464-72 Brüning A, Jückstock J, Blankenstein T, Makovitzky J, Kunze S, Mylonas I. The metastasis-associated gene MTA3 is downregulated in advanced endometrioid adenocarcinomas. Histol Histopathol. 2010 Nov;25(11):1447-56 References This article should be referenced as such: Fujita N, Jaye DL, Kajita M, Geigerman C, Moreno CS, Wade PA. MTA3, a Mi-2/NuRD complex subunit, regulates an invasive growth pathway in breast cancer. Cell. 2003 Apr 18;113(2):207-19 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Brüning A, Mylonas I. MTA3 (metastasis associated 1 family, member 3). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):567-569. 569 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review NMT1 (N-myristoyltransferase 1) Ponniah Selvakumar, Sujeet Kumar, Jonathan R Dimmock, Rajendra K Sharma Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N OW8, Canada (PS, SK, RKS); Cancer Research Unit, Saskatchewan Cancer Agency, 20 Campus Drive, Saskatoon, SK S7N 4H4, Canada (PS, SK, RKS); Drug Design and Discovery Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada (JRD) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/NMT1ID43604ch17q21.html DOI: 10.4267/2042/45997 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology It is a monomer and does not require any cofactor or post-translational modifications. The enzyme follows an ordered Bi Bi reaction mechanism in which the apoenzyme binds myristoyl-CoA to form a NMT1myristoyl-CoA binary complex which subsequently binds to protein/peptide substrates. The catalytic conversion (N-myristoylation) is via a direct nucleophilic addition-elimination reaction. The sequential release of CoA and myristoyl-peptide follows the formation of an enzyme-product complex from the enzyme-substrate complex (Farazi et al., 2001; Wright et al., 2009). N-myristoyltransferases 1 have a common preference for myristoyl-CoA but have divergent peptide substrate specificities and the enzyme is highly selective for myristoyl-CoA in vitro and in vivo (Farazi et al., 2001). The protein belongs to GNAT superfamily of enzymes and consists of a saddle-shaped beta-sheet flanked by a helices. There is a pseudo two fold symmetry with regions corresponding to N- and C-terminal portions of the enzyme. The N-terminal half forms the myristoyl-CoA binding site whereas the C-terminal half forms the major portion of the peptide binding site (Farazi et al., 2001; Wright et al., 2009). A large number of crystal structures of NMT1 from yeast and human isoforms are available in apo and complex form. Comparative analysis of the various NMTs has shown that the peptide binding pocket is more divergent than the myristoyl-CoA-binding site (Farazi et al., 2001; Wright et al., 2009). Further, the phospho-proteome analysis studies have shown that the human isoform is phosphorylated in vivo at position 47 (Beausoleil et al., 2004; Beausoleil et al., 2006; Olsen et al., 2006; Identity Other names: NMT HGNC (Hugo): NMT1 Location: 17q21.31 DNA/RNA Description The gene located on the forward strand and spans a size of 47705 bases. It starts at 43138680 and ends at 43186384 bp from pter. The total number of exons is 12. Transcription Alternate splicing. Pseudogene No known pseudogenes. Protein Description N-myristoyltransferase 1 (NMT 1: EC 2.3.1.97) is a key cellular enzyme which carries out lipid modification by facilitating the attachment of myristate to the N-terminal glycine of several protein molecules. The enzyme's function is indispensible for the growth and development of many eukaryotic organisms and several rotaviruses (Duronio et al., 1989; Duronio et al., 1991; Maurer-Stroh and Eisenhower, 2004; Yang et al., 2005; Wright et al., 2009). The best studied homologue of NMT1 is from the S. cerevisiae (Farazi et al., 2001). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 570 NMT1 (N-myristoyltransferase 1) Selvakumar P, et al. Dephoure et al., 2008; Mayya et al., 2009). However the biological significance of this observation is not yet established. general consesus motif of GXXXS/T (where X is any amino acid) (Boutin, 1997; Resh, 1999; Farazi et al., 2001; Wright et al., 2009; Hannoush and Sun, 2010). Various regular endogenous, physiological enzymes and proteins such as protein kinase A, protein kinase G, NADH-cytochrome b5 reductase, nitric oxide synthase, recoverin, most of the G protein a subunit are the substrates of myristoylation among higher eukaryotes. A detailed list of the substrate proteins is available in a number of reviews elsewhere (Boutin, 1997; Resh, 1999; Maurer-Stroh et al., 2004; Selvakumar et al., 2007). Myristoylation increases protein lipophilicity and is important for the full expression of biological functions of proteins. It controls the functioning of proteins by targeting them to specific localization, promoting specific protein-protein and protein-lipid interactions and ligand-induced conformational changes (Resh, 1999; Farazi et al., 2001; Wright et al., 2009). Expression The enzyme is ubiquitous in expression and often exists as isozymes in vivo, varying in either apparent molecular weight and/or subcellular distribution (Selvakumar et al., 2007; Wright et al., 2009). In humans NMT1 is processed to exist as four distinct isoforms ranging from 49 to 68 kDa in size (Giang and Cravatt, 1998). The longer isoform of 496 amino acids represents the full-length protein whereas the shorter isoform represents a translation product of 416 amino acids that initiates with a methionine at amino acid position 81 in the full-length cDNA (Giang and Cravatt, 1998; Farazi et al., 2001). The shorter isoform of NMT1 may arise from an alternative splice variant or through initiation of translation at an internal methionine. Localisation Implicated in NMT1 is a cytoplasmic enzyme because of Nmyristoylation being a co-translational protein modification. Recently, it has been reported that the extended N-terminal domain of the longer isoform of NMT1 is involved in targeting the enzyme to the ribosome but it is not required for activity in vitro (Glover et al., 1997). Targeting to the ribosome appears to be consistent with its role as a co-translational protein modifier. In previous studies it has been observed that NMT1 activity from various cell lines and tissues is associated with membranous and particulate fraction (Magnuson et al., 1995; Boutin, 1997). However, the enzyme activity in particulate fractions in earlier studies could represent an association with ribosomes, rather than an authentic membrane association. Various cancers Note Altered NMT expression is observed in many types of cancer tissues including those of colon, breast, gallbladder and brain (Selvakumar et al., 2007; Wright et al., 2009). A quantitative RT-PCR investigation of hNMT-1 expression during the progression of different human cancers shows that hNMT-1 is upregulated in breast, colon, lung and on average by 3.7 (p=0.032), 3.1 (p=0.001), 2.3 (p=0.003) and 1.8 (p=0.012) fold, respectively (Chen et al., 2009). These findings are explained by the hypothesis that many of the various proteins/oncoproteins (src, ras etc.) which are overexpressed and activated, during tumorigenesis require myristoylation for their proper function (Boutin, 1997; Resh, 1999; Wright et al., 2009). The elevated NMT activity accounts for the functioning of overexpressed oncoproteins and NMT thus plays a role in cancer progression. The NMT substrate src has elevated activity in human cancers and this contributes to its pathogenicity (Frame, 2002). Inhibiting NMT1 functions has also been shown to reduce proliferation and induce apoptosis in human and murine melanoma cell lines and also to block tumor growth in vivo (Bhandarkar et al., 2008). The siRNA mediated NMT1 knockdown shows that silencing NMT1 inhibits cell replication associated with loss of c-Src activation and its target FAK as well as reduction of various protein kinase regulated pathways (Ducker et al., 2005). The knockdown of either of the isozymes, NMT1 or NMT2 results in apoptosis with NMT2 having a more pronounced effect than NMT1. However, in a mouse model the intratumoral injection mainly of NMT1 siRNA has been shown to be responsible for inhibition of tumor growth (Ducker et al., 2005). It has been concluded that among the two isoforms of NMT Function N-myristoyltransferase1 catalyses the covalent attachment of myristate, a 14 carbon saturated fatty acid, via amide bond to the N-terminal glycine residue of several proteins (Wright et al., 2009; Hannoush and Sun, 2010). This lipidic modification is an irreversible process, however not without exceptions (Hannoush and Sun, 2010). Intially this process was thought to be co-translational in which the addition of myristate on the N-terminal glycine takes place after initial amino acid residues (within 100) have been synthesized by the ribosome (Wilcox et al., 1987). The process follows after the removal of the initiator methionine by a methionine aminopeptidase to expose an available Nterminal glycine. However, now it has been shown to occur post-translationally as well when an internal glycine within a polypeptide chain is exposed following a proteolytic cleavage (Zha et al., 2000; Utsumi et al., 2003; Martin et al., 2008). The Availability of exposed N-terminal glycine is an absolute requirement and the modification occurs on a Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 571 NMT1 (N-myristoyltransferase 1) Selvakumar P, et al. cancer patients have offered an advantage for early detection of colorectal cancer using NMT as a blood based marker (Shrivastav et al., 2007; Kumar et al., 2011). The immunohistochemical analysis shows weak to negative staining for NMT in peripheral blood mononuclear cells (PBMC) of controls, whereas strong positivity is observed in the PBMC of colon cancer patients (Shrivastav et al., 2007; Kumar et al., 2011). In addition, NMT is confined mostly in the nuclei of the bone marrow (BM) mononuclear cells of the colon cancer patients, whereas in the control bone marrow specimens it remained cytoplasmic. The strikingly different NMT expression and its altered localization offers the basis of a potential adjunct investigative tool for screening or diagnosis of patients at risk for, or suspected of having, colon cancer (Shrivastav et al., 2007; Kumar et al., 2011). It has been observed that in colon cancer cell lines, an elevated expression of NMT correlates with high levels of c-Src levels (Rajala et al., 2000a). Further it has been observed that the levels of the myristoylated tyrosine kinases, pp60c-src and pp60cyes are several fold higher in colonic preneoplastic lesions and neoplasms compared with normal colon cells (Bolen et al., 1987; Weber et al., 1992; Termuhlen et al., 1993). Differential expression of pp60c-src has been observed in colonic tumor-derived cell lines (Bolen et al., 1987; Weber et al., 1992) and colonic polyps prone to developing cancer (Cartwright et al., 1990). In the intestinal crypt cells, higher levels of cytoskeletal-associated pp60c-src protein tyrosine kinase activity have been observed along with higher expression of pp60c-yes in the normal intestinal epithelium (Zhao et al., 1990; Cartwright et al., 1993). Studies have revealed that pp60c-src is overexpressed in human colon carcinoma and it has enhanced kinase activity in progressive stages and metastases of human colorectal cancer (Bolen et al., 1987; Termuhlen et al., 1993). Furthermore, it has been shown that src kinase activity is positively regulated by myristoylation and the non-myristoylated c-Src exhibited has reduced kinase activity (Patwardhan and Resh, 2010). The blockages of pp60c-src N-myristoylation in colonic cell lines have been reported to result in depressed colony formation and reduced proliferation (Shoji et al., 1990). (NMT1 and NMT2), both have only partially overlapping functions and that NMT1 is critical for tumor cell proliferation further suggesting that isoformspecific inhibitors might be developed as potential anticancer agents (Ducker et al., 2005). It is now apparent that NMT represents both a valuable clinical marker and therapeutic target for cancer (Boutin, 1997; Ducker et al., 2005; Selvakumar et al., 2007; Wright et al., 2009). A several fold increase in NMT activity in polyps and stage B1 tumors compared to normal colonic mucosa have been proposed to be used as a diagnostic/prognostic tool for early detection of colorectal cancer (Raju et al., 1997; Shrivastav et al., 2007; Kumar et al., 2011). Colorectal cancer Disease Colorectal cancer is associated with significantly high mortality and is one of the most common forms of malignancy world wide (Segal and Saltz, 2009). In the western world, it accounts for the second most common cause of cancer associated deaths (Midgley and Kerr, 2001; Tol and Punt, 2010) and is the fourth most common cause of malignancy in the United States (Wolpin et al., 2007; Wolpin and Mayer, 2008). A majority of colon cancer develop from the precancerous polyps on the lining of the colon which grow over the years to becomes cancerous in nature (Midgley and Kerr, 1999). With the increasing armentarium towards colon cancer (Midgley and Kerr, 1999; Midgley and Kerr, 2001; Wolpin et al., 2007; Wolpin and Mayer, 2008; Segal and Saltz, 2009; Tol and Punt, 2010), it is one of the most curable forms of cancer if detected early. However, due to the lack of early symptoms, the majority of the patients have an advanced disease at presentation (Midgley et al., 2001; Segal and Saltz, 2009). Studies have shown that NMT represents both a valuable marker for clinical diagnosis and as a therapeutic target for colon cancer (Magnuson et al., 1995; Raju et al., 1997; Shrivastav et al., 2007; Kumar et al., 2011). Prognosis A direct relationship has been reported for NMT expression and activity and colon cancer progression (Magnuson et al., 1995; Raju et al., 1997). NMT activity and expression has been shown to be upregulated during the progression of colorectal cancer (Magnuson et al., 1995; Raju et al., 1997) and NMT thus has been proposed as a potential chemotherapeutic target (Felsted et al., 1995). A significantly higher NMT activity in rat colonic tumors and a several fold increase in NMT activity in polyps and stage B1 tumors compared to normal colonic mucosa have indicated that NMT could be used as a diagnostic/prognostic tool for colorectal cancer (Magnuson et al., 1995; Raju et al., 1997; Shrivastav et al., 2007). Altered expression and localization of NMT in the peripheral blood and bone marrow of colon Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Gallbladder cancer Disease Gallbladder cancer, also known as carcinoma of the gallbladder, is extremely rare affecting the gall bladder (the organ behind the liver which stores bile produced by the liver). Gallbladder is a non-essential organ and can be removed without significant consequences. However, since gallbladder cancer is very uncommon and many of its symptoms are similar to those of more common ailments (jaundice, pain, and fever), cancer of the gallbladder is usually not found until it is at an advanced stage and cannot be surgically removed. 572 NMT1 (N-myristoyltransferase 1) Selvakumar P, et al. results in opportunistic infections or malignancies leading to the death of individuals in most of the cases. Prognosis The pathogenic states linked to undesired myristoylation activity includes the myristoylation of viral proteins for their proper maturation and infectivity (Boutin, 1997; Maurer-Stroh and Eisenhower, 2004; Wright et al., 2009). Many of the viral genes are homologues of the tyrosine kinases and require Nmyristoylation for the infectivity of viral particles. In the case of HIV infections, viral proteins Gag and Nef require myristoylation by the host cell NMT to carry out their function properly. Gag is the precursor polyprotein for structural components of the viral capsid and requires myristoylation for intracellular localization and its targeting to the lipid rafts in the plasma membrane during virus assembly (Zhou et al., 1994; Resh, 2004; Wright et al., 2009). Nef on the other hand comprises many virulence factors to modify the cellular environment of infected cells to facilitate viral replication and evade detection by cells of the immune system (Collins et al., 1998). It has been reported that NMT1 myristoylates Gag in vivo and inhibiting NMT1 negatively affects HIV production (Takamune et al., 2008). Prognosis Gallbladder cancer tends to spread to the liver or small intestine and also spreads to lymph nodes through the lymphatic system in the region of the liver resulting in involvement of other lymph nodes and organs. The treatments available are not particularly effective, unless the tumor is very small and found in which case the gallbladder is removed for other reasons. A study of documented gallbladder carcinoma cases has been evaluated for NMT and p53 expression by immunohistochemistry in both in situ and in invasive tumor components (Rajala et al., 2000b). Moderate to strong cytoplasmic positivity for NMT with increased intensity in the invasive component was observed in 60% of the cases. A mild to moderate cytoplasmic staining was revealed in the in situ component in 67% of the cases studied. It has been concluded that increased NMT expression in gall bladder tumors is associated with poor clinical outcomes as evidenced by their mean survival times (Rajala et al., 2000b). Breast cancer Disease Breast cancer originates from the breast tissue, most commonly from the inner lining of milk ducts (ductal carcinoma) or the lobules (lobular carcinoma) that supply the ducts with milk. It is the fifth most common cause of cancer death and comprises 10.4% of all cancer incidences among women worldwide, and is the most common type of non-skin cancer in women. Prognosis It has been observed that in the mammary epithelial cells, the proliferative capacity correlates with NMT activity (Clegg et al., 1999). A study of the NMT profiles in tumourigenic or metastatic breast cancer cell lines have displayed reduced NMT activity and western blot analysis shows that NMT1 is phosphorylated in these breast cancer cells (Shrivastav et al., 2009). Furthermore, patients' breast cancer tissue array revealed strong positivity and high intensity for NMT in malignant breast tissues compared with normal breast cells. In the grade I, II, and III infiltrating ductal carcinoma breast tissues, a gradation in the NMT staining was observed (Shrivastav et al., 2009). It has been concluded that NMT may prove to be an additional diagnostic biomarker for breast cancer. References Bolen JB, Veillette A, Schwartz AM, DeSeau V, Rosen N. Activation of pp60c-src protein kinase activity in human colon carcinoma. Proc Natl Acad Sci U S A. 1987 Apr;84(8):2251-5 Wilcox C, Hu JS, Olson EN. Acylation of proteins with myristic acid occurs cotranslationally. Science. 1987 Nov 27;238(4831):1275-8 Duronio RJ, Towler DA, Heuckeroth RO, Gordon JI. Disruption of the yeast N-myristoyl transferase gene causes recessive lethality. Science. 1989 Feb 10;243(4892):796-800 Cartwright CA, Meisler AI, Eckhart W. Activation of the pp60csrc protein kinase is an early event in colonic carcinogenesis. Proc Natl Acad Sci U S A. 1990 Jan;87(2):558-62 Shoji S, Kurosawa T, Inoue H, Funakoshi T, Kubota Y. Human cellular src gene product: identification of the myristoylated pp60c-src and blockage of its myristoyl acylation with N-fatty acyl compounds resulted in the suppression of colony formation. Biochem Biophys Res Commun. 1990 Dec 31;173(3):894-901 Zhao YH, Krueger JG, Sudol M. Expression of cellular-yes protein in mammalian tissues. Oncogene. 1990 Nov;5(11):1629-35 HIV infection Duronio RJ, Rudnick DA, Johnson RL, Johnson DR, Gordon JI. Myristic acid auxotrophy caused by mutation of S. cerevisiae myristoyl-CoA:protein N-myristoyltransferase. J Cell Biol. 1991 Jun;113(6):1313-30 Disease The human immunodeficiency virus (HIV) is a member of the retrovirus family (lentivirus) that causes acquired immune deficiency syndrome (AIDS). In this syndrome the immune system begins to fail leading to lifethreatening opportunistic infections. The major routes of infections are via the transfer of blood, breast milk, semen, vaginal fluid and the pre-ejaculate. If left untreated, the progressive failure of the immune system Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Weber TK, Steele G, Summerhayes IC. Differential pp60c-src activity in well and poorly differentiated human colon carcinomas and cell lines. J Clin Invest. 1992 Sep;90(3):81521 Cartwright CA, Mamajiwalla S, Skolnick SA, Eckhart W, Burgess DR. Intestinal crypt cells contain higher levels of 573 NMT1 (N-myristoyltransferase 1) Selvakumar P, et al. cytoskeletal-associated pp60c-src protein tyrosine kinase activity than do differentiated enterocytes. Oncogene. 1993 Apr;8(4):1033-9 Utsumi T, Sakurai N, Nakano K, Ishisaka R. C-terminal 15 kDa fragment of cytoskeletal actin is posttranslationally Nmyristoylated upon caspase-mediated cleavage and targeted to mitochondria. FEBS Lett. 2003 Mar 27;539(1-3):37-44 Termuhlen PM, Curley SA, Talamonti MS, Saboorian MH, Gallick GE. Site-specific differences in pp60c-src activity in human colorectal metastases. J Surg Res. 1993 Apr;54(4):293-8 Zhou W, Parent LJ, Wills membrane-binding domain human immunodeficiency interacts with acidic Apr;68(4):2556-69 Beausoleil SA, Jedrychowski M, Schwartz D, Elias JE, Villén J, Li J, Cohn MA, Cantley LC, Gygi SP. Large-scale characterization of HeLa cell nuclear phosphoproteins. Proc Natl Acad Sci U S A. 2004 Aug 17;101(33):12130-5 JW, Resh MD. Identification of a within the amino-terminal region of virus type 1 Gag protein which phospholipids. J Virol. 1994 Maurer-Stroh S, Eisenhaber F. Myristoylation of viral and bacterial proteins. Trends Microbiol. 2004 Apr;12(4):178-85 Felsted RL, Glover CJ, Hartman K. Protein N-myristoylation as a chemotherapeutic target for cancer. J Natl Cancer Inst. 1995 Nov 1;87(21):1571-3 Maurer-Stroh S, Gouda M, Novatchkova M, Schleiffer A, Schneider G, Sirota FL, Wildpaner M, Hayashi N, Eisenhaber F. MYRbase: analysis of genome-wide glycine myristoylation enlarges the functional spectrum of eukaryotic myristoylated proteins. Genome Biol. 2004;5(3):R21 Magnuson BA, Raju RV, Moyana TN, Sharma RK. Increased N-myristoyltransferase activity observed in rat and human colonic tumors. J Natl Cancer Inst. 1995 Nov 1;87(21):1630-5 Resh MD. A myristoyl switch regulates membrane binding of HIV-1 Gag. Proc Natl Acad Sci U S A. 2004 Jan 13;101(2):417-8 Boutin JA. Myristoylation. Cell Signal. 1997 Jan;9(1):15-35 Ducker CE, Upson JJ, French KJ, Smith CD. Two Nmyristoyltransferase isozymes play unique roles in protein myristoylation, proliferation, and apoptosis. Mol Cancer Res. 2005 Aug;3(8):463-76 Glover CJ, Hartman KD, Felsted RL. Human Nmyristoyltransferase amino-terminal domain involved in targeting the enzyme to the ribosomal subcellular fraction. J Biol Chem. 1997 Nov 7;272(45):28680-9 Yang SH, Shrivastav A, Kosinski C, Sharma RK, Chen MH, Berthiaume LG, Peters LL, Chuang PT, Young SG, Bergo MO. N-myristoyltransferase 1 is essential in early mouse development. J Biol Chem. 2005 May 13;280(19):18990-5 Raju RV, Moyana TN, Sharma RK. N-Myristoyltransferase overexpression in human colorectal adenocarcinomas. Exp Cell Res. 1997 Aug 25;235(1):145-54 Beausoleil SA, Villén J, Gerber SA, Rush J, Gygi SP. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat Biotechnol. 2006 Oct;24(10):1285-92 Collins KL, Chen BK, Kalams SA, Walker BD, Baltimore D. HIV-1 Nef protein protects infected primary cells against killing by cytotoxic T lymphocytes. Nature. 1998 Jan 22;391(6665):397-401 Giang DK, Cravatt BF. A second mammalian Nmyristoyltransferase. J Biol Chem. 1998 Mar 20;273(12):65958 Olsen JV, Blagoev B, Gnad F, Macek B, Kumar C, Mortensen P, Mann M. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell. 2006 Nov 3;127(3):63548 Clegg RA, Gordge PC, Miller WR. Expression of enzymes of covalent protein modification during regulated and dysregulated proliferation of mammary epithelial cells: PKA, PKC and NMT. Adv Enzyme Regul. 1999;39:175-203 Selvakumar P, Lakshmikuttyamma A, Shrivastav A, Das SB, Dimmock JR, Sharma RK. Potential role of Nmyristoyltransferase in cancer. Prog Lipid Res. 2007 Jan;46(1):1-36 Midgley R, Kerr D. Colorectal cancer. Lancet. 1999 Jan 30;353(9150):391-9 Shrivastav A, Varma S, Saxena A, DeCoteau J, Sharma RK. N-myristoyltransferase: a potential novel diagnostic marker for colon cancer. J Transl Med. 2007 Nov 16;5:58 Resh MD. Fatty acylation of proteins: new insights into membrane targeting of myristoylated and palmitoylated proteins. Biochim Biophys Acta. 1999 Aug 12;1451(1):1-16 Wolpin BM, Meyerhardt JA, Mamon HJ, Mayer RJ. Adjuvant treatment of colorectal cancer. CA Cancer J Clin. 2007 MayJun;57(3):168-85 Rajala RV, Dehm S, Bi X, Bonham K, Sharma RK. Expression of N-myristoyltransferase inhibitor protein and its relationship to c-Src levels in human colon cancer cell lines. Biochem Biophys Res Commun. 2000a Jul 14;273(3):1116-20 Rajala RV, Radhi JM, Kakkar R, Datla RS, Sharma RK. Increased expression of N-myristoyltransferase in gallbladder carcinomas. Cancer. 2000b May 1;88(9):1992-9 Bhandarkar SS, Bromberg J, Carrillo C, Selvakumar P, Sharma RK, Perry BN, Govindarajan B, Fried L, Sohn A, Reddy K, Arbiser JL. Tris (dibenzylideneacetone) dipalladium, a N-myristoyltransferase-1 inhibitor, is effective against melanoma growth in vitro and in vivo. Clin Cancer Res. 2008 Sep 15;14(18):5743-8 Zha J, Weiler S, Oh KJ, Wei MC, Korsmeyer SJ. Posttranslational N-myristoylation of BID as a molecular switch for targeting mitochondria and apoptosis. Science. 2000 Dec 1;290(5497):1761-5 Dephoure N, Zhou C, Villén J, Beausoleil SA, Bakalarski CE, Elledge SJ, Gygi SP. A quantitative atlas of mitotic phosphorylation. Proc Natl Acad Sci U S A. 2008 Aug 5;105(31):10762-7 Farazi TA, Waksman G, Gordon JI. The biology and enzymology of protein N-myristoylation. J Biol Chem. 2001 Oct 26;276(43):39501-4 Martin DD, Vilas GL, Prescher JA, Rajaiah G, Falck JR, Bertozzi CR, Berthiaume LG. Rapid detection, discovery, and identification of post-translationally myristoylated proteins during apoptosis using a bio-orthogonal azidomyristate analog. FASEB J. 2008 Mar;22(3):797-806 Midgley R, Kerr D. Conventional cytotoxic and novel therapeutic concepts in colorectal cancer. Expert Opin Investig Drugs. 2001 Jun;10(6):1011-9 Seaton KE, Smith CD. N-Myristoyltransferase isozymes exhibit differential specificity for human immunodeficiency virus type 1 Gag and Nef. J Gen Virol. 2008 Jan;89(Pt 1):288-96 Frame MC. Src in cancer: deregulation and consequences for cell behaviour. Biochim Biophys Acta. 2002 Jun 21;1602(2):114-30 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 574 NMT1 (N-myristoyltransferase 1) Selvakumar P, et al. Takamune N, Gota K, Misumi S, Tanaka K, Okinaka S, Shoji S. HIV-1 production is specifically associated with human NMT1 long form in human NMT isozymes. Microbes Infect. 2008 Feb;10(2):143-50 Wright MH, Heal WP, Mann DJ, Tate EW.. Protein myristoylation in health and disease. J Chem Biol. 2009 Nov 7. (Epub ahead of print) Wolpin BM, Mayer RJ. Systemic treatment of colorectal cancer. Gastroenterology. 2008 May;134(5):1296-310 Hannoush RN, Sun J.. The chemical toolbox for monitoring protein fatty acylation and prenylation. Nat Chem Biol. 2010 Jul;6(7):498-506. (REVIEW) Chen L, Ling B, Alcorn J, Yang J.. Quantitative Analysis of the Expression of Human N-myristoyltransferase 1 (hNMT-1) in Cancers. The open Biomarker Journal. 2009; 2: 6-10. Patwardhan P, Resh MD.. Myristoylation and membrane binding regulate c-Src stability and kinase activity. Mol Cell Biol. 2010 Sep;30(17):4094-107. Epub 2010 Jun 28. Mayya V, Lundgren DH, Hwang SI, Rezaul K, Wu L, Eng JK, Rodionov V, Han DK.. Quantitative phosphoproteomic analysis of T cell receptor signaling reveals system-wide modulation of protein-protein interactions. Sci Signal. 2009 Aug 18;2(84):ra46. Tol J, Punt CJ.. Monoclonal antibodies in the treatment of metastatic colorectal cancer: a review. Clin Ther. 2010 Mar;32(3):437-53. (REVIEW) Kumar S, Dimmock JR, Sharma RK.. N-Myristoyltransferase in Colon Cancer: A New Marker for Early Diagnosis. Cancers (2011) (Special Issue "Cancer Diagnosis and Targeted Therapy). (Invited review, Manuscript in preparation) Segal NH, Saltz LB.. Evolving treatment of advanced colon cancer. Annu Rev Med. 2009;60:207-19. (REVIEW) Shrivastav A, Varma S, Senger A, Khandelwal RL, Carlsen S, Sharma RK.. Overexpression of Akt/PKB modulates Nmyristoyltransferase activity in cancer cells. J Pathol. 2009 Jul;218(3):391-8. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) This article should be referenced as such: Selvakumar P, Kumar S, Dimmock JR, Sharma RK. NMT1 (Nmyristoyltransferase 1). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):570-575. 575 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review PAEP (progestagen-associated endometrial protein) Hannu Koistinen, Markku Seppälä Department of Clinical Chemistry, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland (HK, MS) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/PAEPID46067ch9q34.html DOI: 10.4267/2042/45998 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Transcription Identity PAEP mRNA (NM_001018049) has 857 bp. Several alternatively spliced mRNA forms have been described, but for most of these evidence for the corresponding protein lacks. Alternative Splicing and Transcript Diversity database (ASTD) reports 16 different transcripts. Other names: GD; GdA; GdF; GdS; MGC138509; MGC142288; PAEG; PEP; PP14 HGNC (Hugo): PAEP Location: 9q34.3 Local order: Several other lipocalin genes have been mapped on the same chromosomal region. From centromere to telomere (GeneLoc database): lipocalin 1 (tear prealbumin, LCN1) - ENSG00000221613 odorant binding protein 2A (OBP2A) - progestagenassociated endometrial protein (PAEP) ENSG00000237339 LOC138159 ENSG00000236543 - glycosyltransferase 6 domain containing 1 (GLT6D1) - lipocalin 9 (LCN9). Pseudogene Not known. Protein Note Some of the localization studies have employed antibodies, the specificity of which is questionable. Some of the biological studies have utilized short peptides derived from PAEP sequence. It is unclear whether such peptides are present in vivo. Glycosylation plays an important part in modulating/dictating the activity of PAEP. In the literature, PAEP is widely referred to as PP14 and glycodelin. DNA/RNA Note Many other lipocalin genes have similar exon/intron organization. Description Description Maps to chromosome 9: 138453602-138458801 on forward (plus) strand (5200 bases). Gene consists of 7 exons. Promoter region contains, by sequence similarity, 2 forward and two reverse Sp1-like binding sites, four putative glucocorticoid/progesterone response elements (PREs), cAMP responsive element (CRE) and activator protein-1 (AP-1) element. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) PAEP (180 amino acids, of which 18 corresponds to signal sequence) is a 28 kDa secreted glycoprotein, belonging to the kernel lipocalin family. Most family members share three conserved sequence motifs. Although sequence similarity between the family members is low, their three dimensional structures are similar. 576 PAEP (progestagen-associated endometrial protein) Koistinen H, Seppälä M Chromosomal location and gene structure of PAEP. Promoter region shows some of the potential regulatory elements. After translationinitiating codon (ATG) exons of the major transcript are shown in black. Some splicing variants contain also parts outside of these exons. PRE: glucocorticoid/progesterone response element; CRE: cAMP responsive element; Sp1: Sp1 transcription factor binding site; AP-1: activator protein-1 element. seminal vesicles. PAEP is also expressed in other epithelial cells of reproductive tissues, such as fallopian tubes, ovary and the breast. In addition, other secretory epithelia, such as eccrine sweat glands and the bronchus epithelium express PAEP. It is also expressed in differentiated areas of breast cancer, ovarian tumors, endometrial adenocarcinoma, and synovial sarcoma. In addition to epithelial tissues, PAEP has been found in megakaryocytes and erythroid precursor cells. Experimental evidence suggests that PAEP expression is regulated by progesterone/progestins, relaxin, and histone deacetylase inhibitors. Lipocalins are small extracellular proteins, many of which bind small hydrophobic molecules, such as retinol and steroids. There is no evidence that PAEP exhibits similar binding properties. PAEP is a glycoprotein with three potential glycosylation sites. Two of them are glycosylated. Many differentially glycosylated forms have been characterized in these sites. Glycosylation modulates/dictates the biological activity of PAEP. Some of the alternatively spliced mRNAs lack the sequences encoding glycosylation sites and/or the lipocalin signature sequence. Expression Localisation The expression of PAEP is highly regulated in a spatiotemporal fashion. In the female, PAEP is mainly expressed in secretory/decidualized endometrial glands after progesterone exposure. In secretory endometrium, expression becomes detectable four days after ovulation and reaches maximum at the end of the menstrual cycle unless pregnancy ensues. PAEP is one of the major proteins in endometrial secretions. In the male, the highest expression has been reported in Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) PAEP is mostly found in exocrine epithelial cells, from which it is secreted into the gland lumen. In breast cancer, PAEP has been found also in paranuclear vacuoles of lobular carcinoma cells. Function PAEP/PP14/glycodelin regulates the functions of spermatozoa during fertilization in a glycosylation dependent manner. 577 PAEP (progestagen-associated endometrial protein) Koistinen H, Seppälä M Swiss model-deduced tertiary structure of the PAEP monomer. The S-S bridge is shown as cylinder and side chain nitrogen atoms of asparagines of potential glycosylation sites are shown as balls. Below are representative examples of the major complex-type glycans present at the N-glycosylation sites Asn 28 and Asn 63 of amniotic fluid glycodelin-isoform (glycodelin-A) and seminal plasma glycodelinisoform (glycodelin-S). Some of the characteristic epitopes are marked by broken line. The various glycoforms of PAEP have different, sometimes even opposite, biological actions at different phases of the fertilization process. Seminal fluid glycodelin-S binds to the sperm head and inhibits premature capacitation. In the female reproductive tract, spermatozoa come into contact with various PAEP glycoforms, that modulate sperm function, e.g., by preventing premature, progesterone-induced acrosome reaction (glycodelin-F). Glycodelin-A inhibits binding of spermatozoa to the zona pellucida, whereas another glycoform (glycodelin-C) stimulates the same. All these actions are glycosylationdependent. PAEP also regulates immune cell functions, which too are, at least in part, regulated by glycosylation. Different PAEP glycoforms contain diverse bi-, tri-, and tetra-antennary complex-type glycans with varying levels of fucose and sialic acid substitution. Glycodelin-A and -F are the most heavily sialylated and inhibit cell proliferation, induce cell death, and suppress interleukin-2 secretion of Jurkat cells and peripheral blood mononuclear cells. No such Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) immunosuppressive effect has been observed for glycodelin-C and -S carrying less or no sialic acids, or for desialylated glycodelin-A and -F. By its immunosuppressive properties one of the PAEP glycoforms (glycodelin-A) may contribute to immunotolerance at the fetomaternal interface and prevent rejection of the fetal semi-allograft. In early pregnancy, glycodelin-A restrains inappropriate invasion of extravillous cytotrophoblasts by suppressing activity of some key metalloproteinases. In breast and endometrial cancer cell lines, PAEP has been found to revert the malignant phenotype in vitro by inducing morphological differentiation and specific gene expression changes. In a preclinical mouse model, transgenic PAEP expression in breast cancer cells has reduced tumor growth. Homology Most lipocalins do not share high sequence similarity, but they are likely to be homologous. Functional PAEP gene has been found in higher primates. Beta-lactoglobulins represent orthologs of 578 PAEP (progestagen-associated endometrial protein) Koistinen H, Seppälä M PAEP, but they are likely to be functionally different from human PAEP, not least because of their differences in glycosylation. No convincing evidence of a PAEP ortholog in mouse or rat has been reported. Prognosis In sporadic breast cancer, PAEP is associated with low proliferation rate and well-differentiated tumors, whereas in familial "non BRCA1/BRCA2" patients, PAEP expression is associated with a less favorable phenotype and increased risk of metastases. Mutations Reproductive failure Note NCBI SNP database reports 128 PAEP SNPs (Homo sapiens, 13 September 2010). Also HinfI restriction enzyme polymorphism has been reported in Finnish population with 5% frequency for allele A1 and 95% frequency for allele A2. No disease associations for mutations have been described. Note During the period of endometrial receptivity for implantation, reduced PAEP secretion/serum levels have been observed in reproductive failure, e.g. in unexplained infertility or recurrent early pregnancy loss. Disease Unexplained infertility or recurrent miscarriage may result from inadequate implantation and/or placentation. Implicated in Ovarian carcinoma Note PAEP is expressed in both normal and malignant ovarian tissue. PAEP has been localized to the cytoplasm of tumor cells and its staining is more frequent in well-differentiated than in poorly differentiated carcinomas. Nuclear progesterone receptors (PRA and PRB) are often coexpressed with cytoplasmic PAEP. Disease In 2002, ovarian cancer was the 6th most common cancer in women, and 7th most common cause of cancer death. Most malignant neoplasms of the ovary originate from the coelomic epithelium. Prognosis In ovarian serous carcinoma, PAEP expression is associated with a more favorable prognosis, even in patients with the same tumor grade and clinical stage. Polycystic ovary syndrome (PCOS) Note Pregnant women with PCOS who subsequently miscarry show subnormal rise of PAEP serum concentration during the first trimester. Disease PCOS is a common endocrine disorder in fertile-aged women. It is associated with ovulatory disturbance, insulin resistance and androgen excess, and is a frequent cause of menstrual disorders and infertility in women. References Joshi SG, Smith RA, Stokes DK. A progestagen-dependent endometrial protein in human amniotic fluid. J Reprod Fertil. 1980 Nov;60(2):317-21 Breast cancer Julkunen M, Koistinen R, Sjöberg J, Rutanen EM, Wahlström T, Seppälä M. Secretory endometrium synthesizes placental protein 14. Endocrinology. 1986 May;118(5):1782-6 Note In breast cancer tissue, PAEP staining has been found in both estrogen and progesterone receptor negative and positive cancers. PAEP is also present in normal breast tissue. Transfection of PAEP in MCF-7 breast cancer cells reverted the malignant phenotype of the cells by inducing morphological differentiation and specific gene expression changes. Furthermore, these cells showed reduced tumor growth in a preclinical xenograft tumor mouse model. Disease Breast cancer is the most common cancer among women worldwide. Although the prognosis has improved following improved diagnosis and therapies, breast cancer remains an important cause of death among women. Most of the neoplasms of the breast originate from the ductal epithelium, while a minority originates from the lobular epithelium. Family history of breast cancer is associated with a 2-3-fold higher risk of the disease. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Bolton AE, Pockley AG, Clough KJ, Mowles EA, Stoker RJ, Westwood OM, Chapman MG. Identification of placental protein 14 as an immunosuppressive factor in human reproduction. Lancet. 1987 Mar 14;1(8533):593-5 Julkunen M, Seppälä M, Jänne OA. Complete amino acid sequence of human placental protein 14: a progesteroneregulated uterine protein homologous to beta-lactoglobulins. Proc Natl Acad Sci U S A. 1988 Dec;85(23):8845-9 Vaisse C, Atger M, Potier B, Milgrom E. Human placental protein 14 gene: sequence and characterization of a short duplication. DNA Cell Biol. 1990 Jul-Aug;9(6):401-13 Garde J, Bell SC, Eperon IC. Multiple forms of mRNA encoding human pregnancy-associated endometrial alpha 2-globulin, a beta-lactoglobulin homologue. Proc Natl Acad Sci U S A. 1991 Mar 15;88(6):2456-60 Kämäräinen M, Julkunen M, Seppälä M. HinfI polymorphism in the human progesterone associated endometrial protein (PAEP) gene. Nucleic Acids Res. 1991 Sep 25;19(18):5092 Okamoto N, Uchida A, Takakura K, Kariya Y, Kanzaki H, Riittinen L, Koistinen R, Seppälä M, Mori T. Suppression by human placental protein 14 of natural killer cell activity. Am J Reprod Immunol. 1991 Dec;26(4):137-42 579 PAEP (progestagen-associated endometrial protein) Koistinen H, Seppälä M Van Cong N, Vaisse C, Gross MS, Slim R, Milgrom E, Bernheim A. The human placental protein 14 (PP14) gene is localized on chromosome 9q34. Hum Genet. 1991 Mar;86(5):515-8 Taylor RN, Savouret JF, Vaisse C, Vigne JL, Ryan I, Hornung D, Seppälä M, Milgrom E. Promegestone (R5020) and mifepristone (RU486) both function as progestational agonists of human glycodelin gene expression in isolated human epithelial cells. J Clin Endocrinol Metab. 1998 Nov;83(11):4006-12 Kämäräinen M, Leivo I, Julkunen M, Seppälä M. Localization of progesterone-associated endometrial protein mRNA by in-situ hybridization in human pregnancy decidua, endometriosis and borderline endometrioid adenoma. J Mol Endocrinol. 1993 Feb;10(1):71-7 Fazleabas AT, Donnelly KM, Srinivasan S, Fortman JD, Miller JB. Modulation of the baboon (Papio anubis) uterine endometrium by chorionic gonadotrophin during the period of uterine receptivity. Proc Natl Acad Sci U S A. 1999 Mar 2;96(5):2543-8 Morrow DM, Xiong N, Getty RR, Ratajczak MZ, Morgan D, Seppala M, Riittinen L, Gewirtz AM, Tykocinski ML. Hematopoietic placental protein 14. An immunosuppressive factor in cells of the megakaryocytic lineage. Am J Pathol. 1994 Dec;145(6):1485-95 Kämäräinen M, Halttunen M, Koistinen R, von Boguslawsky K, von Smitten K, Andersson LC, Seppälä M. Expression of glycodelin in human breast and breast cancer. Int J Cancer. 1999 Dec 10;83(6):738-42 Kämäräinen M, Riittinen L, Seppälä M, Palotie A, Andersson LC. Progesterone-associated endometrial protein--a constitutive marker of human erythroid precursors. Blood. 1994 Jul 15;84(2):467-73 Koistinen H, Koistinen R, Seppälä M, Burova TV, Choiset Y, Haertlé T. Glycodelin and beta-lactoglobulin, lipocalins with a high structural similarity, differ in ligand binding properties. FEBS Lett. 1999 Apr 30;450(1-2):158-62 Dell A, Morris HR, Easton RL, Panico M, Patankar M, Oehniger S, Koistinen R, Koistinen H, Seppala M, Clark GF. Structural analysis of the oligosaccharides derived from glycodelin, a human glycoprotein with potent immunosuppressive and contraceptive activities. J Biol Chem. 1995 Oct 13;270(41):24116-26 Rachmilewitz J, Riely GJ, Tykocinski ML. Placental protein 14 functions as a direct T-cell inhibitor. Cell Immunol. 1999 Jan 10;191(1):26-33 Tseng L, Zhu HH, Mazella J, Koistinen H, Seppälä M. Relaxin stimulates glycodelin mRNA and protein concentrations in human endometrial glandular epithelial cells. Mol Hum Reprod. 1999 Apr;5(4):372-5 Oehninger S, Coddington CC, Hodgen GD, Seppala M. Factors affecting fertilization: endometrial placental protein 14 reduces the capacity of human spermatozoa to bind to the human zona pellucida. Fertil Steril. 1995 Feb;63(2):377-83 Akerstrom B, Flower DR, Salier JP. Lipocalins: unity in diversity. Biochim Biophys Acta. 2000 Oct 18;1482(1-2):1-8 Clark GF, Oehninger S, Patankar MS, Koistinen R, Dell A, Morris HR, Koistinen H, Seppälä M. A role for glycoconjugates in human development: the human feto-embryonic defence system hypothesis. Hum Reprod. 1996 Mar;11(3):467-73 Flower DR, North AC, Sansom CE. The lipocalin protein family: structural and sequence overview. Biochim Biophys Acta. 2000 Oct 18;1482(1-2):9-24 Flower DR. The lipocalin protein family: structure and function. Biochem J. 1996 Aug 15;318 ( Pt 1):1-14 Gutiérrez G, Ganfornina MD, Sánchez D. Evolution of the lipocalin family as inferred from a protein sequence phylogeny. Biochim Biophys Acta. 2000 Oct 18;1482(1-2):35-45 Kämäräinen M, Leivo I, Koistinen R, Julkunen M, Karvonen U, Rutanen EM, Seppälä M. Normal human ovary and ovarian tumors express glycodelin, a glycoprotein with immunosuppressive and contraceptive properties. Am J Pathol. 1996 May;148(5):1435-43 Halttunen M, Kämäräinen M, Koistinen H. Glycodelin: a reproduction-related lipocalin. Biochim Biophys Acta. 2000 Oct 18;1482(1-2):149-56 Riely GJ, Rachmilewitz J, Koo PH, Tykocinski ML. alpha2macroglobulin modulates the immunoregulatory function of the lipocalin placental protein 14. Biochem J. 2000 Oct 15;351 Pt 2:503-8 Morris HR, Dell A, Easton RL, Panico M, Koistinen H, Koistinen R, Oehninger S, Patankar MS, Seppala M, Clark GF. Gender-specific glycosylation of human glycodelin affects its contraceptive activity. J Biol Chem. 1996 Dec 13;271(50):32159-67 Gao J, Mazella J, Seppala M, Tseng L. Ligand activated hPR modulates the glycodelin promoter activity through the Sp1 sites in human endometrial adenocarcinoma cells. Mol Cell Endocrinol. 2001 May 15;176(1-2):97-102 Kämäräinen M, Seppälä M, Virtanen I, Andersson LC. Expression of glycodelin in MCF-7 breast cancer cells induces differentiation into organized acinar epithelium. Lab Invest. 1997 Dec;77(6):565-73 Mukhopadhyay D, Sundereshan S, Rao C, Karande AA. Placental protein 14 induces apoptosis in T cells but not in monocytes. J Biol Chem. 2001 Jul 27;276(30):28268-73 Koistinen H, Koistinen R, Kämäräinen M, Salo J, Seppälä M. Multiple forms of messenger ribonucleic acid encoding glycodelin in male genital tract. Lab Invest. 1997 May;76(5):683-90 Rachmilewitz J, Riely GJ, Huang JH, Chen A, Tykocinski ML. A rheostatic mechanism for T-cell inhibition based on elevation of activation thresholds. Blood. 2001 Dec 15;98(13):3727-32 Stewart DR, Erikson MS, Erikson ME, Nakajima ST, Overstreet JW, Lasley BL, Amento EP, Seppala M. The role of relaxin in glycodelin secretion. J Clin Endocrinol Metab. 1997 Mar;82(3):839-46 Seppälä M, Koistinen H, Koistinen R. Glycodelins. Trends Endocrinol Metab. 2001 Apr;12(3):111-7 Seppälä M, Taylor RN, Koistinen H, Koistinen R, Milgrom E. Glycodelin: a major lipocalin protein of the reproductive axis with diverse actions in cell recognition and differentiation. Endocr Rev. 2002 Aug;23(4):401-30 Kämäräinen M, Miettinen M, Seppala M, von Boguslawsky K, Benassi MS, Böhling T, Andersson LC. Epithelial expression of glycodelin in biphasic synovial sarcomas. Int J Cancer. 1998 May 18;76(4):487-90 Koistinen H, Easton RL, Chiu PC, Chalabi S, Halttunen M, Dell A, Morris HR, Yeung WS, Seppala M, Koistinen R. Differences in glycosylation and sperm-egg binding inhibition of pregnancyrelated glycodelin. Biol Reprod. 2003 Nov;69(5):1545-51 Seppälä M, Bohn H, Tatarinov Y. Glycodelins. Tumour Biol. 1998;19(3):213-20 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 580 PAEP (progestagen-associated endometrial protein) Koistinen H, Seppälä M Mandelin E, Lassus H, Seppälä M, Leminen A, Gustafsson JA, Cheng G, Bützow R, Koistinen R. Glycodelin in ovarian serous carcinoma: association with differentiation and survival. Cancer Res. 2003 Oct 1;63(19):6258-64 Hautala LC, Koistinen R, Seppälä M, Bützow R, Stenman UH, Laakkonen P, Koistinen H. Glycodelin reduces breast cancer xenograft growth in vivo. Int J Cancer. 2008 Nov 15;123(10):2279-84 Rachmilewitz J, Borovsky Z, Riely GJ, Miller R, Tykocinski ML. Negative regulation of T cell activation by placental protein 14 is mediated by the tyrosine phosphatase receptor CD45. J Biol Chem. 2003 Apr 18;278(16):14059-65 Ohta K, Maruyama T, Uchida H, Ono M, Nagashima T, Arase T, Kajitani T, Oda H, Morita M, Yoshimura Y. Glycodelin blocks progression to S phase and inhibits cell growth: a possible progesterone-induced regulator for endometrial epithelial cell growth. Mol Hum Reprod. 2008 Jan;14(1):17-22 Jakubowicz DJ, Essah PA, Seppälä M, Jakubowicz S, Baillargeon JP, Koistinen R, Nestler JE. Reduced serum glycodelin and insulin-like growth factor-binding protein-1 in women with polycystic ovary syndrome during first trimester of pregnancy. J Clin Endocrinol Metab. 2004 Feb;89(2):833-9 Alok A, Karande AA. The role of glycodelin as an immunemodulating agent at the feto-maternal interface. J Reprod Immunol. 2009 Dec;83(1-2):124-7 Chung MK, Chiu PC, Lee CL, Pang RT, Ng EH, Lee KF, Koistinen R, Koistinen H, Seppala M, Yeung WS. Cumulusassociated alpha2-macroglobulin derivative retains proconceptive glycodelin-C in the human cumulus matrix. Hum Reprod. 2009 Nov;24(11):2856-67 Jayachandran R, Shaila MS, Karande AA. Analysis of the role of oligosaccharides in the apoptotic activity of glycodelin A. J Biol Chem. 2004 Mar 5;279(10):8585-91 Mishan-Eisenberg G, Borovsky Z, Weber MC, Gazit R, Tykocinski ML, Rachmilewitz J. Differential regulation of Th1/Th2 cytokine responses by placental protein 14. J Immunol. 2004 Nov 1;173(9):5524-30 Koistinen H, Hautala LC, Seppälä M, Stenman UH, Laakkonen P, Koistinen R. The role of glycodelin in cell differentiation and tumor growth. Scand J Clin Lab Invest. 2009;69(4):452-9 Chiu PC, Chung MK, Tsang HY, Koistinen R, Koistinen H, Seppala M, Lee KF, Yeung WS. Glycodelin-S in human seminal plasma reduces cholesterol efflux and inhibits capacitation of spermatozoa. J Biol Chem. 2005 Jul 8;280(27):25580-9 Lam KK, Chiu PC, Chung MK, Lee CL, Lee KF, Koistinen R, Koistinen H, Seppala M, Ho PC, Yeung WS. Glycodelin-A as a modulator of trophoblast invasion. Hum Reprod. 2009 Sep;24(9):2093-103 Lee CL, Pang PC, Yeung WS, Tissot B, Panico M, Lao TT, Chu IK, Lee KF, Chung MK, Lam KK, Koistinen R, Koistinen H, Seppälä M, Morris HR, Dell A, Chiu PC. Effects of differential glycosylation of glycodelins on lymphocyte survival. J Biol Chem. 2009 May 29;284(22):15084-96 Koistinen H, Seppälä M, Nagy B, Tapper J, Knuutila S, Koistinen R. Glycodelin reduces carcinoma-associated gene expression in endometrial adenocarcinoma cells. Am J Obstet Gynecol. 2005 Dec;193(6):1955-60 Ish-Shalom E, Gargir A, André S, Borovsky Z, Ochanuna Z, Gabius HJ, Tykocinski ML, Rachmilewitz J. alpha2,6Sialylation promotes binding of placental protein 14 via its Ca2+-dependent lectin activity: insights into differential effects on CD45RO and CD45RA T cells. Glycobiology. 2006 Mar;16(3):173-83 Seppälä M, Koistinen H, Koistinen R, Hautala L, Chiu PC, Yeung WS. Glycodelin in reproductive endocrinology and hormone-related cancer. Eur J Endocrinol. 2009 Feb;160(2):121-33 Hautala LC, Greco D, Koistinen R, Heikkinen T, Heikkilä P, Aittomäki K, Blomqvist C, Koistinen H, Nevanlinna H. Glycodelin expression associates with differential tumour phenotype and outcome in sporadic and familial non-BRCA1/2 breast cancer patients. Breast Cancer Res Treat. 2011 Jul;128(1):85-95 Chiu PC, Chung MK, Koistinen R, Koistinen H, Seppala M, Ho PC, Ng EH, Lee KF, Yeung WS. Glycodelin-A interacts with fucosyltransferase on human sperm plasma membrane to inhibit spermatozoa-zona pellucida binding. J Cell Sci. 2007 Jan 1;120(Pt 1):33-44 Lee CL, Chiu PC, Lam KK, Chan RW, Chu IK, Koistinen R, Koistinen H, Seppälä M, Lee KF, Yeung WS. Glycodelin-A modulates cytokine production of peripheral blood natural killer cells. Fertil Steril. 2010 Jul;94(2):769-71 Chiu PC, Chung MK, Koistinen R, Koistinen H, Seppala M, Ho PC, Ng EH, Lee KF, Yeung WS. Cumulus oophorusassociated glycodelin-C displaces sperm-bound glycodelin-A and -F and stimulates spermatozoa-zona pellucida binding. J Biol Chem. 2007 Feb 23;282(8):5378-88 Poornima BL, Karande AA. Differential sialylation regulates the apoptotic activity of glycodelin A. FEBS Lett. 2007 Sep 4;581(22):4366-70 Scholz C, Toth B, Barthell E, Mylonas I, Weissenbacher T, Friese K, Jeschke U. Glycodelin expression in correlation to grading, nodal involvement and steroid receptor expression in human breast cancer patients. Anticancer Res. 2010 May;30(5):1599-603 Seppälä M, Koistinen H, Koistinen R, Chiu PC, Yeung WS. Glycosylation related actions of glycodelin: gamete, cumulus cell, immune cell and clinical associations. Hum Reprod Update. 2007 May-Jun;13(3):275-87 Tsviliana A, Mayr D, Kuhn C, Kunze S, Mylonas I, Jeschke U, Friese K. Determination of glycodelin-A expression correlated to grading and staging in ovarian carcinoma tissue. Anticancer Res. 2010 May;30(5):1637-40 Uchida H, Maruyama T, Ohta K, Ono M, Arase T, Kagami M, Oda H, Kajitani T, Asada H, Yoshimura Y. Histone deacetylase inhibitor-induced glycodelin enhances the initial step of implantation. Hum Reprod. 2007 Oct;22(10):2615-22 This article should be referenced as such: Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Koistinen H, Seppälä M. PAEP (progestagen-associated endometrial protein). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):576-581. 581 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review SHBG (sex hormone-binding globulin) Nicoletta Fortunati, Maria Graziella Catalano Lab Endocrinologia Oncologica, Dip Oncologia, AOU San Giovanni Battista & Dip Fisiopatologia Clinica, Universita di Torino, Via Genova 3, 10126 Torino, Italy (NF, MGC) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SHBGID42286ch17p13.html DOI: 10.4267/2042/45983 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Transcription Identity Two major SHBG transcripts are known, each originating from a different promoter. The variant 1, which has also been referred to as SHBG-L, encodes the longest protein (isoform 1), while the variant 2 uses an alternate in-frame splice site in the 3' coding region compared to variant 1. These two transcripts differ in their 5' sequence and in the absence of exon 7 in the latter one. Other names: ABP; MGC126834; MGC138391; SBP; TEBG HGNC (Hugo): SHBG Location: 17p13.1 Note This gene encodes a steroid binding protein that was first described as a plasma protein secreted by the liver; lately, it was recognized to be produced also by testis germ cells; the protein is now thought to participate in the regulation of steroid responses at cell level. The encoded protein in biological fluids is a dimer formed from identical or nearly identical monomers; in each monomer one steroid binding pocket has been recognized. SHBG binds androgen and estradiol with different affinity. Alternate promoters and several spliced transcripts were reported. Protein Note 402 amino acids; 43779 Da each subunit: homodimer. Description SHBG is a homodimer; each monomer is constituted of 402 aa, molecular weight 43,7 kDa. The protein consists of a signal peptide (1-29 aa) and 2 laminin Glike domains (domain 1: 45-217 aa; domain 2: 224-390 aa). Each SHBG monomer has an O-linked oligosaccharide at Thr(36) and up to two N-linked oligosaccharides at Asn(380) and Asn(396). DNA/RNA Description The human SHBG gene is located on the short arm of chromosome 17 (17pter-p12) and consists of eight exons. Schematic representation of SHBG gene. Exons are represented by the blue boxes. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 582 SHBG (sex hormone-binding globulin) Fortunati N, Catalano MG Linear structure of SHBG protein. Location of glycosylation sites is shown by red lines. subsequently a specific intracellular pathway leading to cross-talk with the estradiol-activated pathway, finally inhibiting several effects of estradiol in breast cancer cells, e.g. cell proliferation. The Asp327Asn polymorphism of SHBG gene is related to breast cancer risk. Cui et al. observed a significant association of the Asp327Asn polymorphism with reduced breast cancer risk and Becchis et al. reported a significantly higher frequency of the polymorphism in postmenopausal patients with ER-positive breast cancer than in ER-negative; more recently Costantino and co-workers suggested a protective role of this polymorphism since mutated SHBG is more effective than wild type protein in inhibiting estradiol-induced cell proliferation and antiapoptosis, and this is due to the fact that D327N SHBG binds to MCF-7 cells to a greater extent than does wild type protein. Localisation SHBG is secreted by liver into the blood stream and it is synthesized by testis germ cells; it also recognizes a specific binding site located on membranes of sex steroid target cells (e.g. breast, prostate). Function SHBG binds and carries sex steroids, regulating their biological active fraction; it also regulates sex steroid effects in target cells by direct action. Homology Protein S, Gas6, laminin, agrin. Mutations Note CCG-CTG; Pro-Leu156; reported in hyperandrogenism. GAC-AAC; Asp-Asn327; reported in estrogendependent breast cancer. (TAAAA)n promoter, n=6-11; n>8; reported in: polycystic ovary syndrome; CAD in postmenopausal women; reduced bone mineral density in men; metabolic syndrome. Prostate cancer Note Patients with prostate cancer showed lower SHBG levels than benign prostate hypertrophy patients and controls. Alternative splicing of SHBG gene is more pronounced in LNCaP and MCF-7 cancer cell lines; at least six independent transcripts each, resulting from alternative splicing of exons 4, 5, 6, and/or 7 were described. SHBG might be a significant multivariate predictor of lymph node invasion in patients with prostate cancer. The use of preoperative serum SHBG could help to identify patients at risk of lymph node invasion. Implicated in Breast cancer Note Human serum sex hormone-binding globulin (SHBG) regulates the bioavailable fraction of circulating estradiol that is known to be a critical factor in breast cancer. In a case-controlled study within the European Prospective Investigation into Cancer and Nutrition (EPIC), SHBG levels in postmenopausal women who developed breast cancer were confirmed to be significantly lower compared with controls, while no significant difference was observed in premenopausal women. SHBG has a direct effect in breast cancer cells; it interacts with membranes of these cells, initiates Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Type 2 diabetes mellitus Note Epidemiological studies consistently show that circulating SHBG levels are lower in type 2 diabetes patients than in non-diabetic individuals. Low circulating levels of SHBG are a strong predictor of the risk of type 2 diabetes in women and men. Carriers of a variant allele of the SHBG singlenucleotide polymorphism (SNP) rs6259 and carriers of 583 SHBG (sex hormone-binding globulin) Fortunati N, Catalano MG translocation breakpoints in a female patient with hypomelanosis of Ito and choroid plexus papilloma. Eur J Hum Genet. 1997 Mar-Apr;5(2):61-8 a rs6257 variant were associated with a risk of type 2 diabetes following their associated sex hormonebinding globulin levels. Becchis M, Frairia R, Ferrera P, Fazzari A, Ondei S, Alfarano A, Coluccia C, Biglia N, Sismondi P, Fortunati N. The additionally glycosylated variant of human sex hormonebinding globulin (SHBG) is linked to estrogen-dependence of breast cancer. Breast Cancer Res Treat. 1999 Mar;54(2):101-7 Insulin resistance and polycystic ovary syndrome (PCOS) Note SHBG concentrations are inversely associated with insulin resistance, and in turn, with the risk of type 2 diabetes. Women with polycystic ovary syndrome (PCOS) present low SHBG levels that are negatively correlated with body mass index and waist to hip ratio, and are, furthermore, associated with insulin resistance. Grishkovskaya I, Avvakumov GV, Sklenar G, Dales D, Hammond GL, Muller YA. Crystal structure of human sex hormone-binding globulin: steroid transport by a laminin G-like domain. EMBO J. 2000 Feb 15;19(4):504-12 Hogeveen KN, Cousin P, Pugeat M, Dewailly D, Soudan B, Hammond GL. Human sex hormone-binding globulin variants associated with hyperandrogenism and ovarian dysfunction. J Clin Invest. 2002 Apr;109(7):973-81 Breakpoints Selva DM, Hogeveen KN, Seguchi K, Tekpetey F, Hammond GL. A human sex hormone-binding globulin isoform accumulates in the acrosome during spermatogenesis. J Biol Chem. 2002 Nov 22;277(47):45291-8 Note An X;17 translocation breakpoint was characterized in a 5-year-old female with hypomelanosis of Ito (HI) who exhibits characteristic hypopigmented lesions, psychomotor retardation, and choroid plexus papilloma. A chromosome-17-specific DNA fragment was isolated and used to identify cosmid clones crossing the translocation from chromosome 17p13. Exon trapping identified two known genes from chromosome 17: FMR1L2 (the fragile X mental retardation syndrome like protein 2) and SHBG (human sex hormone-binding globulin). Mapping the FMR1L2 and SHBG genes showed that neither gene was disrupted by the translocation. Xita N, Tsatsoulis A, Chatzikyriakidou A, Georgiou I. Association of the (TAAAA)n repeat polymorphism in the sex hormone-binding globulin (SHBG) gene with polycystic ovary syndrome and relation to SHBG serum levels. J Clin Endocrinol Metab. 2003 Dec;88(12):5976-80 Catalano MG, Frairia R, Boccuzzi G, Fortunati N. Sex hormone-binding globulin antagonizes the anti-apoptotic effect of estradiol in breast cancer cells. Mol Cell Endocrinol. 2005 Jan 31;230(1-2):31-7 Cui Y, Shu XO, Cai Q, Jin F, Cheng JR, Cai H, Gao YT, Zheng W. Association of breast cancer risk with a common functional polymorphism (Asp327Asn) in the sex hormone-binding globulin gene. Cancer Epidemiol Biomarkers Prev. 2005 May;14(5):1096-101 Kaaks R, Berrino F, Key T, Rinaldi S, Dossus L, Biessy C, Secreto G, Amiano P, Bingham S, Boeing H, Bueno de Mesquita HB, Chang-Claude J, Clavel-Chapelon F, Fournier A, van Gils CH, Gonzalez CA, Gurrea AB, Critselis E, Khaw KT, Krogh V, Lahmann PH, Nagel G, Olsen A, Onland-Moret NC, Overvad K, Palli D, Panico S, Peeters P, Quirós JR, Roddam A, Thiebaut A, Tjønneland A, Chirlaque MD, Trichopoulou A, Trichopoulos D, Tumino R, Vineis P, Norat T, Ferrari P, Slimani N, Riboli E. Serum sex steroids in premenopausal women and breast cancer risk within the European Prospective Investigation into Cancer and Nutrition (EPIC). J Natl Cancer Inst. 2005 May 18;97(10):755-65 References Siiteri PK, Murai JT, Hammond GL, Nisker JA, Raymoure WJ, Kuhn RW. The serum transport of steroid hormones. Recent Prog Horm Res. 1982;38:457-510 Westphal U. Steroid-protein interactions II. Monogr Endocrinol. 1986;27:1-603 Porto CS, Musto NA, Bardin CW, Gunsalus GL. Binding of an extracellular steroid-binding globulin to membranes and soluble receptors from human breast cancer cells (MCF-7 cells). Endocrinology. 1992 May;130(5):2931-6 Kaaks R, Rinaldi S, Key TJ, Berrino F, Peeters PH, Biessy C, Dossus L, Lukanova A, Bingham S, Khaw KT, Allen NE, Bueno-de-Mesquita HB, van Gils CH, Grobbee D, Boeing H, Lahmann PH, Nagel G, Chang-Claude J, Clavel-Chapelon F, Fournier A, Thiébaut A, González CA, Quirós JR, Tormo MJ, Ardanaz E, Amiano P, Krogh V, Palli D, Panico S, Tumino R, Vineis P, Trichopoulou A, Kalapothaki V, Trichopoulos D, Ferrari P, Norat T, Saracci R, Riboli E. Postmenopausal serum androgens, oestrogens and breast cancer risk: the European prospective investigation into cancer and nutrition. Endocr Relat Cancer. 2005 Dec;12(4):1071-82 Fortunati N, Fissore F, Fazzari A, Berta L, Benedusi-Pagliano E, Frairia R. Biological relevance of the interaction between sex steroid binding protein and its specific receptor of MCF-7 cells: effect on the estradiol-induced cell proliferation. J Steroid Biochem Mol Biol. 1993 May;45(5):435-44 Fortunati N, Fissore F, Fazzari A, Becchis M, Comba A, Catalano MG, Berta L, Frairia R. Sex steroid binding protein exerts a negative control on estradiol action in MCF-7 cells (human breast cancer) through cyclic adenosine 3',5'monophosphate and protein kinase A. Endocrinology. 1996 Feb;137(2):686-92 Eriksson AL, Lorentzon M, Mellström D, Vandenput L, Swanson C, Andersson N, Hammond GL, Jakobsson J, Rane A, Orwoll ES, Ljunggren O, Johnell O, Labrie F, Windahl SH, Ohlsson C. SHBG gene promoter polymorphisms in men are associated with serum sex hormone-binding globulin, androgen and androgen metabolite levels, and hip bone mineral density. J Clin Endocrinol Metab. 2006 Dec;91(12):5029-37 Joseph DR. Sequence and functional relationships between androgen-binding protein/sex hormone-binding globulin and its homologs protein S, Gas6, laminin, and agrin. Steroids. 1997 Aug-Sep;62(8-9):578-88 Zajac V, Kirchhoff T, Levy ER, Horsley SW, Miller A, SteichenGersdorf E, Monaco AP. Characterisation of X;17(q12;p13) Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 584 SHBG (sex hormone-binding globulin) Fortunati N, Catalano MG Alevizaki M, Saltiki K, Xita N, Cimponeriu A, Stamatelopoulos K, Mantzou E, Doukas C, Georgiou I. The importance of the (TAAAA)n alleles at the SHBG gene promoter for the severity of coronary artery disease in postmenopausal women. Menopause. 2008 May-Jun;15(3):461-8 novel ligands and function. Mol Cell Endocrinol. 2010 Mar 5;316(1):13-23 Fortunati N, Catalano MG, Boccuzzi G, Frairia R. Sex Hormone-Binding Globulin (SHBG), estradiol and breast cancer. Mol Cell Endocrinol. 2010 Mar 5;316(1):86-92 Costantino L, Catalano MG, Frairia R, Carmazzi CM, Barbero M, Coluccia C, Donadio M, Genta F, Drogo M, Boccuzzi G, Fortunati N. Molecular mechanisms of the D327N SHBG protective role on breast cancer development after estrogen exposure. Breast Cancer Res Treat. 2009 Apr;114(3):449-56 Grosman H, Fabre B, Mesch V, Lopez MA, Schreier L, Mazza O, Berg G. Lipoproteins, sex hormones and inflammatory markers in association with prostate cancer. Aging Male. 2010 Jun;13(2):87-92 Ding EL, Song Y, Manson JE, Hunter DJ, Lee CC, Rifai N, Buring JE, Gaziano JM, Liu S. Sex hormone-binding globulin and risk of type 2 diabetes in women and men. N Engl J Med. 2009 Sep 17;361(12):1152-63 Pugeat M, Nader N, Hogeveen K, Raverot G, Déchaud H, Grenot C. Sex hormone-binding globulin gene expression in the liver: drugs and the metabolic syndrome. Mol Cell Endocrinol. 2010 Mar 5;316(1):53-9 Nakhla AM, Hryb DJ, Rosner W, Romas NA, Xiang Z, Kahn SM. Human sex hormone-binding globulin gene expressionmultiple promoters and complex alternative splicing. BMC Mol Biol. 2009 May 5;10:37 Rosner W, Hryb DJ, Kahn SM, Nakhla AM, Romas NA. Interactions of sex hormone-binding globulin with target cells. Mol Cell Endocrinol. 2010 Mar 5;316(1):79-85 Xita N, Milionis HJ, Galidi A, Lazaros L, Katsoulis K, Elisaf MS, Georgiou I, Tsatsoulis A. The (TAAAA)n polymorphism of the SHBG gene in men with the metabolic syndrome. Exp Clin Endocrinol Diabetes. 2011 Feb;119(2):126-8 Salonia A, Briganti A, Gallina A, Karakiewicz P, Shariat S, Freschi M, Zanni G, Capitanio U, Bosi E, Rigatti P, Montorsi F. Sex hormone-binding globulin: a novel marker for nodal metastases prediction in prostate cancer patients undergoing extended pelvic lymph node dissection. Urology. 2009 Apr;73(4):850-5 This article should be referenced as such: Fortunati N, Catalano MG. SHBG (sex hormone-binding globulin). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):582-585. Avvakumov GV, Cherkasov A, Muller YA, Hammond GL. Structural analyses of sex hormone-binding globulin reveal Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 585 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review SLC39A6 (solute carrier family 39 (zinc transporter), member 6) Shin Hamada, Kennichi Satoh, Tooru Shimosegawa Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai city, Miyagi, Japan (SH, KS, TS) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/SLC39A6ID44189ch18q12.html DOI: 10.4267/2042/45984 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology prostate, placenta, kidney, pituitary and corpus callosum (Taylor et al., 2003). Elevated expressions in malignancy of epithelial origin, such as pancreatic cancer are reported (Unno et al., 2009). Its expression is also reported in the second heart field progenitors, which contribute to the cardiac outflow tract formation (Barth et al., 2010). Identity Other names: LIV-1; ZIP6 HGNC (Hugo): SLC39A6 Location: 18q12.2 DNA/RNA Localisation Description Located at cell membrane. SLC39A6 gene contains ten exons. The length of this gene is 20864 bases. This gene encodes two transcript variants. Isoform 1 utilizes all of ten exons, while isoform 2 lacks exon 2 and 10. Consequently, isoform 2 gives rise to shorter protein than isoform 1. Function According to the structural similarity, may act as a zinc influx transporter. Accelerates nuclear translocation of the transcriptional factor Snail, the inducer of epithelial-mesenchymal transition (EMT), as a downstream target of STAT3 pathway in the zebrafish gastrula organizer (Yamashita et al., 2004). SLC39A6 is induced by histone deacetylase inhibitors' treatment in cancer cells, and involved in the apoptosis induction by histone deacetylase inhibitors (Ma et al., 2009). Transcription Isoform 1; 3637 bases mRNA; 2265 bases of coding region. Isoform 2; 1681 bases mRNA; 1299 bases of coding region. Pseudogene None reported. Homology Protein Mus musculus Slc39a6; Rattus norvegicus Slc39a6; Bos Taurus SLC39A6; Pan troglodytes SLC39A6. Description Mutations SLC39A6 encodes the zinc transporter ZIP6. Isoform 1 consists of 755 amino acids; Isoform 2 consists of 433 amino acids. The molecular weight of ZIP6 is 85 kDa. SLC39A6 is a multi-pass membrane protein and showing the characteristics of zinc transporter (Taylor and Nicholson, 2003). Note No disease related mutations are reported. Implicated in Pancreatic cancer Expression Note SLC39A6 is highly expressed in pancreatic cancer cell ZIP6 is highly expressed in normal breast tissue, Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 586 SLC39A6 (solute carrier family 39 (zinc transporter), member 6) Hamada S, et al. line and pancreatic cancer tissue. Knockdown of SLC39A6 expression in the human pancreatic cancer cell line Panc-1 resulted in the reduced tumorigenicity in nude mice and acquisition of epithelial phenotype, such as increased E-cadherin expression (Unno et al., 2009). Taylor KM, Nicholson RI. The LZT proteins; the LIV-1 subfamily of zinc transporters. Biochim Biophys Acta. 2003 Apr 1;1611(1-2):16-30 Breast cancer Kasper G, Weiser AA, Rump A, Sparbier K, Dahl E, Hartmann A, Wild P, Schwidetzky U, Castaños-Vélez E, Lehmann K. Expression levels of the putative zinc transporter LIV-1 are associated with a better outcome of breast cancer patients. Int J Cancer. 2005 Dec 20;117(6):961-73 Yamashita S, Miyagi C, Fukada T, Kagara N, Che YS, Hirano T. Zinc transporter LIVI controls epithelial-mesenchymal transition in zebrafish gastrula organizer. Nature. 2004 May 20;429(6989):298-302 Note SLC39A6 regulates the expression level of E-cadherin, a epithelial marker in human breast cancer cell line MCF-7 (Shen et al., 2009). Higher expression of SLC39A6 in breast cancer tissue correlates with the better outcome of breast cancer patients (Kasper et al., 2005). SLC39A6 is induced upon the treatment of breast cancer and cervical cancer cell lines by the histone deacetylase inhibitor, TSA. Knockdown of SLC39A6 resulted in the decreased cell death of TSA-treated cancer cells, which indicates the requirement of SLC39A6 during the apoptosis induction (Ma et al., 2009). Zhao L, Chen W, Taylor KM, Cai B, Li X. LIV-1 suppression inhibits HeLa cell invasion by targeting ERK1/2-Snail/Slug pathway. Biochem Biophys Res Commun. 2007 Nov 9;363(1):82-8 Ma X, Ma Q, Liu J, Tian Y, Wang B, Taylor KM, Wu P, Wang D, Xu G, Meng L, Wang S, Ma D, Zhou J. Identification of LIV1, a putative zinc transporter gene responsible for HDACiinduced apoptosis, using a functional gene screen approach. Mol Cancer Ther. 2009 Nov;8(11):3108-16 Shen H, Qin H, Guo J. Concordant correlation of LIV-1 and Ecadherin expression in human breast cancer cell MCF-7. Mol Biol Rep. 2009 Apr;36(4):653-9 Cervical cancer Unno J, Satoh K, Hirota M, Kanno A, Hamada S, Ito H, Masamune A, Tsukamoto N, Motoi F, Egawa S, Unno M, Horii A, Shimosegawa T. LIV-1 enhances the aggressive phenotype through the induction of epithelial to mesenchymal transition in human pancreatic carcinoma cells. Int J Oncol. 2009 Oct;35(4):813-21 Note SLC39A6 is involved in the cellular invasion of HeLa cells by controlling the ERK-mediated Snail and Slug expression (Zhao et al., 2007). Barth JL, Clark CD, Fresco VM, Knoll EP, Lee B, Argraves WS, Lee KH. Jarid2 is among a set of genes differentially regulated by Nkx2.5 during outflow tract morphogenesis. Dev Dyn. 2010 Jul;239(7):2024-33 References Taylor KM, Morgan HE, Johnson A, Hadley LJ, Nicholson RI. Structure-function analysis of LIV-1, the breast cancerassociated protein that belongs to a new subfamily of zinc transporters. Biochem J. 2003 Oct 1;375(Pt 1):51-9 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) This article should be referenced as such: Hamada S, Satoh K, Shimosegawa T. SLC39A6 (solute carrier family 39 (zinc transporter), member 6). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):586-587. 587 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review TRPV1 (transient receptor potential cation channel, subfamily V, member 1) Massimo Nabissi, Giorgio Santoni School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino (MC), Italy (MN, GS) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/TRPV1ID50368ch17p13.html DOI: 10.4267/2042/45985 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity including a coding region and a 5' and a 3' non-coding region. Other names: DKFZp434K0220; VR1 HGNC (Hugo): TRPV1 Location: 17p13.2 Local order: Colocalized with another transient receptor potential channel gene (TRPV3). Transcription There are four transcript variants encoding the same protein, but with different segments in the 5' UTR (var.1, var.2, var.3, var.4) and one alternative splice variant lacking exon 7 (TRPV1b). TRPV1 gene transcription was demonstrated in different cells and tissues, but no data are available on TRPV1 variant expression profiles. DNA/RNA Description TRPV1 gene consists of 17 exons and 17 introns Schematic representation of human TRPV1 gene and neighbouring family gene. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 588 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) Nabissi M, Santoni G Genomic structure of human TRPV1. In gene the exon number and relative length in bp are shown. In cDNA, the coding region is shown by open bars. The non-traslated regions are shown by black filled bars. The different 5' UTR TRPV1 splice variants with relative 5' UTR length are described in table. The TRPV1 splice variant (TRPV1b) is described in table. Hyperlink to FASTA nucleotide sequences of all TRPV1 cDNAs are inserted. Schematic representation of TRPV1 protein. Double broken line is representative of cellular membrane, transmembrane domains are numbered. Red spot indicates the position of the three ankyrin repeat domains and a representative image of the structural ankyrin repeat unit containing two antiparallel helices and a beta-hairpin, with repeats that are stacked in a superhelical arrangement is shows in black box (from NCBI Conserved Domains), N and C (-terminal domains). SP (signal peptide region), ANK (ankyrin regions, red box), TM (trasmembrane domain, grey box), ED (extracellular domain, blue box), CD (cytoplasmatic domain, orange box), PFD (pore forming domain, green box). An association domain (AD) in 685-713 region has been found necessary for self-association. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 589 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) Nabissi M, Santoni G Expression Protein TRPV1 has also been found in different brain region, such as in dopaminergic neurones of the substantia nigra, hippocampal pyramidal neurones, hypothalamic neurones, neurones in the locus coeruleus, and in various layers of the cortex as in small to medium diameter primary afferent fibres, In non-neuronal cells TRPV1 has been found in keratinocytes, bladder urothelium, smooth muscle, liver, polymorphonuclear granulocytes, pancreatic beta-cells, endothelial cells, lymphocytes, thymocytes and macrophages. Moreover by expression profile studies more cells and tissues has been analyzed for TRPV1 expression. Description The canonical form comprises 839 aa (MW~96 kDa) and is composed of six transmembrane spanning domains and a pore forming region between transmembrane domains 5 and 6. The N-terminal and C-terminal tails are in cytoplasmatic side. Three N terminal ankyrin (ANK) repeats are present in Nterminal tail. The variant form TRPV1b is identical to TRPV1 except for the partial deletion of the third ankyrin repeat domain and adjoining polypeptide sequence. Aminoacid modifications has been found (according to Swiss-Prot) in different residues (Table 1). The N-terminal intracellular domain appears to play a pivotal role in intracellular activation of TRPV1, in fact, by mutagenesis analysis a loss of sensitivity to capsaicin has been found related to residue Tyr-511 (Gavva et al., 2004). Modification of a single Nterminal cysteine altered activation of TRPV1 by pungent compounds ranging from onions to garlic (Salazar et al., 2008). The N-terminal intracellular domain also interacts with adjacent modulatory proteins and with the C-terminal intracellular domain. In the closed state, the N-terminal domain is likely exposed to the binding of ATP and a C-terminal region residues interact with PIP-2, facilitating channel activation. In contrast, a desensitized state may be promoted through the interaction of the N- and Cterminal domains through modulatory action involving calcium-calmodulin interacting regions. Moreover, the ankyrin repeat domains residing within the N-terminal intracellular domain forming a region of three repeats spanning amino acids participating in protein-protein (subunit) interactions (Bork, 1993). The presence of concave binding surfaces for ATP within the ANK regions suggest a role of ANKs in modulating channel activation and function (Lishko et al., 2007). Aminoacid Residue modification 117 Phosphoserine 145 Phosphoserine 371 Phosphoserine 502 Phosphoserine 604 Glycosylation 705 Phosphoserine 775 Phosphoserine 801 Phosphoserine 821 Phosphoserine Localisation TRPV1 is expressed in discrete spots in the plasma membrane and cytosol of different cell types (e.g. urothelial cells). Moreover, dorsal root ganglion (DRG) neurons express ectopic but functional TRPV1 channels in the endoplasmic reticulum (ER) (TRPV1(ER)). Function TRPV1 agonists. TRPV1 is a non-selective cation channel, belonging to the superfamily of TRP channels. TRPV1 agonists are of exogenous and endogenous origins. Exogenous agonist are of natural, semisynthetic and synthetic origin. The natural compounds include dietary derived compounds as: capsaicinoids, capsinoids, piperine, allicin, alliin, eugenol and gingerol or non dietary plant compounds as resiniferatoxin, ∆9-tetrahydrocannabinol, cannabidiol and venom from animal origins (Pertwee, 2005; Vriens et al., 2009). Moreover, other environmental irritants as well as noxious heat (> 43-45 °C) has been found to act as TRPV1 agonist. The existence of endogenous vanilloid agonists, a class of compounds referred to as endovanilloids, as TRPV1 channels modulators as been also investigated. TRPV1 has been found to be activated by biogenic amines like Narachidonylethanolamine (AEA, anandamide), Narachidonoyldopamine (NADA), Noleoylethanolamine (OLEA), N-arachidonolylserine, and various N-acyltaurines and N-acylsalsolinols. Various lipids from the fatty acid pool have also been identified as TRPV1 activators, as inflammatory compounds such as bradykinin, products of the lipoxygenases (12-HPETE and leukotriene B4, 5(S)HPETE (hydroperoxyeicosatetranoic acid) and/or leukotriene B4) (Van Der Stelt and Di Marzo, 2004). Also nerve growth factor (NGF), an inflammatory mediator is known to activate/sensitize TRPV1 through the TrkA receptor, act primarily through phosphoinositide-3- kinase (PI3K) and mitogen activated protein kinase (MAPK) signaling pathways (Chuang et al., 2001). Table 1. Aminoacid number and type of putative modification in TRPV1 protein. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 590 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) phosphorylation of TRPV1 by PKC has been located at Ser502 and Ser800 (Bhave et al., 2003). IGF-I (insulin growth factor I). Insulin and IGF-I increase translocation of TRPV1 to the plasma membrane via activation of IGF receptors, which, in turn, induced PI(3) kinase and PKC activation (Van Buren et al., 2005). CdK5 (cyclin-dependent kinase 5). CdK5 can directly phosphorylate Thr407 in TRPV1, while inhibition of CdK5 activity decreases TRPV1 function and Ca2+ influx (Pareek et al., 2007). TRPV1 antagonists. Natural TRPV1 antagonists are actually restricted to two plant derived compounds, the thapsigargin that is the irritant principle of Thapsia garganica L. and yohimbine, an indole alkaloid from the tree Corynanthe yohimbe K. The endogenous TRPV1 antagonists discovery up to now are dynorphins, adenosine, various dietary omega-3 fatty acids like eicosapentaenoic and linolenic acids, the endogenous fatty acid amide hydrolase (FAAH) and different polyamines as putrescine, spermidine, and spermine permeate. The most active non-natural compound that act as TRPV1 antagonist are capsazepine and 5-iodoRTX. Ligand-binding site. By comparative analysis of the primary structure of theTRPV1 and by mutagenesis studies has been revealed a critical role for Tyr511 and Ser512 (between the second intracellular loop and TM3), confirming that the vanilloid binding site is located intracellulary, moreover a third critical residue in the putative TM4 segment (Leu547) was indicated as relevant in ligand-binding. The effect of extracellular protons (as Ca2+), acts primarily by increasing channel opening, rather than interacting directly with the vanilloid binding site. Related TRPV1 pathways intracellular Homology 86% identity with Mus musculus TRPV1, 85% with Rattus norvegicus TRPV1, 65% with human TRPV3. Implicated in Bone cancer Note Bone cancer leads to osteoclast activation, which promotes acidosis and consequently TRPV1 activation in sensory fibers. The correlation between TRPV1 activation and bone cancer pain was demonstrated by the evaluation of the RTX analgesic effects of pharmacological blockade of TRPV1. So, TRPV1 activation plays a critical role in the generation of bone cancer pain, and bone cancer increases TRPV1 expression within distinct subpopulation of DRG neurons (Niiyama et al., 2007). signaling EGFR (epidermal growth factor receptor). TRPV1 has been found to down-regulate epidermal growth factor receptor (EGFR) expression. Interaction of TRPV1 terminal cytosolic domain with EGFR induces EGFR ubiquitination and degradation. Moreover, by transfection of TRPV1 in HEK293 cells a decreased EGFR protein expression was observed (Bode et al., 2009). Fas/CD95. Activation of TRPV1 with capsaicin, in low-grade urothelial cancer cells, induced a TRPV1dependent G0/G1 cell cycle arrest and apoptosis by inducing transcription of pro-apoptotic genes Fas/CD95, Bcl-2 and caspases, and by activation of the DNA damage response pathway. Moreover, CPS stimulation induced a TRPV1-dependent redistribution and its clustering with Fas/CD95. In addition, an involvement of capsaicin in activation of the ATM kinase/p53 pathways was found (Amantini et al., 2009). PKA (protein kinase A). TRPV1 are found phosphorylated by PKA in the amino terminus Ser116 and Thr370 and involved in desensitisation while phosphorylation of Ser116 by PKA inhibits dephosphorylation of TRPV1 caused by capsaicin exposure (Mohapatra and Nau, 2003). PKC (protein kinase C). Several inflammatory mediators, like ATP, bradykinin, prostaglandins and trypsin or tryptase activated Gq coupled receptors and induced PKC-dependent phosphorylation of TRPV1 (Moriyama et al., 2003). PKC dependent phosphorylation of TRPV1 potentiates capsaicin- or proton-evoked responses and reduces temperature 'threshold' for TRPV1 activation. Direct Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Nabissi M, Santoni G Skin cancer Oncogenesis TRPV1 is highly and specifically expressed in both premalignant (leukoplasia) and low-grade papillary skin carcinoma, whereas its expression is substantially absent in invasive carcinoma. Recently, TRPV1 has been found to exhibit tumor suppressive activity on skin carcinogenesis in mice because of its ability to down-regulate epidermal growth factor receptor (EGFR) expression; conversely, loss of TRPV1 expression resulted in marked increase in papilloma development. TRPV1 by interacting with EGFR through its terminal cytosolic domain, facilitates Cblmediated EGFR ubiquitination and subsequently its degradation via the lysosomal pathway. In addition, ectopic TRPV1 expression in HEK293 cells resulted in decreased EGFR protein expression, and higher EGFR levels were observed in the skin of TRPV1 deficient mice (TRPV1-/-) as compared to wild-type control animals (Marincsák et al., 2009; Hwang et al., 2010). Urothelial cancer Oncogenesis Changes in the TRPV1 expression occur during the development of human urothelial cancer. Thus, transitional cell carcinoma (TCC) show a progressive decrease in TRPV1 expression as the tumor stage increases. Treatment of low-grade RT4 urothelial 591 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) Nabissi M, Santoni G However, resinferatoxin (RTX), a potent TRPV1 agonist, induced apoptosis by targeting mitochondrial respiration, and decreased pancreatic cancer cell growth in a TRPV1-independent manner (Hartel et al., 2006). cancer cells with a specific TRPV1 agonist, capsaicin (CPS) induced a TRPV1-dependent G0/G1 cell cycle arrest and apoptosis. These events were associated with the transcription of pro-apoptotic genes including Fas/CD95, Bcl-2 and caspases, and with the activation of the DNA damage response pathway. Moreover, stimulation of TRPV1 by CPS significantly increased Fas/CD95 protein expression and more importantly induced a TRPV1-dependent redistribution and clustering of Fas/CD95 that co-localized with the vanilloid receptor, suggesting that Fas/CD95 ligandindependent TRPV1-mediated Fas/CD95 clustering results in death-inducing signaling complex formation and triggering of apoptotic signaling through both the extrinsic and intrinsic mitochondrial-dependent pathways. Moreover, we found that CPS activates the ATM kinase involved in p53 Ser15, Ser20 and Ser392 phosphorylation. ATM activation is involved in Fas/CD95 up-regulation and co-clustering with TRPV1 as well as in urothelial cancer cell growth and apoptosis. Finally, the role of TRPV1 mRNA downregulation as a negative prognostic factor in patients with bladder cancer has been reported. By univariate analysis, cumulative survival curves calculated according to the Kaplan-Meier method for the canonic prognostic parameters such as tumor grade and high stage (pT4), lymph nodes and distant diagnosed metastasis, reached significance. Notably, the reduction of TRPV1 mRNA expression was associated with a shorter survival of urothelial cancer patients (P=0.008). On multivariate Cox regression analysis, TRPV1 mRNA expression retained its significance as an independent risk factor, also in a subgroup of patients without diagnosed metastasis (M0). These findings may be particularly important in the stratification of urothelial cancer patients with higher risk of tumor progression for the choice of therapy options (Amantini et al., 2009; Kalogris et al., 2010). Cervical cancer Oncogenesis TRPV1 expression has been reported in human cervical cancer cell lines and tissues, and the endocannabinod anandamide (AEA) induced TRPV1-dependent tumor cell apoptosis. In addition, TRPV1 stimulation completely reverted the cannabidiol (CBD)-mediated inhibitory effect on human cervical cancer cell invasion by blocking CBD-induced increase of TIMP-1, a MMP inhibitor both at mRNA and protein levels, and ERK1/ERK2 and p38MAPK activation (Contassot et al., 2004a; Contassot et al., 2004b). Prostate cancer Oncogenesis A functional TRPV1 channel is expressed in human prostate cancer cells (PC3 and LNCaP) and in prostate hyperplasic tissue. Moreover, increased TRPV1 mRNA and protein expression was found in human prostate cancer tissues as compared to prostate hyperplastic and healthy donors, and this increase correlated with degree of malignancy. CPS induced a growth inhibition and apoptosis of PC3 prostate cancer cells, but in TRPV1independent manner, through ROS generation, mitochondrial inner transmembrane potential dissipation and caspase-3 activation. Moreover, CPS or the specific antagonist capsazepin inhibited tumor growth in vivo, in a xenograft human prostate PC3 cancer model. By contrast, in androgen-responsive LNCaP prostate cancer cells, CPS was found to stimulate TRPV1-dependent cell proliferation. CPS effects were attributable to decreased ceramide levels and to activation of Akt/PKB and ERK pathways, and were associated with increased androgen receptor expression (Sanchez et al., 2005). Glioblastoma Oncogenesis TRPV1 mRNA and protein expression was evidenced in normal astrocytes and glioma cells and tissues. Its expression inversely correlated with glioma grading, with a marked loss of TRPV1 expression in the majority of grade IV glioblastoma tissues. TRPV1 activation by CPS induced apoptosis of U373MG glioma cells, and involved rise of Ca2+ influx, p38MAPK activation, mitochondrial permeability transmembrane pore opening and transmembrane potential dissipation, and caspase-3 activation (Amantini et al., 2007). Oncogenesis TRPV1 expression has been also demonstrated on the plasma membrane of rat pheochromocytoma-derived PC12 cell line. PC12 stimulation by CPS resulted in TRPV1-dependent nitric oxide synthase (iNOS) expression. CPS exposure triggered Ca2+ influx, which in turn enhanced mitochondrial Ca2+ accumulation and promoted NO generation, events that have been associated with tumor progression (Qiao et al., 2004). Pancreatic cancer Hepatocarcinoma Oncogenesis Human pancreatic cancer, significantly expressed increased levels of TRPV1 mRNA and protein. Oncogenesis Hepatocarcinoma patients show high TRPV1 expression that is associated with increased disease-free survival (Miao et al., 2008). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Pheochromocytoma 592 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) expressing afferent nerve fibers (Blumensohn et al., 2002). Digestive tract diseases Note TRPV1 sensitive sensory nerves are densely distributed in the gastrointestinal system, and one of the important roles of these nerves is the preservation of the tissues integrity from the exposed to aggressive compounds, such as protons and activated enzymes. Moreover, activation of TRPV1 either by endogenous or by exogenous agonists exerts hypotensive effects or protective effects against gastrointestinal injury. Therefore, TRPV1 is not only a prime target for the pharmacological control of pain but also a useful target for drug development to treat various gastrointestinal diseases. The function of TRPV1 visceral sensitivity and hypersensitivity tends to be well established. It was shown the involvement of TRPV1 in the regulation of gastrointestinal motility and absorption, visceral sensation and visceral hypersensitivity (Holzer, 2010). Cardiovascular diseases Note TRPV1 is expressed in cardiac spinal sympathetic sensory fibers. During cardiac ischemia these fibers are essential for the sympathoexcitatory reflex, which is associated with increased blood pressure and chest pain. Acidosis TRPV1 activation and ischemia provides the organism with a mechanism, which relays painful information to the brain. Conversely, the release substance P (SP), neurokinin A (NKA) and CGRP by the nerve fiber itself has beneficial effects, which helping to reduce the effects of ischemia and acidosis. Some data indicated that spinal cord stimulation (SCS) used to improve peripheral blood flow in selected populations of patients with ischemia is mediated via VR-1 containing sensory fibers. Treatment of patients with the TRPV1 agonist RTX result in a SCS-induced vasodilation indicating a cardioprotective role for TRPV1 (Wu et al., 2006). Respiratory system diseases Note TRPV1 is expressed on vagal afferent C fibers in the lungs and may be activated by intense heat, acidic solutions, endocannabinoids, metabolites of arachidonic acid, capsaicin and ROS.The role of TRPV1 in respiratory system is correlated to date indicating that acidic solutions as other TRPV1inducing stimuli lead to C-fiber-mediated respiratory reflexes and activation of these fibers leads to bronchoconstriction, mucus secretion, bradycardia and hypotension, in addition to cough and airway irritation (Taylor-Clark and Undem, 2006). Diabetes Note A fundamental role for insulin responsive TRPV1+ in pancreatic sensory neurons in controlling islet inflammation and insulin resistance function and diabetes pathoetiology has been demonstrated. Infact, eliminating these neurons in diabetes-prone NOD mice prevents insulitis and diabetes. In type 2 diabetes administration of capsaicin and RTX which desensitize TRPV1 result in improved glucose tolerance through enhancement of insulin secretion and decreased plasma insulin levels. So ablation of TRPV1-expressing neurons which innervate the pancreas through neonatal capsaicin treatment prevents the insulitis and pancreatic beta-cell destruction that normally occurs in these animals (Gram et al., 2007; Razavi et al., 2006). Bladder diseases Note The role of TRPV1 in overactive (irritable) bladder disease has been shown in TRPV1 knockout mice where differences in their response to bladder injury when compared to their wild-type counterparts. TRPV1 knockout mice didn't develop bladder overactivity during acute bladder inflammation, suggesting a role for TRPV1 in bladder inflammatory states. Moreover, in patients diagnosed with neurogenic detrusor overactivity (NDO), higher levels of TRPV1 immunoreactivity in the urothelium and in the number of nerve fibers were found, compared to control (Apostolidis et al., 2005). Itch Note TRPV1 is expressed on the "pruriceptor subpopulation" of mechano insensitive fibers and the itch-selective sensory afferents respond to capsaicin. Itch sensation can be modulate by changing skin temperature and pH, to common TRPV1 activator stimuli. Therefore, TRPV1 may function as a 'central integrator' molecule in the itch pathway (Yosipovitch et al., 2005; Ghilardi et al., 2005). Diseases of the basal ganglia Note TRPV1 plays a role in dopaminergic mechanisms associated with schizophrenia and Parkinson's disease. Exposure of mesencephalic dopaminergic neurons to the TRPV1 agonist capsaicin triggers cell death, while exposure to TRPV1 antagonists prevents these effects. In addition, schizophrenic patients tend to display reduced pain sensitivity and a diminished skin flare response to niacin, suggesting a defects in TRPV1- Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Nabissi M, Santoni G References Bork P. Hundreds of ankyrin-like repeats in functionally diverse proteins: mobile modules that cross phyla horizontally? Proteins. 1993 Dec;17(4):363-74 Chuang HH, Prescott ED, Kong H, Shields S, Jordt SE, Basbaum AI, Chao MV, Julius D. Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2mediated inhibition. Nature. 2001 Jun 21;411(6840):957-62 593 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) Blumensohn R, Ringler D, Eli I. Pain perception in patients with schizophrenia. J Nerv Ment Dis. 2002 Jul;190(7):481-3 Van Buren JJ, Bhat S, Rotello R, Pauza ME, Premkumar LS. Sensitization and translocation of TRPV1 by insulin and IGF-I. Mol Pain. 2005 Apr 27;1:17 Bhave G, Hu HJ, Glauner KS, Zhu W, Wang H, Brasier DJ, Oxford GS, Gereau RW 4th. Protein kinase C phosphorylation sensitizes but does not activate the capsaicin receptor transient receptor potential vanilloid 1 (TRPV1). Proc Natl Acad Sci U S A. 2003 Oct 14;100(21):12480-5 Yosipovitch G, Fast K, Bernhard JD. Noxious heat and scratching decrease histamine-induced itch and skin blood flow. J Invest Dermatol. 2005 Dec;125(6):1268-72 Hartel M, di Mola FF, Selvaggi F, Mascetta G, Wente MN, Felix K, Giese NA, Hinz U, Di Sebastiano P, Büchler MW, Friess H. Vanilloids in pancreatic cancer: potential for chemotherapy and pain management. Gut. 2006 Apr;55(4):519-28 Mohapatra DP, Nau C. Desensitization of capsaicin-activated currents in the vanilloid receptor TRPV1 is decreased by the cyclic AMP-dependent protein kinase pathway. J Biol Chem. 2003 Dec 12;278(50):50080-90 Hosseini A, Koskela LR, Ehrén I, Aguilar-Santelises M, Sirsjö A, Wiklund NP. Enhanced formation of nitric oxide in bladder carcinoma in situ and in BCG treated bladder cancer. Nitric Oxide. 2006 Dec;15(4):337-43 Moriyama T, Iida T, Kobayashi K, Higashi T, Fukuoka T, Tsumura H, Leon C, Suzuki N, Inoue K, Gachet C, Noguchi K, Tominaga M. Possible involvement of P2Y2 metabotropic receptors in ATP-induced transient receptor potential vanilloid receptor 1-mediated thermal hypersensitivity. J Neurosci. 2003 Jul 9;23(14):6058-62 Razavi R, Chan Y, Afifiyan FN, Liu XJ, Wan X, Yantha J, Tsui H, Tang L, Tsai S, Santamaria P, Driver JP, Serreze D, Salter MW, Dosch HM. TRPV1+ sensory neurons control beta cell stress and islet inflammation in autoimmune diabetes. Cell. 2006 Dec 15;127(6):1123-35 Amantini C, Mosca M, Lucciarini R, Perfumi M, Morrone S, Piccoli M, Santoni G. Distinct thymocyte subsets express the vanilloid receptor VR1 that mediates capsaicin-induced apoptotic cell death. Cell Death Differ. 2004 Dec;11(12):134256 Sánchez AM, Sánchez MG, Malagarie-Cazenave S, Olea N, Díaz-Laviada I. Induction of apoptosis in prostate tumor PC-3 cells and inhibition of xenograft prostate tumor growth by the vanilloid capsaicin. Apoptosis. 2006 Jan;11(1):89-99 Contassot E, Tenan M, Schnüriger V, Pelte MF, Dietrich PY. Arachidonyl ethanolamide induces apoptosis of uterine cervix cancer cells via aberrantly expressed vanilloid receptor-1. Gynecol Oncol. 2004 Apr;93(1):182-8 Taylor-Clark T, Undem BJ. Transduction mechanisms in airway sensory nerves. J Appl Physiol. 2006 Sep;101(3):950-9 Wu M, Komori N, Qin C, Farber JP, Linderoth B, Foreman RD. Sensory fibers containing vanilloid receptor-1 (VR-1) mediate spinal cord stimulation-induced vasodilation. Brain Res. 2006 Aug 30;1107(1):177-84 Contassot E, Wilmotte R, Tenan M, Belkouch MC, Schnüriger V, de Tribolet N, Burkhardt K, Dietrich PY. Arachidonylethanolamide induces apoptosis of human glioma cells through vanilloid receptor-1. J Neuropathol Exp Neurol. 2004 Sep;63(9):956-63 Amantini C, Mosca M, Nabissi M, Lucciarini R, Caprodossi S, Arcella A, Giangaspero F, Santoni G. Capsaicin-induced apoptosis of glioma cells is mediated by TRPV1 vanilloid receptor and requires p38 MAPK activation. J Neurochem. 2007 Aug;102(3):977-90 Gavva NR, Klionsky L, Qu Y, Shi L, Tamir R, Edenson S, Zhang TJ, Viswanadhan VN, Toth A, Pearce LV, Vanderah TW, Porreca F, Blumberg PM, Lile J, Sun Y, Wild K, Louis JC, Treanor JJ. Molecular determinants of vanilloid sensitivity in TRPV1. J Biol Chem. 2004 May 7;279(19):20283-95 Gram DX, Ahrén B, Nagy I, Olsen UB, Brand CL, Sundler F, Tabanera R, Svendsen O, Carr RD, Santha P, Wierup N, Hansen AJ. Capsaicin-sensitive sensory fibers in the islets of Langerhans contribute to defective insulin secretion in Zucker diabetic rat, an animal model for some aspects of human type 2 diabetes. Eur J Neurosci. 2007 Jan;25(1):213-23 Qiao S, Li W, Tsubouchi R, Murakami K, Yoshino M. Role of vanilloid receptors in the capsaicin-mediated induction of iNOS in PC12 cells. Neurochem Res. 2004 Apr;29(4):687-93 Van Der Stelt M, Di Marzo V. Endovanilloids. Putative endogenous ligands of transient receptor potential vanilloid 1 channels. Eur J Biochem. 2004 May;271(10):1827-34 Lishko PV, Procko E, Jin X, Phelps CB, Gaudet R. The ankyrin repeats of TRPV1 bind multiple ligands and modulate channel sensitivity. Neuron. 2007 Jun 21;54(6):905-18 Apostolidis A, Brady CM, Yiangou Y, Davis J, Fowler CJ, Anand P. Capsaicin receptor TRPV1 in urothelium of neurogenic human bladders and effect of intravesical resiniferatoxin. Urology. 2005 Feb;65(2):400-5 Niiyama Y, Kawamata T, Yamamoto J, Omote K, Namiki A. Bone cancer increases transient receptor potential vanilloid subfamily 1 expression within distinct subpopulations of dorsal root ganglion neurons. Neuroscience. 2007 Aug 24;148(2):560-72 Ghilardi JR, Röhrich H, Lindsay TH, Sevcik MA, Schwei MJ, Kubota K, Halvorson KG, Poblete J, Chaplan SR, Dubin AE, Carruthers NI, Swanson D, Kuskowski M, Flores CM, Julius D, Mantyh PW. Selective blockade of the capsaicin receptor TRPV1 attenuates bone cancer pain. J Neurosci. 2005 Mar 23;25(12):3126-31 Pareek TK, Keller J, Kesavapany S, Agarwal N, Kuner R, Pant HC, Iadarola MJ, Brady RO, Kulkarni AB. Cyclin-dependent kinase 5 modulates nociceptive signaling through direct phosphorylation of transient receptor potential vanilloid 1. Proc Natl Acad Sci U S A. 2007 Jan 9;104(2):660-5 Lazzeri M, Vannucchi MG, Spinelli M, Bizzoco E, Beneforti P, Turini D, Faussone-Pellegrini MS. Transient receptor potential vanilloid type 1 (TRPV1) expression changes from normal urothelium to transitional cell carcinoma of human bladder. Eur Urol. 2005 Oct;48(4):691-8 Jara-Oseguera A, Simon SA, Rosenbaum T. TRPV1: on the road to pain relief. Curr Mol Pharmacol. 2008 Nov;1(3):255-69 Miao X, Liu G, Xu X, Xie C, Sun F, Yang Y, Zhang T, Hua S, Fan W, Li Q, Huang S, Wang Q, Liu G, Zhong D. High expression of vanilloid receptor-1 is associated with better prognosis of patients with hepatocellular carcinoma. Cancer Genet Cytogenet. 2008 Oct;186(1):25-32 Pertwee RG. Pharmacological actions of cannabinoids. Handb Exp Pharmacol. 2005;(168):1-51 Sanchez MG, Sanchez AM, Collado B, Malagarie-Cazenave S, Olea N, Carmena MJ, Prieto JC, Diaz-Laviada I I. Expression of the transient receptor potential vanilloid 1 (TRPV1) in LNCaP and PC-3 prostate cancer cells and in human prostate tissue. Eur J Pharmacol. 2005 May 16;515(1-3):20-7 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Nabissi M, Santoni G Salazar H, Llorente I, Jara-Oseguera A, García-Villegas R, Munari M, Gordon SE, Islas LD, Rosenbaum T. A single Nterminal cysteine in TRPV1 determines activation by pungent 594 TRPV1 (transient receptor potential cation channel, subfamily V, member 1) compounds from onion and garlic. Nat Neurosci. 2008 Mar;11(3):255-61 Marincsák R, Tóth BI, Czifra G, Márton I, Rédl P, Tar I, Tóth L, Kovács L, Bíró T. Increased expression of TRPV1 in squamous cell carcinoma of the human tongue. Oral Dis. 2009 Jul;15(5):328-35 Vriens J, Nilius B, Vennekens R. Herbal compounds and toxins modulating TRP channels. Curr Neuropharmacol. 2008 Mar;6(1):79-96 Marincsák R, Tóth BI, Czifra G, Márton I, Rédl P, Tar I, Tóth L, Kovács L, Bíró T. Increased expression of TRPV1 in squamous cell carcinoma of the human tongue. Oral Dis. 2009 Jul;15(5):328-35 Amantini C, Ballarini P, Caprodossi S, Nabissi M, Morelli MB, Lucciarini R, Cardarelli MA, Mammana G, Santoni G. Triggering of transient receptor potential vanilloid type 1 (TRPV1) by capsaicin induces Fas/CD95-mediated apoptosis of urothelial cancer cells in an ATM-dependent manner. Carcinogenesis. 2009 Aug;30(8):1320-9 Kalogris C, Caprodossi S, Amantini C, Lambertucci F, Nabissi M, Morelli MB, Farfariello V, Filosa A, Emiliozzi MC, Mammana G, Santoni G. Expression of transient receptor potential vanilloid-1 (TRPV1) in urothelial cancers of human bladder: relation to clinicopathological and molecular parameters. Histopathology. 2010 Nov;57(5):744-52 Bode AM, Cho YY, Zheng D, Zhu F, Ericson ME, Ma WY, Yao K, Dong Z. Transient receptor potential type vanilloid 1 suppresses skin carcinogenesis. Cancer Res. 2009 Feb 1;69(3):905-13 Ramer R, Merkord J, Rohde H, Hinz B. Cannabidiol inhibits cancer cell invasion via upregulation of tissue inhibitor of matrix metalloproteinases-1. Biochem Pharmacol. 2010 Apr 1;79(7):955-66 Czifra G, Varga A, Nyeste K, Marincsák R, Tóth BI, Kovács I, Kovács L, Bíró T. Increased expressions of cannabinoid receptor-1 and transient receptor potential vanilloid-1 in human prostate carcinoma. J Cancer Res Clin Oncol. 2009 Apr;135(4):507-14 Holzer P. TRP Channels in the Digestive System. Curr Pharm Biotechnol. 2011 Jan 1;12(1):24-34 Gallego-Sandín S, Rodríguez-García A, Alonso MT, GarcíaSancho J. The endoplasmic reticulum of dorsal root ganglion neurons contains functional TRPV1 channels. J Biol Chem. 2009 Nov 20;284(47):32591-601 This article should be referenced as such: Nabissi M, Santoni G. TRPV1 (transient receptor potential cation channel, subfamily V, member 1). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):588-595. Malagarie-Cazenave S, Olea-Herrero N, Vara D, Díaz-Laviada I. Capsaicin, a component of red peppers, induces expression of androgen receptor via PI3K and MAPK pathways in prostate LNCaP cells. FEBS Lett. 2009 Jan 5;583(1):141-7 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Nabissi M, Santoni G 595 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review WRAP53 (WD repeat containing, antisense to TP53) Marianne Farnebo Karolinska Institutet, Cancer Center Karolinska (CCK) R8 :04, 17176 Stockholm, Sweden (MF) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/WRAP53ID50705ch17p13.html DOI: 10.4267/2042/45986 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Protein Other names: FLJ10385; TCAB1; WDR79 HGNC (Hugo): WRAP53 Location: 17p13.1 Description 548 amino acids; 75 kDa protein; contains from N-term to C-term, a proline-rich region (aa 8-57), a WD40 domain, 5 repeats (160-441), and a glycin-rich region (533-545). DNA/RNA Expression Description Widely expressed, overexpressed in cancer. The WRAP53 gene encompasses 16 kb of DNA; 13 exons (three non-coding alternative start exons: exon 1alpha, 1beta and 1gamma. Exon 1alpha directly overlaps the first exon of TP53 in an antisense fashion by up to 227 base pairs (bp), depending on transcription start site (TSS) usage. Exon 1gamma of WRAP53 is located in the first intron of TP53 overlapping the previously identified transcript Hp53int1 in an antisense fashion. Localisation Cytoplasm and nucleus (enriched in Cajal bodies). Function Essential for Cajal body formation and maintenance. Targets the SMN complex, scaRNAs and telomerase enzyme (via TERC) to Cajal bodies. Inhibition of WRAP53 triggers mitochondrial-dependent apoptosis specifically in cancer cells. Transcription Homology At least 17 splice variants. 1.9 kb mRNA; 1647 bp open reading frame. Regulatory antisense RNA Expression: widely expressed at low levels. Localisation: cytoplasm and nucleus. Function: regulates p53 mRNA levels by interacting with the 5'UTR of p53 mRNA. Homology: conserved in mouse. Diseases implication currently not analysed. Highly-conserved in mammals, the WD40 domain is conserved from human to fly. Mutations Note Single nucleotide polymorphisms (SNPs) in women with breast cancer (see below). Pseudogene Germinal Not known. Not reported. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 596 WRAP53 (WD repeat containing, antisense to TP53) Farnebo M Mahmoudi S, Henriksson S, Corcoran M, Méndez-Vidal C, Wiman KG, Farnebo M. Wrap53, a natural p53 antisense transcript required for p53 induction upon DNA damage. Mol Cell. 2009 Feb 27;33(4):462-71 Somatic Not reported. Implicated in Schildkraut JM, Goode EL, Clyde MA, Iversen ES, Moorman PG, Berchuck A, Marks JR, Lissowska J, Brinton L, Peplonska B, Cunningham JM, Vierkant RA, Rider DN, Chenevix-Trench G, Webb PM, Beesley J, Chen X, Phelan C, Sutphen R, Sellers TA, Pearce L, Wu AH, Van Den Berg D, Conti D, Elund CK, Anderson R, Goodman MT, Lurie G, Carney ME, Thompson PJ, Gayther SA, Ramus SJ, Jacobs I, Krüger Kjaer S, Hogdall E, Blaakaer J, Hogdall C, Easton DF, Song H, Pharoah PD, Whittemore AS, McGuire V, Quaye L, AntonCulver H, Ziogas A, Terry KL, Cramer DW, Hankinson SE, Tworoger SS, Calingaert B, Chanock S, Sherman M, GarciaClosas M. Single nucleotide polymorphisms in the TP53 region and susceptibility to invasive epithelial ovarian cancer. Cancer Res. 2009 Mar 15;69(6):2349-57 Breast and ovarian cancer Note Single nucleotide polymorphisms (SNPs) in WRAP53 were found to be overrepresented in women with breast cancer, in particular estrogen receptor negative breast cancer. The same SNPs were also associated with aggressive ovarian cancer. The SNPs are located in the coding region of WRAP53 and results in the amino acid change R68G. Spinal muscular atrophy (SMA) Tycowski KT, Shu MD, Kukoyi A, Steitz JA. A conserved WD40 protein binds the Cajal body localization signal of scaRNP particles. Mol Cell. 2009 Apr 10;34(1):47-57 Note WRAP53 targets the SMN complex to Cajal Bodies. WRAP53 and SMN association is disrupted in SMA patients suggesting a role of WRAP53 in SMA pathogenesis. Disease Spinal muscular atrophy (SMA) is a common neurodegenerative disorder caused by reduced levels of SMN due to mutations or deletions of the SMN1 gene. SMA is the leading genetic cause of infant mortality worldwide, affecting approximately 1 in 6000 infants. Venteicher AS, Abreu EB, Meng Z, McCann KE, Terns RM, Veenstra TD, Terns MP, Artandi SE. A human telomerase holoenzyme protein required for Cajal body localization and telomere synthesis. Science. 2009 Jan 30;323(5914):644-8 Mahmoudi S, Henriksson S, Weibrecht I, Smith S, Söderberg O, Strömblad S, Wiman KG, Farnebo M. WRAP53 is essential for Cajal body formation and for targeting the survival of motor neuron complex to Cajal bodies. PLoS Biol. 2010 Nov 2;8(11):e1000521 Mahmoudi S, Henriksson S, Farnebo L, Roberg K, Farnebo M.. WRAP53 promotes cancer cell survival and is a potential target for cancer therapy. Cell Death Dis. 2011;2:e114;doi:10.1038/cddis.2010.90. References Garcia-Closas M, Kristensen V, Langerød A, Qi Y, Yeager M, Burdett L, Welch R, Lissowska J, Peplonska B, Brinton L, Gerhard DS, Gram IT, Perou CM, Børresen-Dale AL, Chanock S. Common genetic variation in TP53 and its flanking genes, WDR79 and ATP1B2, and susceptibility to breast cancer. Int J Cancer. 2007 Dec 1;121(11):2532-8 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) This article should be referenced as such: Farnebo M. WRAP53 (WD repeat containing, antisense to TP53). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):596-597. 597 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review YBX1 (Y box binding protein 1) Valentina Evdokimova, Alexey Sorokin Institute of Protein Research, Pushchino, Moscow Region 142290, Russian Federation (VE, AS) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/YBX1ID46554ch1p34.html DOI: 10.4267/2042/45987 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology boxes located between -1855 and -422 nucleotides (relative to the start of exon 1) and several GT and GC boxes. The gene also contains a large and highly conserved CpG island at the immediate 5' promoter region which extends to the first exon encoding 5' UTR of YBX1 mRNA. The region between nucleotides -119 to +127 was shown to be essential for transcriptional activity in the reporter assays (Makino et al., 1996). YBX1 is constitutively expressed in multiple human tissues and its expression can be further induced by the E-box-binding transcription factors such as c-myc (Uramoto et al., 2002), Twist (Shiota et al., 2008) and Math2 (Ohashi et al., 2009). Identity Other names: BP-8; CSDA2; CSDB; DBPB; MDRNF1; MGC104858; MGC110976; MGC117250; NSEP-1; NSEP1; YB-1; YB1 HGNC (Hugo): YBX1 Location: 1p34.2 Local order: The human YBX1 gene maps on 1p34 between the PPIH and the LOC100287607 loci. DNA/RNA Description Transcription The human YBX1 gene consists of 8 exons and 7 introns spanning a 19.2-kb genomic region. Intron number 1 is phase 1 (between 1st and 2nd base of codon). Introns number 2 and 6 are phase 2 (between 2nd and 3rd base of codon). Introns number 3, 4, 5 are phase 0 (between codons). According to the SNP source (dbSNP NCBI), non-synonymous polymorphism has been reported for the codons 30 (rs11558135), 237 (rs3887881), 251 (rs55676223), and 261 (rs3887879). The YBX1 promoter region contains no typical TATA or CCAAT box, but has multiple E- The main processed mRNA is 1514 bp. It encompasses exons 1-8. The 70-amino acid cold-shock domain (CSD) is encoded separately by exons 2-5. Four additional splice variants in human were predicted (Ensembl), two of which (YBX1-004 and YBX1-201) preserve exons 2 and 3 coding for core elements of the CSD, the RNP1 and RNP2 motifs, respectively. An alternative transcript for ctYB-1, the YBX1 homologous gene in C. tentans, has been reported (Nashchekin et al., 2007). Genomic organization of YBX1. Box = exon (blue = 5'UTR, yellow = CDS, light red = 3'UTR). Line = intron. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 598 YBX1 (Y box binding protein 1) Evdokimova V, Sorokin A Pseudogene Description Symbol NCBI gene ID Position Introns/ exons ORF nuclease sensitive element binding protein 1 pseudogene bA327L3.4 158373 9p13.1 Intronless Stops 1477/1515 after E88 (97%) Y box binding protein YBX1P1 1 pseudogene 1 50631 4q23.3 Intronless 1496 bp - 1488/1531 (97%) 20/1511 (1%) Y box binding protein YBX1P2 1 pseudogene 2 646531 7q22.3 2 exons, 1431/1529 Stops 1 intron, after E88 (93%) 1553 bp 43/1529 (2%) Intronless, 820 bp 26/353 (7%) Y box binding protein 10013101 LOC100131012 7q36.1 1 pseudogene 2 287/353 (81%) 2/1515 (0%) The C-terminal region of YB-1 is responsible for sequence-nonspecific binding to DNA and RNA and mediation of protein-protein interactions (Wolffe, 1994; Sommerville and Ladomery, 1996). An inverted CCAAT-box found in HLA class II gene promoters, a so-called Y-box, was originally determined as the YB-1 binding motif (Didier et al., 1988). Later studies have concluded that YB-1 rather recognizes the DNA structure than a defined nucleotide sequence, making prediction of its target genes not feasible with conventional in silico analyses (Swamynathan et al., 1998). YB-1 is also capable of unwinding DNA and RNA duplexes, especially those containing mismatches, thereby promoting strand exchange and formation of perfectly matched duplex structures (Skabkin et al., 2001; Gaudreault et al., 2004). Protein Description The YBX1 gene encodes the Y-box protein 1 (YB-1) which consists of 324 amino acid residues and has the isoelectric point 10.3. Theoretical MW is 35924, however YB-1 is known to migrate as a ~45-50 kDa protein in SDS-polyacrylamide gels due to its anomalous electrophoretic mobility. YB-1 belongs to the family of multifunctional DNA/RNA binding proteins that are highly conserved throughout evolution and found in eukaryotes, prokaryotes and archaea. The most conserved region in YB-1 is the 80 amino acid CSD which exhibits >40% identity and >60% similarity to the major E. coli cold shock protein CspA (Matsumoto and Wolffe, 1998; Sommerville, 1999). The CSD possesses RNP1 and RNP2-like consensus motifs and is represented by a five-stranded beta-barrel structure which creates a surface rich in aromatic and basic amino acids that may act as a large nucleic acidbinding site (Wolffe et al., 1992; Wolffe, 1994). The CSD has a preference for binding single-stranded pyrimidine-rich sequences. The N-terminal AP domain of YB-1 is similar to that found in several other transcription factors and may thus be important for its transcriptional activity. This region is also essential for interaction with p53 and modulation of p53-mediated transcription (Okamoto et al., 2000), and for association with actin microfilaments and mRNA compartmentalization (Ruzanov et al., 1999). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Identity Gaps with YBX1 Expression According to Human Protein Atlas, YB-1 is variably expressed in most normal human tissues. Its expression is elevated in multiple cancer types (Kohno et al., 2003). Localisation Mostly cytosolic. Shuttles between cytoplasm and nucleus. Localized in cytoplasmic stress granules and processing bodies containing untranslated mRNAs (Kedersha and Anderson, 2007). Nuclear translocation is induced in response to various stresses, including adenoviral infection (Holm et al., 2002), hyperthermia (Stein et al., 2001), DNA damage (Kohno et al., 2003) and activation of 599 YBX1 (Y box binding protein 1) Evdokimova V, Sorokin A Structural and functional organization of YB-1. YB-1 is composed by three domains: N-terminal Ala/Pro rich (AP) domain, cold shock domain (CSD) and the C-terminal domain (CTD) containing clusters of positively and negatively charged amino acids. Indicated are some known molecular partners of YB-1 and sites of their interactions (from Sorokin et al., 2005). The arrow indicates proteasomal cleavage sites. PI3K-Akt signaling (Sutherland et al., 2005). 2003). Overall, YB-1 is considered as an important regulator of growth- and stress-associated genes. mRNA translation and stability. YB-1 (p50) is known as a major structural component of messenger ribonucleoprotein particles (mRNPs) which exerts positive or negative effects on translation, depending on the amount bound to mRNA (Evdokimova and Ovchinnikov, 1999). YB-1 regulates translational activity of many growth- and differentiation-associated mRNAs, including Snail1, and selectively protects capped mRNAs against degradation (Evdokimova et al., 2001; Evdokimova et al., 2006; Evdokimova et al., 2009). YB-1 appears to play a role in stabilization of short-lived mRNAs, including IL-2 (Chen et al., 2000), GM-CSF (Capowski et al., 2001) and VEGF (Coles et al., 2004). DNA repair and stress response. YB-1 is involved in base excision and mismatch repair pathways via interaction with multiple DNA repair proteins including glycosylase NEIL2, DNA polymerase beta and delta, DNA ligase III, APE1, MSH2, Ku80, WRN, endonuclease III, etc (Marenstein et al., 2001; Gaudreault et al., 2004; Das et al., 2007). YB-1 also directly binds and promotes separation of DNA strands that contain mismatches or are modified by cisplatin (Ise et al., 1999; Skabkin et al., 2001; Gaudreault et al., 2004). Various stresses, including DNA damage, adenovirus infection and hyperthermia, induce nuclear Function The diverse biological functions of YB-1 appear to arise from its broad nucleic acid binding properties. YB-1 has been implicated in pre-mRNA splicing, transcriptional regulation, mRNA translation and stability as well as in chromatin remodelling, DNA repair and environmental stress responses (Kohno et al., 2003; Matsumoto and Bay, 2005). Splicing. YB-1 regulates splice site selection via direct binding to splicing recognition motifs in pre-mRNA, including A/C-rich exon enhancers (Stickeler et al., 2001) or via interaction with splicing factors from the SR family (Li et al., 2003; Raffetseder et al., 2003). Transcription. YB-1 is capable of binding to promoters of many genes, many of which lack the Ybox, and either activates or represses transcription. Among the genes activated by YB-1 are thymidine kinase, proliferating cell nuclear antigen (PCNA), cyclin A and cyclin B1, DNA topoisomerase II alpha, gelatinase A, matrix metalloproteinase 2, multidrug resistance 1 (MDR1), EGFR and protein tyrosine phosphatase 1B. Genes that are transcriptionally repressed by YB-1 include MHC class II, collagen alpha1, granulocyte-macrophage colony-stimulating factor (GM-CSF), etc (reviewed in Ladomery and Sommerville, 1995; Kohno et al., 2003; Kuwano et al., Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 600 YBX1 (Y box binding protein 1) Evdokimova V, Sorokin A translocation of YB-1 (Ohga et al., 1996; Kohno et al., 2003) and its proteasomal cleavage (Sorokin et al., 2005). Accumulation of the full-length and/or truncated YB-1 proteins in the nucleus is associated with increased survival and multidrug resistance (Kohno et al., 2003; Sorokin et al., 2005). YB-1 knock-out in mice is lethal (Lu et al., 2005; Lu et al., 2006). Fibroblasts derived from YB-1(-/-) embryos exhibit a reduced ability to respond to oxidative, genotoxic and oncogene-induced stresses, further implicating YB-1 in stress responses and embryonic development. Tumorigenesis. YB-1 is frequently overexpressed in multiple human cancers (reviewed in Kohno et al., 2003; Kuwano et al., 2003). In many cases, YB-1 levels are elevated in the nucleus, positively correlating with multiple drug resistance and poor patient outcome (Bargou et al., 1997; Janz et al., 2002). Ectopic expression of YB-1 in breast cancer cells and mouse models stimulated tumor growth (Bergmann et al., 2005; Sutherland et al., 2005). Yet, the role of YB-1 in tumorigenesis is controversial. YB-1 overexpression blocked oncogenic transformation caused by PI3K or Akt (Bader et al., 2003). These apparently contradictory results were proposed to be due to differential localization of YB-1; its interference with oncogenic transformation is associated with cytosolic localization and a consequent function in translational control (Bader and Vogt, 2004; Bader and Vogt, 2005). induced chromosomal instability and tumorigenesis (Bergmann et al., 2005). YB-1 effects on tumorigenesis are likely dependent on cellular signaling. It blocks oncogenic transformation induced by Akt or PI3K but not by Src, Jun or Qin oncoproteins (Bader et al., 2003), and decreases proliferation of tumor cells with activated MAPK-Ras signaling, while inducing their metastatic ability (Evdokimova et al., 2009). Prognosis Nuclear YB-1 is considered as a marker of poor clinical outcome. Patients with high YB-1 levels are likely to benefit from dose-intensified chemotherapy regimens (Gluz et al., 2009). Prostate cancer Note YB-1 is upregulated during prostate cancer tumor progression and is reported to increase P-glycoprotein activity (Giménez-Bonafé et al., 2004). Lung cancer Note Nuclear YB-1 is associated with poor survival and expression of HER2/ErbB2 and HER3/ErbB3 in nonsmall cell lung cancer (Kashihara et al., 2009). Prognosis Patients with nuclear YB-1 expression and p53 mutations appear to have the worst prognosis (median survival 3 months), while best outcome was found in patients with no nuclear YB-1 and wild-type p53 (Gessner et al., 2004). Homology YB-1 is highly homologous to human DbpA (12p13; expressed predominantly in heart and muscle) and DbpC/contrin (17p11; expressed exclusively in germ cells). They share greater than 90% identity within the CSD and a high degree of similarity in the N- and Cterminal domains, including C-terminal clusters of basic and acidic amino acids. Mouse orthologues are YB-1 (encoded by Ybx1; 99% overall aminoacid identity with human YB-1), MSY2 (Ybx2; ~93% identity with contrin) and MSY4 (~86% identity with DbpA). Colon cancer Note YB-1 expression levels are elevated in colorectal carcinoma and positively correlate with DNA topoisomerase II alpha and PCNA expression but not with P-gp (Shibao et al., 1999). In colon cancer cells, YB-1 accumulates in the nuclei in response to vinblastin and is associated with development of vinblastin resistance and elevated expression of P-gp (Vaiman et al., 2007). Mutations Ovarian cancer Note Mutations in YBX1 are not reported. Note YB-1 levels are elevated in the nuclei of cisplatinresistant cancer cell lines and cancer patients, indicating that nuclear YB-1 may be associated with acquired cisplatin resistance in ovarian cancers (Yahata et al., 2002). Prognosis Co-expression of YB-1 and P-gp is indicative of unfavourable prognosis in ovarian cancer (Huang et al., 2004). Implicated in Breast cancer Note Elevated expression and nuclear localization of YB-1 is associated with increased proliferation, multidrug resistance and tumor aggressiveness across all tumor subtypes. Nuclear localization positively correlates with increased expression of MDR1/P-gp and HER2/ErbB2 (Bargou et al., 1997; Saji et al., 2003; Fujii et al., 2008; Habibi et al., 2008). Enforced YB-1 expression in mammary glands of transgenic mice Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Haematopoietic malignancies Disease Large B-cell lymphoma, multiple myeloma. 601 YBX1 (Y box binding protein 1) Evdokimova V, Sorokin A human Y-box binding protein (YB-1) gene promoter. Nucleic Acids Res. 1996 May 15;24(10):1873-8 Nuclear expression of YB-1 is associated with P-gp expression and poor response to chemotherapy in large B-cell lymphoma (Xu et al., 2009). YB-1 is strongly expressed in normal plasma cell precursor blasts as well as in a multiple myeloma tumor specimens and cell lines but not in normal bone marrow or plasma cells. Its expression is associated with an immature morphology, a highly proliferative phenotype and doxorubicin resistance, indicating its involvement in drug resistance and disease progression in multiple myeloma (Chatterjee et al., 2008). Ohga T, Koike K, Ono M, Makino Y, Itagaki Y, Tanimoto M, Kuwano M, Kohno K. Role of the human Y box-binding protein YB-1 in cellular sensitivity to the DNA-damaging agents cisplatin, mitomycin C, and ultraviolet light. Cancer Res. 1996 Sep 15;56(18):4224-8 Sommerville J, Ladomery M. Masking of mRNA by Y-box proteins. FASEB J. 1996 Mar;10(4):435-43 Bargou RC, Jürchott K, Wagener C, Bergmann S, Metzner S, Bommert K, Mapara MY, Winzer KJ, Dietel M, Dörken B, Royer HD. Nuclear localization and increased levels of transcription factor YB-1 in primary human breast cancers are associated with intrinsic MDR1 gene expression. Nat Med. 1997 Apr;3(4):447-50 Bone and soft tissue tumors Disease Rhabdomyosarcoma, synovial sarcoma and osteosarcoma. Nuclear expression of YB-1 protein positively correlates with P-gp expression and a higher proliferative index in embryonal (ERMS) but not in alveolar rhabdomyosarcoma (ARMS) (Oda et al., 2008), synovial sarcoma (Oda et al., 2003) and osteosarcoma (Oda et al., 1998). Matsumoto K, Wolffe AP. Gene regulation by Y-box proteins: coupling control of transcription and translation. Trends Cell Biol. 1998 Aug;8(8):318-23 Oda Y, Sakamoto A, Shinohara N, Ohga T, Uchiumi T, Kohno K, Tsuneyoshi M, Kuwano M, Iwamoto Y. Nuclear expression of YB-1 protein correlates with P-glycoprotein expression in human osteosarcoma. Clin Cancer Res. 1998 Sep;4(9):2273-7 Swamynathan SK, Nambiar A, Guntaka RV. Role of singlestranded DNA regions and Y-box proteins in transcriptional regulation of viral and cellular genes. FASEB J. 1998 May;12(7):515-22 Melanoma Note YB-1 expression is increased in melanoma cells compared to benign melanocytes, and nuclear YB-1 is found in invasive and metastatic melanoma cells. YB-1 expression is associated with increased proliferation, tumor invasion and chemoresistance (Schittek et al., 2007). Evdokimova VM, Ovchinnikov LP. Translational regulation by Y-box transcription factor: involvement of the major mRNAassociated protein, p50. Int J Biochem Cell Biol. 1999 Jan;31(1):139-49 Ise T, Nagatani G, Imamura T, Kato K, Takano H, Nomoto M, Izumi H, Ohmori H, Okamoto T, Ohga T, Uchiumi T, Kuwano M, Kohno K. Transcription factor Y-box binding protein 1 binds preferentially to cisplatin-modified DNA and interacts with proliferating cell nuclear antigen. Cancer Res. 1999 Jan 15;59(2):342-6 Nervous system tumors Disease Glioblastoma, neuroblastoma. YB-1 levels are elevated in pediatric glioblastoma (Faury et al., 2007) and neuroblastoma (Wachowiak et al., 2010). Prognosis In neuroblastoma, no correlation of YB-1 expression with survival, risk factors or stage of the disease was found. Ruzanov PV, Evdokimova VM, Korneeva NL, Hershey JW, Ovchinnikov LP. Interaction of the universal mRNA-binding protein, p50, with actin: a possible link between mRNA and microfilaments. J Cell Sci. 1999 Oct;112 ( Pt 20):3487-96 Shibao K, Takano H, Nakayama Y, Okazaki K, Nagata N, Izumi H, Uchiumi T, Kuwano M, Kohno K, Itoh H. Enhanced coexpression of YB-1 and DNA topoisomerase II alpha genes in human colorectal carcinomas. Int J Cancer. 1999 Dec 10;83(6):732-7 Sommerville J. Activities of cold-shock domain proteins in translation control. Bioessays. 1999 Apr;21(4):319-25 References Chen CY, Gherzi R, Andersen JS, Gaietta G, Jürchott K, Royer HD, Mann M, Karin M. Nucleolin and YB-1 are required for JNK-mediated interleukin-2 mRNA stabilization during T-cell activation. Genes Dev. 2000 May 15;14(10):1236-48 Didier DK, Schiffenbauer J, Woulfe SL, Zacheis M, Schwartz BD. Characterization of the cDNA encoding a protein binding to the major histocompatibility complex class II Y box. Proc Natl Acad Sci U S A. 1988 Oct;85(19):7322-6 Okamoto T, Izumi H, Imamura T, Takano H, Ise T, Uchiumi T, Kuwano M, Kohno K. Direct interaction of p53 with the Y-box binding protein, YB-1: a mechanism for regulation of human gene expression. Oncogene. 2000 Dec 14;19(54):6194-202 Wolffe AP, Tafuri S, Ranjan M, Familari M. The Y-box factors: a family of nucleic acid binding proteins conserved from Escherichia coli to man. New Biol. 1992 Apr;4(4):290-8 Wolffe AP. Structural and functional properties of the evolutionarily ancient Y-box family of nucleic acid binding proteins. Bioessays. 1994 Apr;16(4):245-51 Capowski EE, Esnault S, Bhattacharya S, Malter JS. Y boxbinding factor promotes eosinophil survival by stabilizing granulocyte-macrophage colony-stimulating factor mRNA. J Immunol. 2001 Nov 15;167(10):5970-6 Ladomery M, Sommerville J. A role for Y-box proteins in cell proliferation. Bioessays. 1995 Jan;17(1):9-11 Evdokimova V, Ruzanov P, Imataka H, Raught B, Svitkin Y, Ovchinnikov LP, Sonenberg N. The major mRNA-associated protein YB-1 is a potent 5' cap-dependent mRNA stabilizer. EMBO J. 2001 Oct 1;20(19):5491-502 Makino Y, Ohga T, Toh S, Koike K, Okumura K, Wada M, Kuwano M, Kohno K. Structural and functional analysis of the Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 602 YBX1 (Y box binding protein 1) Evdokimova V, Sorokin A Marenstein DR, Ocampo MT, Chan MK, Altamirano A, Basu AK, Boorstein RJ, Cunningham RP, Teebor GW. Stimulation of human endonuclease III by Y box-binding protein 1 (DNAbinding protein B). Interaction between a base excision repair enzyme and a transcription factor. J Biol Chem. 2001 Jun 15;276(24):21242-9 Saji H, Toi M, Saji S, Koike M, Kohno K, Kuwano M. Nuclear expression of YB-1 protein correlates with P-glycoprotein expression in human breast carcinoma. Cancer Lett. 2003 Feb 20;190(2):191-7 Bader AG, Vogt PK. An essential role for protein synthesis in oncogenic cellular transformation. Oncogene. 2004 Apr 19;23(18):3145-50 Skabkin MA, Evdokimova V, Thomas AA, Ovchinnikov LP. The major messenger ribonucleoprotein particle protein p50 (YB-1) promotes nucleic acid strand annealing. J Biol Chem. 2001 Nov 30;276(48):44841-7 Coles LS, Bartley MA, Bert A, Hunter J, Polyak S, Diamond P, Vadas MA, Goodall GJ. A multi-protein complex containing cold shock domain (Y-box) and polypyrimidine tract binding proteins forms on the vascular endothelial growth factor mRNA. Potential role in mRNA stabilization. Eur J Biochem. 2004 Feb;271(3):648-60 Stein U, Jürchott K, Walther W, Bergmann S, Schlag PM, Royer HD. Hyperthermia-induced nuclear translocation of transcription factor YB-1 leads to enhanced expression of multidrug resistance-related ABC transporters. J Biol Chem. 2001 Jul 27;276(30):28562-9 Gaudreault I, Guay D, Lebel M. YB-1 promotes strand separation in vitro of duplex DNA containing either mispaired bases or cisplatin modifications, exhibits endonucleolytic activities and binds several DNA repair proteins. Nucleic Acids Res. 2004;32(1):316-27 Stickeler E, Fraser SD, Honig A, Chen AL, Berget SM, Cooper TA. The RNA binding protein YB-1 binds A/C-rich exon enhancers and stimulates splicing of the CD44 alternative exon v4. EMBO J. 2001 Jul 16;20(14):3821-30 Gessner C, Woischwill C, Schumacher A, Liebers U, Kuhn H, Stiehl P, Jürchott K, Royer HD, Witt C, Wolff G. Nuclear YB-1 expression as a negative prognostic marker in nonsmall cell lung cancer. Eur Respir J. 2004 Jan;23(1):14-9 Holm PS, Bergmann S, Jurchott K, Lage H, Brand K, Ladhoff A, Mantwill K, Curiel DT, Dobbelstein M, Dietel M, Gansbacher B, Royer HD. YB-1 relocates to the nucleus in adenovirusinfected cells and facilitates viral replication by inducing E2 gene expression through the E2 late promoter. J Biol Chem. 2002 Mar 22;277(12):10427-34 Giménez-Bonafé P, Fedoruk MN, Whitmore TG, Akbari M, Ralph JL, Ettinger S, Gleave ME, Nelson CC. YB-1 is upregulated during prostate cancer tumor progression and increases P-glycoprotein activity. Prostate. 2004 May 15;59(3):337-49 Janz M, Harbeck N, Dettmar P, Berger U, Schmidt A, Jürchott K, Schmitt M, Royer HD. Y-box factor YB-1 predicts drug resistance and patient outcome in breast cancer independent of clinically relevant tumor biologic factors HER2, uPA and PAI-1. Int J Cancer. 2002 Jan 20;97(3):278-82 Huang X, Ushijima K, Komai K, Takemoto Y, Motoshima S, Kamura T, Kohno K. Co-expression of Y box-binding protein-1 and P-glycoprotein as a prognostic marker for survival in epithelial ovarian cancer. Gynecol Oncol. 2004 May;93(2):28791 Uramoto H, Izumi H, Ise T, Tada M, Uchiumi T, Kuwano M, Yasumoto K, Funa K, Kohno K. p73 Interacts with c-Myc to regulate Y-box-binding protein-1 expression. J Biol Chem. 2002 Aug 30;277(35):31694-702 Bader AG, Vogt PK. Inhibition of protein synthesis by Y boxbinding protein 1 blocks oncogenic cell transformation. Mol Cell Biol. 2005 Mar;25(6):2095-106 Yahata H, Kobayashi H, Kamura T, Amada S, Hirakawa T, Kohno K, Kuwano M, Nakano H. Increased nuclear localization of transcription factor YB-1 in acquired cisplatin-resistant ovarian cancer. J Cancer Res Clin Oncol. 2002 Nov;128(11):621-6 Bergmann S, Royer-Pokora B, Fietze E, Jürchott K, Hildebrandt B, Trost D, Leenders F, Claude JC, Theuring F, Bargou R, Dietel M, Royer HD. YB-1 provokes breast cancer through the induction of chromosomal instability that emerges from mitotic failure and centrosome amplification. Cancer Res. 2005 May 15;65(10):4078-87 Bader AG, Felts KA, Jiang N, Chang HW, Vogt PK. Y boxbinding protein 1 induces resistance to oncogenic transformation by the phosphatidylinositol 3-kinase pathway. Proc Natl Acad Sci U S A. 2003 Oct 14;100(21):12384-9 Lu ZH, Books JT, Ley TJ. YB-1 is important for late-stage embryonic development, optimal cellular stress responses, and the prevention of premature senescence. Mol Cell Biol. 2005 Jun;25(11):4625-37 Kohno K, Izumi H, Uchiumi T, Ashizuka M, Kuwano M. The pleiotropic functions of the Y-box-binding protein, YB-1. Bioessays. 2003 Jul;25(7):691-8 Matsumoto K, Bay BH. Significance of the Y-box proteins in human cancers. J Mol Genet Med. 2005 Aug 3;1(1):11-7 Kuwano M, Uchiumi T, Hayakawa H, Ono M, Wada M, Izumi H, Kohno K. The basic and clinical implications of ABC transporters, Y-box-binding protein-1 (YB-1) and angiogenesisrelated factors in human malignancies. Cancer Sci. 2003 Jan;94(1):9-14 Sorokin AV, Selyutina AA, Skabkin MA, Guryanov SG, Nazimov IV, Richard C, Th'ng J, Yau J, Sorensen PH, Ovchinnikov LP, Evdokimova V. Proteasome-mediated cleavage of the Y-box-binding protein 1 is linked to DNAdamage stress response. EMBO J. 2005 Oct 19;24(20):360212 Li J, Hawkins IC, Harvey CD, Jennings JL, Link AJ, Patton JG. Regulation of alternative splicing by SRrp86 and its interacting proteins. Mol Cell Biol. 2003 Nov;23(21):7437-47 Oda Y, Ohishi Y, Saito T, Hinoshita E, Uchiumi T, Kinukawa N, Iwamoto Y, Kohno K, Kuwano M, Tsuneyoshi M. Nuclear expression of Y-box-binding protein-1 correlates with Pglycoprotein and topoisomerase II alpha expression, and with poor prognosis in synovial sarcoma. J Pathol. 2003 Feb;199(2):251-8 Sutherland BW, Kucab J, Wu J, Lee C, Cheang MC, Yorida E, Turbin D, Dedhar S, Nelson C, Pollak M, Leighton Grimes H, Miller K, Badve S, Huntsman D, Blake-Gilks C, Chen M, Pallen CJ, Dunn SE. Akt phosphorylates the Y-box binding protein 1 at Ser102 located in the cold shock domain and affects the anchorage-independent growth of breast cancer cells. Oncogene. 2005 Jun 16;24(26):4281-92 Raffetseder U, Frye B, Rauen T, Jürchott K, Royer HD, Jansen PL, Mertens PR. Splicing factor SRp30c interaction with Y-box protein-1 confers nuclear YB-1 shuttling and alternative splice site selection. J Biol Chem. 2003 May 16;278(20):18241-8 Evdokimova V, Ruzanov P, Anglesio MS, Sorokin AV, Ovchinnikov LP, Buckley J, Triche TJ, Sonenberg N, Sorensen PH. Akt-mediated YB-1 phosphorylation activates translation of silent mRNA species. Mol Cell Biol. 2006 Jan;26(1):277-92 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 603 YBX1 (Y box binding protein 1) Evdokimova V, Sorokin A Lu ZH, Books JT, Ley TJ. Cold shock domain family members YB-1 and MSY4 share essential functions during murine embryogenesis. Mol Cell Biol. 2006 Nov;26(22):8410-7 receptor or HER-2 across all tumor subtypes. Breast Cancer Res. 2008;10(5):R86 Oda Y, Kohashi K, Yamamoto H, Tamiya S, Kohno K, Kuwano M, Iwamoto Y, Tajiri T, Taguchi T, Tsuneyoshi M. Different expression profiles of Y-box-binding protein-1 and multidrug resistance-associated proteins between alveolar and embryonal rhabdomyosarcoma. Cancer Sci. 2008 Apr;99(4):726-32 Das S, Chattopadhyay R, Bhakat KK, Boldogh I, Kohno K, Prasad R, Wilson SH, Hazra TK. Stimulation of NEIL2mediated oxidized base excision repair via YB-1 interaction during oxidative stress. J Biol Chem. 2007 Sep 28;282(39):28474-84 Faury D, Nantel A, Dunn SE, Guiot MC, Haque T, Hauser P, Garami M, Bognár L, Hanzély Z, Liberski PP, Lopez-Aguilar E, Valera ET, Tone LG, Carret AS, Del Maestro RF, Gleave M, Montes JL, Pietsch T, Albrecht S, Jabado N. Molecular profiling identifies prognostic subgroups of pediatric glioblastoma and shows increased YB-1 expression in tumors. J Clin Oncol. 2007 Apr 1;25(10):1196-208 Shiota M, Izumi H, Onitsuka T, Miyamoto N, Kashiwagi E, Kidani A, Yokomizo A, Naito S, Kohno K. Twist promotes tumor cell growth through YB-1 expression. Cancer Res. 2008 Jan 1;68(1):98-105 Evdokimova V, Tognon C, Ng T, Ruzanov P, Melnyk N, Fink D, Sorokin A, Ovchinnikov LP, Davicioni E, Triche TJ, Sorensen PH. Translational activation of snail1 and other developmentally regulated transcription factors by YB-1 promotes an epithelial-mesenchymal transition. Cancer Cell. 2009 May 5;15(5):402-15 Kedersha N, Anderson P. Mammalian stress granules and processing bodies. Methods Enzymol. 2007;431:61-81 Nashchekin D, Masich S, Soop T, Kukalev A, Kovrigina E, Nashchekina O, Daneholt B. Two splicing isoforms of the Ybox protein ctYB-1 appear on the same mRNA molecule. FEBS J. 2007 Jan;274(1):202-11 Gluz O, Mengele K, Schmitt M, Kates R, Diallo-Danebrock R, Neff F, Royer HD, Eckstein N, Mohrmann S, Ting E, Kiechle M, Poremba C, Nitz U, Harbeck N. Y-box-binding protein YB-1 identifies high-risk patients with primary breast cancer benefiting from rapidly cycled tandem high-dose adjuvant chemotherapy. J Clin Oncol. 2009 Dec 20;27(36):6144-51 Schittek B, Psenner K, Sauer B, Meier F, Iftner T, Garbe C. The increased expression of Y box-binding protein 1 in melanoma stimulates proliferation and tumor invasion, antagonizes apoptosis and enhances chemoresistance. Int J Cancer. 2007 May 15;120(10):2110-8 Kashihara M, Azuma K, Kawahara A, Basaki Y, Hattori S, Yanagawa T, Terazaki Y, Takamori S, Shirouzu K, Aizawa H, Nakano K, Kage M, Kuwano M, Ono M. Nuclear Y-box binding protein-1, a predictive marker of prognosis, is correlated with expression of HER2/ErbB2 and HER3/ErbB3 in non-small cell lung cancer. J Thorac Oncol. 2009 Sep;4(9):1066-74 Vaiman AV, Stromskaya TP, Rybalkina EY, Sorokin AV, Ovchinnikov LP, Stavrovskaya AA. Development of drug resistance in the population of colon cancer cells under the effect of multifunctional protein YB-1. Bull Exp Biol Med. 2007 Apr;143(4):463-6 Ohashi S, Fukumura R, Higuchi T, Kobayashi S. YB-1 transcription in the postnatal brain is regulated by a bHLH transcription factor Math2 through an E-box sequence in the 5'UTR of the gene. Mol Cell Biochem. 2009 Jul;327(1-2):267-75 Chatterjee M, Rancso C, Stühmer T, Eckstein N, Andrulis M, Gerecke C, Lorentz H, Royer HD, Bargou RC. The Y-box binding protein YB-1 is associated with progressive disease and mediates survival and drug resistance in multiple myeloma. Blood. 2008 Apr 1;111(7):3714-22 Xu W, Zhou L, Qin R, Tang H, Shen H. Nuclear expression of YB-1 in diffuse large B-cell lymphoma: correlation with disease activity and patient outcome. Eur J Haematol. 2009 Oct;83(4):313-9 Fujii T, Kawahara A, Basaki Y, Hattori S, Nakashima K, Nakano K, Shirouzu K, Kohno K, Yanagawa T, Yamana H, Nishio K, Ono M, Kuwano M, Kage M. Expression of HER2 and estrogen receptor alpha depends upon nuclear localization of Y-box binding protein-1 in human breast cancers. Cancer Res. 2008 Mar 1;68(5):1504-12 Wachowiak R, Thieltges S, Rawnaq T, Kaifi JT, Fiegel H, Metzger R, Quaas A, Mertens PR, Till H, Izbicki JR. Y-boxbinding protein-1 is a potential novel tumour marker for neuroblastoma. Anticancer Res. 2010 Apr;30(4):1239-42 Habibi G, Leung S, Law JH, Gelmon K, Masoudi H, Turbin D, Pollak M, Nielsen TO, Huntsman D, Dunn SE. Redefining prognostic factors for breast cancer: YB-1 is a stronger predictor of relapse and disease-specific survival than estrogen Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) This article should be referenced as such: Evdokimova V, Sorokin A. YBX1 (Y box binding protein 1). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):598-604. 604 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review ZBTB33 (zinc finger and BTB domain containing 33) Michael R Dohn, Albert B Reynolds Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA (MRD, ABR) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/ZBTB33ID43785chXq24.html DOI: 10.4267/2042/45988 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology DNA/RNA Kaiso was originally identified in a yeast two-hybrid screen as a binding partner for the Armadillo repeat domain protein p120-catenin (CTNND1) and has subsequently been found to also interact with the p120catenin-related protein delta-catenin. The N-terminal POZ/BTB domain mediates Kaiso interactions with NCoR, the CTC-binding factor (CTCF), and Znf131, as well as Kaiso homodimerization. Description Expression DNA consists of three exons, the third of which contains the coding region. Kaiso is ubiquitously expressed. Transcription In various mammalian cell lines Kaiso localizes nearly exclusively to the nucleus, but in normal and tumor tissues Kaiso is predominantly detected in the cytoplasm. During mitosis a pool of Kaiso localizes to microtubles and centrosomes. Identity Other names: ZNF-kaiso; ZNF348 HGNC (Hugo): ZBTB33 Location: Xq24 Localisation Transcription of this gene produces transcript variants 1 (5324 bp) and 2 (5225 bp) that encode the same protein. Variant 2 lacks exon 2 in the 5' UTR. Pseudogene Function None. Via its zinc finger domain, Kaiso binds DNA and functions as both a repressor and activator of transcription. Kaiso recognizes methylated CpG dinucleotides as well as a sequence-specific site (TCCTGCNA). While several genes are repressed by Kaiso (including matrilysin, siamois, c-Fos, cyclin-D1, c-Myc, Wnt11, MMP-7 and MTA2), rapsyn is the only reported gene to be activated by Kaiso. Protein Description ZBTB33/Kaiso (hereafter Kaiso) is a member of the BTB/POZ (Broad complex, Tramtrak, Bric à brac/Pox virus and zinc finger)-zinc finger family of transcription factors. ZBTB33/Kaiso genomic sequence (7.64 Kb) is composed of three exons (green). The coding region (red) is within exon 3. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 605 ZBTB33 (zinc finger and BTB domain containing 33) Dohn MR, Reynolds AB ZBTB33/Kaiso contains an N-terminal POZ/BTB domain (green), two acidic regions (blue), and three C-terminal zinc finger domains (red). and non-canonical Wnt-signaling were thought to contribute to this phenotype, but subsequent studies determined that Kaiso's main role in early Xenopus development is restricted to the maintenence of transcriptional silencing. However, disruption of the Kaiso gene in mice did not reveal any abnormalities in development or gene expression. Homology Mus musculus - Zbtb33; Rattus norvegicus - Zbtb33; Xenopus laevis - zbtb33; Danio rerio - zbtb33; Pan troglodytes - ZBTB33; Bos Taurus - ZBTB33; Gallus gallus - ZBTB33. Mutations References Note None reported. Daniel JM, Reynolds AB. The catenin p120(ctn) interacts with Kaiso, a novel BTB/POZ domain zinc finger transcription factor. Mol Cell Biol. 1999 May;19(5):3614-23 Implicated in Prokhortchouk A, Hendrich B, Jørgensen H, Ruzov A, Wilm M, Georgiev G, Bird A, Prokhortchouk E. The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev. 2001 Jul 1;15(13):1613-8 Lung cancer Note Immunohistochemical analysis of 294 cases of nonsmall cell lung cancer, including 50 cases of paired lymph node metastases, revealed a correlation of cytoplasmic Kaiso staining with poor prognosis, and shRNA-mediated knockdown of Kaiso enhances proliferative and invasive capabilities of several lung cancer cell lines. Daniel JM, Spring CM, Crawford HC, Reynolds AB, Baig A. The p120(ctn)-binding partner Kaiso is a bi-modal DNA-binding protein that recognizes both a sequence-specific consensus and methylated CpG dinucleotides. Nucleic Acids Res. 2002 Jul 1;30(13):2911-9 Yoon HG, Chan DW, Reynolds AB, Qin J, Wong J. N-CoR mediates DNA methylation-dependent repression through a methyl CpG binding protein Kaiso. Mol Cell. 2003 Sep;12(3):723-34 Colon cancer Note Kaiso has been shown to repress expression of methylated tumor suppressor genes, and depletion of Kaiso sensitizes colon cancer cell lines to chemotherapy. Moreover, Kaiso is upregulated in intestinal tumors in mice, and a delayed onset of intestinal tumorigenesis is observed when Kaiso-null mice are crossed with the tumor-susceptible ApcMin/+ mice. Kim SW, Park JI, Spring CM, Sater AK, Ji H, Otchere AA, Daniel JM, McCrea PD. Non-canonical Wnt signals are modulated by the Kaiso transcriptional repressor and p120catenin. Nat Cell Biol. 2004 Dec;6(12):1212-20 Rodova M, Kelly KF, VanSaun M, Daniel JM, Werle MJ. Regulation of the rapsyn promoter by kaiso and delta-catenin. Mol Cell Biol. 2004 Aug;24(16):7188-96 Ruzov A, Dunican DS, Prokhortchouk A, Pennings S, Stancheva I, Prokhortchouk E, Meehan RR. Kaiso is a genome-wide repressor of transcription that is essential for amphibian development. Development. 2004 Dec;131(24):6185-94 Gastric cancer Note A study of the Helicobacter pylori-induced inflammatory response, which, if persistent, increases the risk for gastric adenocarcinoma, revealed that H. pylori induces nuclear translocation of the Kaisobinding partner p120-catenin. Nuclear p120-catenin then relieves Kaiso-mediated repression of MMP-7, which is often overexpressed in premalignant and malignant gastric lesions. Defossez PA, Kelly KF, Filion GJ, Pérez-Torrado R, Magdinier F, Menoni H, Nordgaard CL, Daniel JM, Gilson E. The human enhancer blocker CTC-binding factor interacts with the transcription factor Kaiso. J Biol Chem. 2005 Dec 30;280(52):43017-23 Park JI, Kim SW, Lyons JP, Ji H, Nguyen TT, Cho K, Barton MC, Deroo T, Vleminckx K, Moon RT, McCrea PD. Kaiso/p120-catenin and TCF/beta-catenin complexes coordinately regulate canonical Wnt gene targets. Dev Cell. 2005 Jun;8(6):843-54 Vertebrate development Soubry A, van Hengel J, Parthoens E, Colpaert C, Van Marck E, Waltregny D, Reynolds AB, van Roy F. Expression and nuclear location of the transcriptional repressor Kaiso is regulated by the tumor microenvironment. Cancer Res. 2005 Mar 15;65(6):2224-33 Note Injection of Xenopus laevis embryos with morpholinos targeting xKaiso leads to a developmental delay during gastrulation. Initially, Kaiso's roles in both general transcription repression and in regulation of canonical Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Spring CM, Kelly KF, O'Kelly I, Graham M, Crawford HC, Daniel JM. The catenin p120ctn inhibits Kaiso-mediated 606 ZBTB33 (zinc finger and BTB domain containing 33) Dohn MR, Reynolds AB transcriptional repression of the beta-catenin/TCF target gene matrilysin. Exp Cell Res. 2005 May 1;305(2):253-65 poor prognosis in non-small cell lung cancer. BMC Cancer. 2009 Jun 9;9:178 Prokhortchouk A, Sansom O, Selfridge J, Caballero IM, Salozhin S, Aithozhina D, Cerchietti L, Meng FG, Augenlicht LH, Mariadason JM, Hendrich B, Melnick A, Prokhortchouk E, Clarke A, Bird A. Kaiso-deficient mice show resistance to intestinal cancer. Mol Cell Biol. 2006 Jan;26(1):199-208 Ruzov A, Savitskaya E, Hackett JA, Reddington JP, Prokhortchouk A, Madej MJ, Chekanov N, Li M, Dunican DS, Prokhortchouk E, Pennings S, Meehan RR. The nonmethylated DNA-binding function of Kaiso is not required in early Xenopus laevis development. Development. 2009 Mar;136(5):729-38 Daniel JM. Dancing in and out of the nucleus: p120(ctn) and the transcription factor Kaiso. Biochim Biophys Acta. 2007 Jan;1773(1):59-68 Donaldson NS, Nordgaard CL, Pierre CC, Kelly KF, Robinson SC, Swystun L, Henriquez R, Graham M, Daniel JM. Kaiso regulates Znf131-mediated transcriptional activation. Exp Cell Res. 2010 Jun 10;316(10):1692-705 Lopes EC, Valls E, Figueroa ME, Mazur A, Meng FG, Chiosis G, Laird PW, Schreiber-Agus N, Greally JM, Prokhortchouk E, Melnick A. Kaiso contributes to DNA methylation-dependent silencing of tumor suppressor genes in colon cancer cell lines. Cancer Res. 2008 Sep 15;68(18):7258-63 Soubry A, Staes K, Parthoens E, Noppen S, Stove C, Bogaert P, van Hengel J, van Roy F.. The transcriptional repressor Kaiso localizes at the mitotic spindle and is a constituent of the pericentriolar material. PLoS One. 2010 Feb 15;5(2):e9203. Ogden SR, Wroblewski LE, Weydig C, Romero-Gallo J, O'Brien DP, Israel DA, Krishna US, Fingleton B, Reynolds AB, Wessler S, Peek RM Jr. p120 and Kaiso regulate Helicobacter pylori-induced expression of matrix metalloproteinase-7. Mol Biol Cell. 2008 Oct;19(10):4110-21 This article should be referenced as such: Dohn MR, Reynolds AB. ZBTB33 (zinc finger and BTB domain containing 33). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):605-607. Dai SD, Wang Y, Miao Y, Zhao Y, Zhang Y, Jiang GY, Zhang PX, Yang ZQ, Wang EH. Cytoplasmic Kaiso is associated with Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 607 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication t(3;5)(p21;q32) Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH) Published in Atlas Database: December 2010 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t0305p21q32ID2139.html DOI: 10.4267/2042/45989 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology colony stimulating factor-1 (CSF1). Upon binding of CSF1, CSF1R tyrosine phosphorylation is induced leading to RAS/RAF/MAPK, PI3K/AKT/mTOR and JAK/STAT (specifically STAT1, STAT3, and STAT5) pathways activation. CSF1R activation by CSF1 results in increased growth, proliferation and differentiation (Fischer et al., 2008). Clinics and pathology Disease MKPL-1 cell line, established from a 66-year-old male patient with an acute megakaryoblastic leukemia (M7AML) and a karyotype apparently with -21,+3mar (Takeuchi et al., 1992), re-analysed for tyrosine kinase dysregulation (Gu et al., 2007). Result of the chromosomal anomaly Epidemiology Only one case to date. Hybrid gene Genes involved and proteins Description Fusion of RBM6 exon 2 to CSF1R exon 12; the reciprocal CSF1R-RBM6 was not detected. RBM6 Location 3p21 Protein From N-term to C-term, contains a BTB/POZ domain (mediates homomeric dimerization) and decamer repeat domains, responsible for multimerization/selfassociation of the protein, RRM1 and RRM2 (RNA recognition motif) domains, an octamer repeat, a C2H2 zinc finger, a nuclear localisation signal, and a G-patch (made of highly conserved glycines; may have RNA binding functions). RNA-binding protein. Binds poly(G). Splicing factor (Heath et al., 2010). Fusion protein Description The RBM6-CSF1R fusion protein consists of the amino terminal 36 amino acids of RBM6, fused to the carboxy terminal 399 amino acids of CSF1R, including a polymerisation domain of RBM6, and the tyrosine kinase domain of CSF1R. Oncogenesis Constitutive tyrosine kinase activation. References CSF1R Takeuchi S, Sugito S, Uemura Y, Miyagi T, Kubonishi I, Taguchi H, Enzan H, Ohtsuki Y, Miyoshi I. Acute megakaryoblastic leukemia: establishment of a new cell line (MKPL-1) in vitro and in vivo. Leukemia. 1992 Jun;6(6):588-94 Location 5q32 Protein Contains Ig-like domains (extracellular), a transmembrane domain, and a split tyrosine kinase domain (intracellular), from N-term to C-term. Transmembrane glycoprotein, receptor for the ligand Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Gu TL, Mercher T, Tyner JW, Goss VL, Walters DK, Cornejo MG, Reeves C, Popova L, Lee K, Heinrich MC, Rush J, Daibata M, Miyoshi I, Gilliland DG, Druker BJ, Polakiewicz RD. A novel fusion of RBM6 to CSF1R in acute megakaryoblastic leukemia. Blood. 2007 Jul 1;110(1):323-33 608 t(3;5)(p21;q32) Huret JL Fischer JA, Rossetti S, Sacchi N.. CSF1R (colony stimulating factor 1 receptor, formerly McDonough feline sarcoma viral (vfms) oncogene homolog). Atlas Genet Cytogenet Oncol Haematol. April 2008. http://AtlasGeneticsOncology.org/Genes/CSF1RID40161ch5q3 2.html nascent transcripts. Chromosome Res. 2010 Dec;18(8):85172. Epub 2010 Nov 18. This article should be referenced as such: Huret JL. t(3;5)(p21;q32). Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):608-609. Heath E, Sablitzky F, Morgan GT.. Subnuclear targeting of the RNA-binding motif protein RBM6 to splicing speckles and Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 609 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Deep Insight Section Role of HB-EGF in cancer Rosalyn M Adam Urological Diseases Research Center, Enders Research Building, Rm 1077, Children's Hospital Boston, 300 Longwood Avenue, Boston MA 02115, USA (RMA) Published in Atlas Database: November 2010 Online updated version : http://AtlasGeneticsOncology.org/Deep/HB-EGFInCancerID20090.html DOI: 10.4267/2042/45990 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology generate recognition motifs for interactors that mediate downstream signaling cascades. Among the ERBB proteins, ERBB2 is an orphan receptor with no known ligand, whereas ERBB3 lacks functional intrinsic tyrosine kinase activity. In addition, the EGF-like growth factors show specificity in ERBB binding, with some factors such as EGF, amphiregulin (AREG) and TGFα selective for ERBB1 but others such as HB-EGF and betacellulin (BTC) able to interact with ERBB1 and ERBB4. Consequently, as a result of differential intrinsic receptor activity, ligand selectivity and modulation of ligand availability by interactions with heparin, a wide range of downstream responses can be evoked following ligand-ERBB interaction (Citri and Yarden, 2006). Introduction Peptide growth factors regulate diverse processes from cell survival and proliferation to migration and programmed cell death. Due to their central role in growth regulation, growth factors are major players in the development and progression of cancer. Among this broad class of molecules, those comprising the EGFlike family are amongst the best characterized. In this Deep Insight, I will elaborate on the evidence implicating one such protein, heparin-binding EGF-like growth factor (HB-EGF), in tumor biology and how its activity may be targeted for therapeutic gain. HB-EGF: a member of the EGF-like growth factor family HB-EGF: structure, molecular interactions and function Heparin-binding EGF-like growth factor (HB-EGF) is a member of the epidermal growth factor (EGF)-like growth factor family of proteins that bind to and activate the EGF receptor (EGFR) and its associated receptors ERBB2, ERBB3 and ERBB4. The extended family comprises 15 members, all of which conform broadly to common structural framework centered around 6 cysteine residues in the sequence: CX7CX45CX10-13CXCX8GXRC. Disulphide bond formation between 3 pairs of cysteines gives rise to the characteristic 3-looped EGF-like motif that mediates high-affinity binding to receptors (reviewed in Wilson et al., 2009). The EGF-like growth factors achieve their effects through interaction with one or more ErbB receptor tyrosine kinases. The ERBB receptors are class I transmembrane proteins that homo- or heterodimerize following ligand binding and undergo autophosphorylation on defined tyrosine residues to Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) HB-EGF was originally identified as a secreted product of macrophages that was purified on the basis of its high affinity for heparin (Higashiyama et al., 1991). The gene is encoded on the long arm of chromosome 5, at 5q23, and gives rise to a 208 amino acid protein of 20-22 kDa in size. Determination of the primary peptide sequence revealed the presence in HB-EGF of an extended N-terminal domain that was absent in the prototypical EGFR ligands EGF and transforming growth factor-alpha (TGFα). Notably, the N-terminal sequence in HB-EGF is enriched in basic amino acids that are positively charged at physiological pH and enable interaction with negatively charged heparin sulphate proteoglycans both on the cell surface and in the extracellular matrix. HB-EGF is synthesized as a single pass membraneanchored precursor with a short cytoplasmic tail (Figure 1). 610 Role of HB-EGF in cancer Adam RM Figure 1. Molecular structure of HB-EGF. The figure illustrates in schematic form the secondary structure of HB-EGF. The protein possesses a short cytoplasmic region, a single transmembrane domain, and an ectodomain that harbors the 3-looped EGF-like motif characteristic of this growth factor family. Vertical arrows indicate primary sites of ectodomain cleavage. Molecules that interact with HBEGF are indicated in blue. Membrane-anchored proHB-EGF undergoes a number of post-translational modifications ranging from Olinked glycosylation of the N-terminal ectodomain, Nterminal truncations, phosphorylation of the cytoplasmic domain and regulated cleavage of the entire ectodomain. The significance of these modifications will be considered in subsequent sections. Among the EGFR ligands, proHB-EGF is notable for the number of proteins and other molecules with which it interacts, including transmembrane receptors, adhesion molecules, and transcriptional regulators as described below. (i) Transmembrane receptors: the best-studied functions of HB-EGF are as a ligand for the EGFR/ErbB1 and the related receptor ErbB4. Highaffinity interactions with receptors are mediated via the 3-looped EGF-like motif and result in receptor autophosphorylation and initiation of downstream signaling cascades. Interestingly, the biological effects evoked by HB-EGF binding to ErbB1 and ErbB4 are distinct, with the former typically promoting proliferation but the latter stimulating chemotaxis and migration (Elenius et al., 1997). Interactions between the membrane-anchored form of HB-EGF and ErbB receptors expressed on adjacent cells also mediate both cell survival (Singh et al., 2007) and intercellular adhesion functions (Raab et al., 1996; Paria et al., 1999). More recently, radioligand binding assays with 125 I-labeled HB-EGF using the breast cancer cell line MDA-MB 453 revealed the existence of a novel receptor that was subsequently identified as the metalloendopeptidase N-arginine dibasic convertase (NRDc). In that study, NRDc was found to enhance HB-EGF stimulated migration of tumor cells in an EGFR/Erb1-dependent manner (Nishi et al., 2001). (ii) Heparan sulphate proteoglycans (HSPGs): HB-EGF was purified from the conditioned medium of a Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) macrophage-like cell line on the basis of its high affinity for heparin (Higashiyama et al., 1991). Interaction of HB-EGF with heparin, in the form of HSPGs present on cell surfaces and in the extracellular matrix is known to enhance ErbB receptor binding affinity as well as bioactivity (Paria et al., 1999; Higashiyama et al., 1993). (iii) CD9 and integrins: CD9 is a member of the tetraspanin family of transmembrane proteins that interacts with membrane-anchored HB-EGF via its heparin-binding domain (Sakuma et al., 1997) and upregulates its ability to stimulate juxtacrine activation of the EGFR expressed on adjacent cells (Higashiyama et al., 1995). HB-EGF and CD9 were also found to be co-expressed in gastric cancers (Murayama et al., 2002), although the prognostic significance of these observations has not been determined. CD9 and HBEGF were also demonstrated to exist in complex with integrin α3β1 at sites of cell-cell junctions (Nakamura et al., 1995) where the multiprotein complex waspredicted to participate in cell-cell adhesion. (iv) Cytoplasmic tail interactors: several binding partners for the cytoplasmic domain of proHB-EGF were identified using either yeast 2-hybrid or coimmunoprecipitation strategies, including the cochaperone BAG-1 and the transcriptional repressors PLZF and Bcl6. Interaction between BAG-1 and proHB-EGF was found to augment the prosurvival function of proHB-EGF (Lin et al., 2001). Conversely, association between the C-terminal fragment of HBEGF that is liberated following ectodomain shedding, with PLZF or Bcl6 leads to nuclear export or degradation, respectively of the transcriptional repressors and a resulting inhibition of repressive activity (Nanba et al., 2003; Kinugasa et al., 2007; Hirata et al., 2009). The relevance of these interactions to cancer will be considered in more detail below. 611 Role of HB-EGF in cancer Adam RM Figure 2. Regulated processing and activity of HB-EGF. ProHB-EGF expressed on the plasma membrane undergoes ectodomain cleavage mediated by enzymes of the matrix metalloproteinase (MMP) and a disintegrin and metalloproteinase (ADAM) families (A). Release of the mature, soluble protein facilitates both autocrine (B) and paracrine (C) activation of ERBB receptors expressed on the same or adjacent cells, respectively. The membrane-anchored proHB-EGF can also activate ERBB receptors via juxtacrine signaling (D). In addition to release of the ectodomain, regulated cleavage releases the Cterminal cytoplasmic fragment, HB-EGF-C (E), which can translocate to the nucleus to effect either nuclear export (F) or degradation (G), respectively, of the transcriptional repressors PLZF or Bcl6. revealed no differences in processing with wild type MEFs (Weskamp et al., 2002) suggesting that there is some redundancy among ADAM factors that cleave proHB-EGF. Elegant studies using cells from mice deficient in specific ADAM family members identified ADAM-17/TACE as the primary mediator of HB-EGF cleavage (Sahin et al., 2004). Importantly, ADAM-17 itself is upregulated in a range of tumor types (Tanaka et al., 2005) and at least part of its association with tumor progression is likely to reflect increased processing of EGFR ligands including HB-EGF. Ectodomain shedding of HB-EGF has also been implicated in EGFR transactivation in tumor cells downstream of multiple discrete agonists including Gprotein coupled receptor activators (Filardo et al., 2000; Madarame et al., 2003; Schäfer et al., 2004a; Schäfer et al., 2004b; Yano et al., 2004; Itoh et al., 2005), Ser/Thr kinase activators (Ebi et al., 2010), ligands for gp130 cytokine receptors (Ogunwobi and Beales, 2008) and others. EGFR transactivation by GPCR-dependent HBEGF cleavage has been discussed recently (reviewed in Higashiyama et al., 2008) and will not be considered further here. Although much attention has focused on the fate of the soluble HB-EGF that is liberated following precursor cleavage, the C-terminal fragment of HB-EGF that remains following ectodomain shedding has also been shown to be functional, independently of proHB-EGF. HB-EGF-C, comprising both the transmembrane and cytoplasmic domains, was demonstrated to undergo nuclear translocation following cleavage of the HB- Regulated processing of proHBEGF Like all ERBB ligands, HB-EGF is synthesized as a membrane-anchored precursor that is trafficked to the plasma membrane and subsequently processed to yield the mature, soluble growth factors. Regulated processing of the HB-EGF precursor represents a critical control point in ligand function since it represents the conversion from a membrane-anchored, non-diffusible state to a diffusible protein that has a greatly expanded sphere of influence on surrounding cells and tissues (Figure 2). In addition to normal post-translational maturation of HB-EGF, regulation of precursor processing is highly relevant to cancer, since many of the enzymes that cleave HB-EGF and other EGFR ligands are themselves upregulated in cancer versus normal cells (Murphy, 2008). The signals and enzymes responsible for liberation of the HB-EGF ectodomain will be considered in the following sections. Several metalloproteinases have been implicated in ectodomain shedding of proHB-EGF including matrix metalloproteinase-3 (MMP-3) (Suzuki et al., 1997), MMP-7 (Yu et al., 2002), ADAM10 (Yan et al., 2002), ADAM12 (Asakura et al., 2002) and TNFα-converting enzyme (TACE)/ADAM17 (Sahin et al., 2004). In forced expression experiments, ADAM9 was demonstrated to promote both basal and TPAstimulated proHB-EGF processing (Izumi et al., 1998). However, subsequent evaluation of ADAM9-/- MEFs Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 612 Role of HB-EGF in cancer Adam RM EGF ectodomain (Nanba et al., 2003; Nanba et al., 2004; Toki et al., 2005). Detection of the C-terminal fragment of HB-EGF in nuclei was consistent with an earlier report from our group identifying nuclear localization of HB-EGF as a feature of aggressive disease in bladder cancer (Adam et al., 2003). Nuclear localization of HB-EGF-C was accompanied by nuclear export of the promyelocytic leukemia zinc finger (PLZF) protein, identified as an interactor for HB-EGFC by yeast 2-hybrid analysis (Nanba et al., 2003). PLZF is a sequence-specific transcriptional repressor and inhibitor of cell cycle transit that achieves its effect by binding via its zinc finger domains to the promoters of target genes such as cyclin A (Yeyati et al., 1999). Export from the nucleus therefore prevents it from exerting its inhibitory function, resulting in enhanced movement through the cell cycle (Nanba et al., 2003). Interestingly, Bcl6, another transcriptional repressor, was also demonstrated to interact with HBEGF-C by a similar mechanism. In contrast to PLZF, however, binding to HB-EGF-C led to Bcl6 degradation and attenuation of its negative regulatory activity (Kinugasa et al., 2007; Hirata et al., 2009). In light of the dual growth stimulatory effects of proHB-EGF cleavage, resulting in liberation of the HBEGF ectodomain that can promote autocrine and paracrine stimulation of tumor cells, and the HB-EGFC carboxyl terminal fragment, that induces cell cycle transit, attempts have been made to target both biological consequences pharmacologically to achieve tumor cell inhibition (Shimura et al., 2008). sufficiency in growth signals; (ii) limitless replicative potential; (iii) resistance to growth inhibitory signals; (iv) evasion of apoptosis; (v) ability to migrate, invade and metastasize; and (vi) ability to evoke sustained angiogenesis. Recently, it has been argued that the list should be updated to include inflammation as an additional hallmark of cancer (Colotta et al., 2009). In the following sections, we will consider how HB-EGF relates functionally these features. Self-sufficiency in growth signals and limitless replicative potential As a ligand for members of the ErbB family of receptor tyrosine kinases, it is well established that HB-EGF can promote proliferation of a wide range of cells, including tumor cells from diverse cancer types. HBEGF gene expression is a target of several oncogenes including v-jun (Fu et al., 1999), Raf and Ras (McCarthy et al., 1995), and can therefore mediate growth-promoting effects subsequent to oncogenic transformation. The growth promoting effects of HB-EGF are mediated largely, although not exclusively, through binding to ErbB receptors on the plasma membrane. HB-EGF binding to EGFR/ErbB1 activates downstream signaling that converges on the Raf/Ras/MEK/Erk and phosphoinositide-3-kinase (PI3K)/Akt pathways to promote survival and proliferation (reviewed in Yarden and Sliwkowski, 2001). However, recent studies have demonstrated receptor-independent activities for HBEGF-C, the C-terminal fragment of HB-EGF that remains after ectodomain cleavage. In particular HBEGF-C has been shown to inhibit the transcriptionrepressing capabilities of PLZF and Bcl6 through either nuclear export or degradation, respectively (Nanba et al., 2003; Kinugasa et al., 2007). This resulted in enhanced expression of cyclin A and cyclin D2, together with increased cell cycle transit. Resistance to growth inhibitory signals evasion of apoptosis HB-EGF has been implicated as a survival factor for multiple cell types exposed to growth inhibitory stimuli. One of the earliest demonstrations of HB-EGFmediated cell survival revealed that whereas proHBEGF could prevent TGFβ-induced apoptosis in hepatoma cells in culture, this function could not be replicated with soluble HB-EGF (Miyoshi et al., 1997). That study provided the first demonstration of discrete functions for the soluble and cell-associated forms of HB-EGF, a concept that has been borne out in many subsequent studies both in non-malignant and tumor cells (Takemura et al., 1997; Singh et al., 2007; Ray et al., 2009). It is important to appreciate that HB-EGF expressed by cells in the microenvironment has also been implicated in tumor cell survival. Circulating cells such as T lymphocytes and macrophages that infiltrate tumors have been demonstrated to secrete HB-EGF that can act on tumor cells as well as other components critical for HB-EGF in cancer HB-EGF expression is altered in a number of cancer types including bladder (Adam et al., 2003; Kramer et al., 2007), breast (Ito et al., 2001c; Yotsumoto et al., 2010), colon (Ito et al., 2001a), hepatic (Inui et al., 1994), ovarian (Miyamoto et al., 2004; Tanaka et al., 2005), pancreatic (Kobrin et al., 1994; Ito et al., 2001b) and prostate cancers (Freeman et al., 1998) as well as gliomas (Mishima et al., 1998). In addition to quantitative increases in its expression in tumor versus non-tumor tissue, HB-EGF has also been found to undergo qualitative changes including altered subcellular localization, and cleavage to release N- and C-terminal fragments that mediate oncogenic behaviors. Notably, although HB-EGF in cancer is typically expressed in epithelial cells, we and others have reported robust HB-EGF expression in the stroma (Freeman et al., 1998; Adam et al., 1999) and endothelium (Nolan-Stevaux et al., 2010) in certain organs that exerts profound paracrine effects on tumor cells. To understand the potential functions of HB-EGF in cancer, it is instructive to consider the defining characteristics of tumor cells In their seminal article published 10 years ago Hanahan and Weinberg defined six features or 'hallmarks' characteristic of tumor cells (Hanahan and Weinberg, 2000). These are: (i) self Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 613 Role of HB-EGF in cancer Adam RM neuroendocrine tumorigenesis (Nolan-Stevaux et al., 2010). In that study the authors identified discrete roles for HB-EGF expressed by tumor endothelial and perivascular cells, and TGFα released by cancer cells both of which act through the EGFR to promote angiogenesis and tumor cell survival/growth, respectively. Ability to migrate, invade and metastasize To exit the primary tumor and disseminate to distant sites in the body, tumor cells must acquire the ability to migrate, intravasate, survive in the circulation, extravasate and establish in the secondary site. HBEGF has been shown to promote prostate cancer cell migration and invasion both directly and as an intermediate in EGFR transactivation by G-protein coupled receptor agonists (Madarame et al., 2003; Schäfer et al., 2004a; Cáceres et al., 2008). HBEGF was also found to participate in the epithelialmesenchymal transition (EMT) in gastric and ovarian cancer cells (Yagi et al., 2008). Gastric cancer cells exposed to the pathogen Helicobacter pylori, displayed increased MMP-7- and gastrin-dependent HB-EGF shedding and induction of EMT-associated genes. Inhibition of either gastrin or MMP-7 in vitro, or gastrin in vivo suppressed expression of HB-EGF and EMT-associated genes (Yin et al., 2010). Treatment of ovarian cancer cells with recombinant HB-EGF reduced E-cadherin levels and upregulated expression of Snail, a key regulator of the EMT. Conversely RNAi-mediated silencing of Snail attenuated HB-EGF expression and release of HB-EGF into the medium. Together these findings led the authors to conclude that HB-EGF could promote ovarian cancer metastasis through induction of the EMT (Yagi et al., 2008). Interestingly, Wang and colleagues demonstrated opposing effects on E-cadherin expression in pancreatic cells by retention of HB-EGF on the membrane. In cells either expressing non-cleavable proHB-EGF or treated with an inhibitor of HB-EGF ectodomain shedding, E-cadherin levels were up-regulated as a result of inhibition of ZEB1, a transcriptional repressor for E-cadherin (Wang et al., 2007b). Increased Ecadherin not only attenuated cell motility, but also sensitized cells to chemotherapy-induced apoptosis. Regulation of neuroendocrine differentiation and inflammation Although not strictly defined as 'hallmarks' of cancer, HB-EGF is known to participate in two additional processes linked to development and progression of cancer, namely inflammation and neuroendocrine differentiation. In intestinal cells exposed to cytokines (Mehta and Besner, 2003) or intestinal tissue exposed to ischemia/reperfusion injury (Rocourt et al., 2007) HB-EGF was found to exert anti-inflammatory activity in part through downregulation of NFκB and the ensuing reduction in expression of pro-inflammatory cytokines. In contrast, however, HB-EGF expression by mesenchymal cells in the liver was upregulated in tumor expansion such as endothelial cells and pericytes (Blotnick et al., 1994; Peoples et al., 1995). The cytokine CXCL12 was shown to promote HB-EGF release from mononuclear phagocytes and subsequent activation of the EGFR/ErbB1 and initiation of prosurvival signaling in tumor cells (Rigo et al., 2010). This in turn stimulated release of the macrophage mitogen GM-CSF to further promote HB-EGF in a growth stimulatory loop. Resistance to growth inhibition and evasion of apoptosis are relevant not only to cancer initiation, where cells lose responsiveness to normal cell death signals, but also in the setting of cancer treatment where tumor cells develop resistance to cytotoxic agents. Several recent reports have identified HB-EGF as a key mediator of treatment resistance and several tumor types. Exposure of cancer cells to either conventional chemotherapy or treatment with small molecule inhibitors was found to upregulate HB-EGF expression, release and activation of the EGFR, thereby enhancing survival signaling (Johnson et al., 2005; Yotsumoto et al., 2010). Both transcriptional and posttranscriptional mechanisms have been proposed to account for increased HB-EGF levels, including AP1/NFkappaB-dependent transcription (Wang et al., 2007a; Sorensen et al., 2006) and enhanced mRNA stability contributing to upregulation of HB-EGF protein expression. Ability to evoke sustained angiogenesis Decades of work by Judah Folkman and colleagues led to the concept that tumor growth beyond a defined size is an angiogenesis-dependent process (reviewed in Bishop-Bailey, 2009) i.e. requiring the development of a tumor blood supply. Using bladder cancer cells stably expressing either soluble or membrane-anchored HBEGF, Ongusaha and colleagues demonstrated that HBEGF was a potent inducer of several oncogenic behaviors including growth and migration in vitro as well as xenograft growth and angiogenesis in vivo (Ongusaha et al., 2004). Consistent with distinct functions for soluble and membrane-anchored HBEGF, expression of non-cleavable proHB-EGF was unable to replicate the tumorigenic potential of either soluble or wild type proHB-EGF. The HB-EGF sheddase ADAM17 has also been implicated in pathological neovascularization. Studies in which ADAM17 was deleted conditionally in either endothelial cells or pericytes using tissue-specific promoters to drive Cre recombinase expression demonstrated that loss of ADAM17 expression specifically in endothelial cells attenuated growth of implanted tumor cells (Weskamp et al., 2002). Significantly, effects of ADAM17 ablation could be restored by administration of exogenous HB-EGF, consistent with a role for ADAM17-dependent release of HBEGF in regulation of angiogenesis. The proangiogenic function of HBEGF was verified in an independent study that employed the RIP1-Tag2 mouse model of pancreatic Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 614 Role of HB-EGF in cancer Adam RM migration and invasion in vitro and to diminish growth, promote apoptosis and suppress angiogenesis in xenografts in nude mice (Sanui et al., 2010; Miyamoto et al., 2004; Martarelli et al., 2009). In addition, enhanced antitumor activity of CRM197 has been observed in combination with conventional chemotherapeutic agents such as paclitaxel (Yagi et al., 2009; Sanui et al., 2010). Although the results of CRM197 combination chemotherapy are provocative, it is important to note that exposure of cells to chemotherapeutic agents has been shown to upregulate HB-EGF levels that may in turn promote resistance to chemotherapy (Wang et al., 2007a). However, by careful attention to scheduling of drug administration, chemotherapy-induced upregulation of HB-EGF could be exploited to sensitize tumor cells to HB-EGFtargeted agents. Based on its demonstrated bioactivity, CRM197 has been administered to patients with advanced, treatmentrefractory malignant disease (Buzzi et al., 2004). One potential limitation of this strategy is that many in the general population are immunized against diphtheria and therefore may have innate resistance to CRM197 delivered systemically. Nevertheless, objective antitumor activity was observed in a small proportion of patients, with 3 responses and stable disease in a further 6 patients. Moreover, CRM197 demonstrated reasonable bioavailability, and toxicity associated with the treatment was deemed acceptable. Although the effect of CRM197 in that study was modest, more recent demonstrations of enhanced bioactivity in combination with conventional chemotherapeutic agents (Yagi et al., 2009; Sanui et al., 2010) suggests CRM197 may still have utility as an anti-cancer agent. As noted earlier, cleavage of membrane-anchored proHB-EGF represents a major control point for regulation of HB-EGF bioactivity. Consequently, several groups have focused on this event as a means to inhibit HB-EGF-dependent regulation of tumor cell behavior. Fridman and colleagues described the identification of selective inhibitors that could prevent shedding of ERBB ligands in vitro and went on to demonstrate potent anti-tumor effects in a range of assays, including survival pathway activation and growth and survival of xenografts (Fridman et al., 2007). Although such inhibitors are inhibiting shedding of multiple EGF-like ligands, in addition to HB-EGF, these results suggest the potential for combined inhibition of ligand shedding and ERBB receptor activation with small molecule inhibitors. In addition to preventing release of soluble HB-EGF, pharmacological inhibition of MMP/ADAM activity using KB-R7785 also suppressed generation and nuclear translocation of the HB-EGF C-terminal fragment (Shimura et al., 2008). This resulted in growth arrest, induction of apoptosis and decreased expression of proliferation-associated genes. inflamed tissue and augmented by pro-inflammatory stimuli (Sagmeister et al., 2008). Moreover, increased HB-EGF expression contributed to enhanced DNA synthesis and mitogenesis in premalignant hepatocytes consistent with a facilitative role for HB-EGF in hepatocarcinogenesis. In certain cancer types, such as prostate cancer the presence of neuroendocrine differentiation is associated with more aggressive tumors and worse patient outcome (Slovin, 2006). Our group showed that HBEGF could drive the neuroendocrine phenotype in prostate cancer cells in vitro and in vivo (Kim et al., 2002; Adam et al., 2002). Notably, cells exposed to HB-EGF continued to traverse the cell cycle in contrast to previous reports showing inducers of NE differentiation promoting cell cycle arrest (Cox et al., 2000; Wang et al., 2004). Moreover, HB-EGF induced downregulation of androgen receptor (AR) expression in xenografts as well as AR expression and activity in vitro (Adam et al., 2002). Subsequent analysis revealed HB-EGF-mediated AR inhibition occurred through an mTOR-dependent mechanism involving cap-dependent mRNA translation (Cinar et al., 2005). HB-EGF as a therapeutic target In light of the involvement of HB-EGF in multiple aspects of tumor development, progression and metastasis, it is not surprising that attempts have been made to target it for therapeutic benefit. Promising targeting strategies include prevention of ligand binding to the EGFR, inhibition of proHBEGF cleavage and subsequent release of ectodomain and Cterminal fragments and exploitation of proHB-EGF as the receptor for diphtheria toxin. In this section, we will review the approaches used to inhibit HB-EGF and their potential for clinical use. In situations where HB-EGF is overexpressed in tumors, some of its effects can obviously be blocked in the presence of either function blocking anti-EGFR antibodies or small molecule inhibitors of the intrinsic kinase domain. However this topic has been covered in many excellent reviews (Laskin and Sandler, 2004; Jimeno and Hidalgo, 2005) and will not be considered further here. A number of studies have exploited the identity of proHB-EGF as the receptor for diphtheria toxin (DT) in human cells by treating cells, and in some cases patients, with a non-toxic DT mutant termed CRM197. CRM197 harbors a point mutation (G52E) in the DT A chain that diminishes its ability to perform the ADPribosylation of elongation factor 2 and inhibition of protein synthesis characteristic of wild type DT (Mekada and Uchida, 1985). Although CRM197 lacks the toxicity of DT, it nonetheless exerts potent growth inhibitory effects through binding to the EGF-like domain of cell surface and soluble HB-EGF (Mitamura et al., 1995; Kageyama et al., 2007). In experimental evaluation, CRM197 alone has been found to induce apoptosis and inhibit oncogenic behaviors such as Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 615 Role of HB-EGF in cancer Adam RM growth factor-like growth factor and basic fibroblast growth factor: a potential pathologic role. Proc Natl Acad Sci U S A. 1995 Jul 3;92(14):6547-51 Summary HB-EGF plays multiple important roles in cancer, and has been implicated in each of the hallmarks characteristic of neoplastic disease. Recent work has emphasized the potential for HB-EGF as a viable therapeutic target for certain cancer types, through a variety of strategies. Moreover, its unique identity among ERBB ligands as a receptor for DT/CRM197 may provide novel avenues for rational targeting in the context of cancer therapeutics. The future challenge will be to realize this potential and to translate our knowledge of this inimitable molecule into treatments that benefit humankind. Raab G, Kover K, Paria BC, Dey SK, Ezzell RM, Klagsbrun M. Mouse preimplantation blastocysts adhere to cells expressing the transmembrane form of heparin-binding EGF-like growth factor. Development. 1996 Feb;122(2):637-45 Elenius K, Paul S, Allison G, Sun J, Klagsbrun M. Activation of HER4 by heparin-binding EGF-like growth factor stimulates chemotaxis but not proliferation. EMBO J. 1997 Mar 17;16(6):1268-78 Miyoshi E, Higashiyama S, Nakagawa T, Hayashi N, Taniguchi N. Membrane-anchored heparin-binding epidermal growth factor-like growth factor acts as a tumor survival factor in a hepatoma cell line. J Biol Chem. 1997 May 30;272(22):1434955 References Sakuma T, Higashiyama S, Hosoe S, Hayashi S, Taniguchi N. CD9 antigen interacts with heparin-binding EGF-like growth factor through its heparin-binding domain. J Biochem. 1997 Aug;122(2):474-80 Mekada E, Uchida T. Binding properties of diphtheria toxin to cells are altered by mutation in the fragment A domain. J Biol Chem. 1985 Oct 5;260(22):12148-53 Higashiyama S, Abraham JA, Miller J, Fiddes JC, Klagsbrun M. A heparin-binding growth factor secreted by macrophagelike cells that is related to EGF. Science. 1991 Feb 22;251(4996):936-9 Suzuki M, Raab G, Moses MA, Fernandez CA, Klagsbrun M. Matrix metalloproteinase-3 releases active heparin-binding EGF-like growth factor by cleavage at a specific juxtamembrane site. J Biol Chem. 1997 Dec 12;272(50):31730-7 Higashiyama S, Abraham JA, Klagsbrun M. Heparin-binding EGF-like growth factor stimulation of smooth muscle cell migration: dependence on interactions with cell surface heparan sulfate. J Cell Biol. 1993 Aug;122(4):933-40 Takemura T, Kondo S, Homma T, Sakai M, Harris RC. The membrane-bound form of heparin-binding epidermal growth factor-like growth factor promotes survival of cultured renal epithelial cells. J Biol Chem. 1997 Dec 5;272(49):31036-42 Blotnick S, Peoples GE, Freeman MR, Eberlein TJ, Klagsbrun M. T lymphocytes synthesize and export heparin-binding epidermal growth factor-like growth factor and basic fibroblast growth factor, mitogens for vascular cells and fibroblasts: differential production and release by CD4+ and CD8+ T cells. Proc Natl Acad Sci U S A. 1994 Apr 12;91(8):2890-94 Freeman MR, Paul S, Kaefer M, Ishikawa M, Adam RM, Renshaw AA, Elenius K, Klagsbrun M. Heparin-binding EGFlike growth factor in the human prostate: synthesis predominantly by interstitial and vascular smooth muscle cells and action as a carcinoma cell mitogen. J Cell Biochem. 1998 Mar 1;68(3):328-38 Inui Y, Higashiyama S, Kawata S, Tamura S, Miyagawa J, Taniguchi N, Matsuzawa Y. Expression of heparin-binding epidermal growth factor in human hepatocellular carcinoma. Gastroenterology. 1994 Dec;107(6):1799-804 Izumi Y, Hirata M, Hasuwa H, Iwamoto R, Umata T, Miyado K, Tamai Y, Kurisaki T, Sehara-Fujisawa A, Ohno S, Mekada E. A metalloprotease-disintegrin, MDC9/meltrin-gamma/ADAM9 and PKCdelta are involved in TPA-induced ectodomain shedding of membrane-anchored heparin-binding EGF-like growth factor. EMBO J. 1998 Dec 15;17(24):7260-72 Kobrin MS, Funatomi H, Friess H, Buchler MW, Stathis P, Korc M. Induction and expression of heparin-binding EGF-like growth factor in human pancreatic cancer. Biochem Biophys Res Commun. 1994 Aug 15;202(3):1705-9 Mishima K, Higashiyama S, Asai A, Yamaoka K, Nagashima Y, Taniguchi N, Kitanaka C, Kirino T, Kuchino Y. Heparin-binding epidermal growth factor-like growth factor stimulates mitogenic signaling and is highly expressed in human malignant gliomas. Acta Neuropathol. 1998 Oct;96(4):322-8 Higashiyama S, Iwamoto R, Goishi K, Raab G, Taniguchi N, Klagsbrun M, Mekada E. The membrane protein CD9/DRAP 27 potentiates the juxtacrine growth factor activity of the membrane-anchored heparin-binding EGF-like growth factor. J Cell Biol. 1995 Mar;128(5):929-38 Adam RM, Borer JG, Williams J, Eastham JA, Loughlin KR, Freeman MR. Amphiregulin is coordinately expressed with heparin-binding epidermal growth factor-like growth factor in the interstitial smooth muscle of the human prostate. Endocrinology. 1999 Dec;140(12):5866-75 McCarthy SA, Samuels ML, Pritchard CA, Abraham JA, McMahon M. Rapid induction of heparin-binding epidermal growth factor/diphtheria toxin receptor expression by Raf and Ras oncogenes. Genes Dev. 1995 Aug 15;9(16):1953-64 Fu S, Bottoli I, Goller M, Vogt PK. Heparin-binding epidermal growth factor-like growth factor, a v-Jun target gene, induces oncogenic transformation. Proc Natl Acad Sci U S A. 1999 May 11;96(10):5716-21 Mitamura T, Higashiyama S, Taniguchi N, Klagsbrun M, Mekada E. Diphtheria toxin binds to the epidermal growth factor (EGF)-like domain of human heparin-binding EGF-like growth factor/diphtheria toxin receptor and inhibits specifically its mitogenic activity. J Biol Chem. 1995 Jan 20;270(3):1015-9 Paria BC, Elenius K, Klagsbrun M, Dey SK. Heparin-binding EGF-like growth factor interacts with mouse blastocysts independently of ErbB1: a possible role for heparan sulfate proteoglycans and ErbB4 in blastocyst implantation. Development. 1999 May;126(9):1997-2005 Nakamura K, Iwamoto R, Mekada E. Membrane-anchored heparin-binding EGF-like growth factor (HB-EGF) and diphtheria toxin receptor-associated protein (DRAP27)/CD9 form a complex with integrin alpha 3 beta 1 at cell-cell contact sites. J Cell Biol. 1995 Jun;129(6):1691-705 Yeyati PL, Shaknovich R, Boterashvili S, Li J, Ball HJ, Waxman S, Nason-Burchenal K, Dmitrovsky E, Zelent A, Licht JD. Leukemia translocation protein PLZF inhibits cell growth and expression of cyclin A. Oncogene. 1999 Jan 28;18(4):92534 Peoples GE, Blotnick S, Takahashi K, Freeman MR, Klagsbrun M, Eberlein TJ. T lymphocytes that infiltrate tumors and atherosclerotic plaques produce heparin-binding epidermal Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 616 Role of HB-EGF in cancer Adam RM Cox ME, Deeble PD, Bissonette EA, Parsons SJ. Activated 3',5'-cyclic AMP-dependent protein kinase is sufficient to induce neuroendocrine-like differentiation of the LNCaP prostate tumor cell line. J Biol Chem. 2000 May 5;275(18):13812-8 Yan Y, Shirakabe K, Werb Z. The metalloprotease Kuzbanian (ADAM10) mediates the transactivation of EGF receptor by G protein-coupled receptors. J Cell Biol. 2002 Jul 22;158(2):2216 Yu WH, Woessner JF Jr, McNeish JD, Stamenkovic I. CD44 anchors the assembly of matrilysin/MMP-7 with heparinbinding epidermal growth factor precursor and ErbB4 and regulates female reproductive organ remodeling. Genes Dev. 2002 Feb 1;16(3):307-23 Filardo EJ, Quinn JA, Bland KI, Frackelton AR Jr. Estrogeninduced activation of Erk-1 and Erk-2 requires the G proteincoupled receptor homolog, GPR30, and occurs via transactivation of the epidermal growth factor receptor through release of HB-EGF. Mol Endocrinol. 2000 Oct;14(10):1649-60 Adam RM, Danciu T, McLellan DL, Borer JG, Lin J, Zurakowski D, Weinstein MH, Rajjayabun PH, Mellon JK, Freeman MR. A nuclear form of the heparin-binding epidermal growth factorlike growth factor precursor is a feature of aggressive transitional cell carcinoma. Cancer Res. 2003 Jan 15;63(2):484-90 Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000 Jan 7;100(1):57-70 Ito Y, Higashiyama S, Takeda T, Okada M, Matsuura N. Bimodal expression of heparin-binding EGF-like growth factor in colonic neoplasms. Anticancer Res. 2001 MarApr;21(2B):1391-4 Madarame J, Higashiyama S, Kiyota H, Madachi A, Toki F, Shimomura T, Tani N, Oishi Y, Matsuura N. Transactivation of epidermal growth factor receptor after heparin-binding epidermal growth factor-like growth factor shedding in the migration of prostate cancer cells promoted by bombesin. Prostate. 2003 Nov 1;57(3):187-95 Ito Y, Higashiyama S, Takeda T, Yamamoto Y, Wakasa KI, Matsuura N. Expression of heparin-binding epidermal growth factor-like growth factor in pancreatic adenocarcinoma. Int J Pancreatol. 2001;29(1):47-52 Ito Y, Takeda T, Higashiyama S, Noguchi S, Matsuura N. Expression of heparin-binding epidermal growth factor-like growth factor in breast carcinoma. Breast Cancer Res Treat. 2001 May;67(1):81-5 Mehta VB, Besner GE. Inhibition of NF-kappa B activation and its target genes by heparin-binding epidermal growth factor-like growth factor. J Immunol. 2003 Dec 1;171(11):6014-22 Nanba D, Mammoto A, Hashimoto K, Higashiyama S. Proteolytic release of the carboxy-terminal fragment of proHBEGF causes nuclear export of PLZF. J Cell Biol. 2003 Nov 10;163(3):489-502 Lin J, Hutchinson L, Gaston SM, Raab G, Freeman MR. BAG1 is a novel cytoplasmic binding partner of the membrane form of heparin-binding EGF-like growth factor: a unique role for proHB-EGF in cell survival regulation. J Biol Chem. 2001 Aug 10;276(32):30127-32 Buzzi S, Rubboli D, Buzzi G, Buzzi AM, Morisi C, Pironi F. CRM197 (nontoxic diphtheria toxin): effects on advanced cancer patients. Cancer Immunol Immunother. 2004 Nov;53(11):1041-8 Nishi E, Prat A, Hospital V, Elenius K, Klagsbrun M. N-arginine dibasic convertase is a specific receptor for heparin-binding EGF-like growth factor that mediates cell migration. EMBO J. 2001 Jul 2;20(13):3342-50 Laskin JJ, Sandler AB. Epidermal growth factor receptor: a promising target in solid tumours. Cancer Treat Rev. 2004 Feb;30(1):1-17 Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001 Feb;2(2):127-37 Miyamoto S, Hirata M, Yamazaki A, Kageyama T, Hasuwa H, Mizushima H, Tanaka Y, Yagi H, Sonoda K, Kai M, Kanoh H, Nakano H, Mekada E. Heparin-binding EGF-like growth factor is a promising target for ovarian cancer therapy. Cancer Res. 2004 Aug 15;64(16):5720-7 Adam RM, Kim J, Lin J, Orsola A, Zhuang L, Rice DC, Freeman MR. Heparin-binding epidermal growth factor-like growth factor stimulates androgen-independent prostate tumor growth and antagonizes androgen receptor function. Endocrinology. 2002 Dec;143(12):4599-608 Nanba D, Toki F, Higashiyama S. Roles of charged amino acid residues in the cytoplasmic domain of proHB-EGF. Biochem Biophys Res Commun. 2004 Jul 23;320(2):376-82 Asakura M, Kitakaze M, Takashima S, Liao Y, Ishikura F, Yoshinaka T, Ohmoto H, Node K, Yoshino K, Ishiguro H, Asanuma H, Sanada S, Matsumura Y, Takeda H, Beppu S, Tada M, Hori M, Higashiyama S. Cardiac hypertrophy is inhibited by antagonism of ADAM12 processing of HB-EGF: metalloproteinase inhibitors as a new therapy. Nat Med. 2002 Jan;8(1):35-40 Ongusaha PP, Kwak JC, Zwible AJ, Macip S, Higashiyama S, Taniguchi N, Fang L, Lee SW. HB-EGF is a potent inducer of tumor growth and angiogenesis. Cancer Res. 2004 Aug 1;64(15):5283-90 Kim J, Adam RM, Freeman MR. Activation of the Erk mitogenactivated protein kinase pathway stimulates neuroendocrine differentiation in LNCaP cells independently of cell cycle withdrawal and STAT3 phosphorylation. Cancer Res. 2002 Mar 1;62(5):1549-54 Sahin U, Weskamp G, Kelly K, Zhou HM, Higashiyama S, Peschon J, Hartmann D, Saftig P, Blobel CP. Distinct roles for ADAM10 and ADAM17 in ectodomain shedding of six EGFR ligands. J Cell Biol. 2004 Mar 1;164(5):769-79 Schäfer B, Gschwind A, Ullrich A. Multiple G-protein-coupled receptor signals converge on the epidermal growth factor receptor to promote migration and invasion. Oncogene. 2004 Jan 29;23(4):991-9 Murayama Y, Miyagawa J, Shinomura Y, Kanayama S, Isozaki K, Yamamori K, Mizuno H, Ishiguro S, Kiyohara T, Miyazaki Y, Taniguchi N, Higashiyama S, Matsuzawa Y. Significance of the association between heparin-binding epidermal growth factorlike growth factor and CD9 in human gastric cancer. Int J Cancer. 2002 Apr 1;98(4):505-13 Schäfer B, Marg B, Gschwind A, Ullrich A. Distinct ADAM metalloproteinases regulate G protein-coupled receptorinduced cell proliferation and survival. J Biol Chem. 2004 Nov 12;279(46):47929-38 Weskamp G, Cai H, Brodie TA, Higashyama S, Manova K, Ludwig T, Blobel CP. Mice lacking the metalloproteasedisintegrin MDC9 (ADAM9) have no evident major abnormalities during development or adult life. Mol Cell Biol. 2002 Mar;22(5):1537-44 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Wang Q, Horiatis D, Pinski J. Interleukin-6 inhibits the growth of prostate cancer xenografts in mice by the process of neuroendocrine differentiation. Int J Cancer. 2004 Sep 10;111(4):508-13 617 Role of HB-EGF in cancer Adam RM Yano S, Macleod RJ, Chattopadhyay N, Tfelt-Hansen J, Kifor O, Butters RR, Brown EM. Calcium-sensing receptor activation stimulates parathyroid hormone-related protein secretion in prostate cancer cells: role of epidermal growth factor receptor transactivation. Bone. 2004 Sep;35(3):664-72 Singh AB, Sugimoto K, Harris RC. Juxtacrine activation of epidermal growth factor (EGF) receptor by membraneanchored heparin-binding EGF-like growth factor protects epithelial cells from anoikis while maintaining an epithelial phenotype. J Biol Chem. 2007 Nov 9;282(45):32890-901 Cinar B, De Benedetti A, Freeman MR. Post-transcriptional regulation of the androgen receptor by Mammalian target of rapamycin. Cancer Res. 2005 Apr 1;65(7):2547-53 Wang F, Liu R, Lee SW, Sloss CM, Couget J, Cusack JC. Heparin-binding EGF-like growth factor is an early response gene to chemotherapy and contributes to chemotherapy resistance. Oncogene. 2007 Mar 29;26(14):2006-16 Itoh Y, Joh T, Tanida S, Sasaki M, Kataoka H, Itoh K, Oshima T, Ogasawara N, Togawa S, Wada T, Kubota H, Mori Y, Ohara H, Nomura T, Higashiyama S, Itoh M. IL-8 promotes cell proliferation and migration through metalloproteinase-cleavage proHB-EGF in human colon carcinoma cells. Cytokine. 2005 Mar 21;29(6):275-82 Wang F, Sloss C, Zhang X, Lee SW, Cusack JC. Membranebound heparin-binding epidermal growth factor like growth factor regulates E-cadherin expression in pancreatic carcinoma cells. Cancer Res. 2007 Sep 15;67(18):8486-93 Jimeno A, Hidalgo M. Blockade of epidermal growth factor receptor (EGFR) activity. Crit Rev Oncol Hematol. 2005 Mar;53(3):179-92 Cáceres M, Tobar N, Guerrero J, Smith PC, Martínez J. c-junNH2JNK mediates invasive potential and EGFR activation by regulating the expression of HB-EGF in a urokinase-stimulated pathway. J Cell Biochem. 2008 Feb 15;103(3):986-93 Johnson FM, Saigal B, Donato NJ. Induction of heparin-binding EGF-like growth factor and activation of EGF receptor in imatinib mesylate-treated squamous carcinoma cells. J Cell Physiol. 2005 Nov;205(2):218-27 Higashiyama S, Iwabuki H, Morimoto C, Hieda M, Inoue H, Matsushita N. Membrane-anchored growth factors, the epidermal growth factor family: beyond receptor ligands. Cancer Sci. 2008 Feb;99(2):214-20 Tanaka Y, Miyamoto S, Suzuki SO, Oki E, Yagi H, Sonoda K, Yamazaki A, Mizushima H, Maehara Y, Mekada E, Nakano H. Clinical significance of heparin-binding epidermal growth factor-like growth factor and a disintegrin and metalloprotease 17 expression in human ovarian cancer. Clin Cancer Res. 2005 Jul 1;11(13):4783-92 Murphy G. The ADAMs: signalling scissors in the tumour microenvironment. Nat Rev Cancer. 2008 Dec;8(12):929-41 Ogunwobi OO, Beales IL. Leptin stimulates the proliferation of human oesophageal adenocarcinoma cells via HB-EGF and Tgfalpha mediated transactivation of the epidermal growth factor receptor. Br J Biomed Sci. 2008;65(3):121-7 Toki F, Nanba D, Matsuura N, Higashiyama S. Ectodomain shedding of membrane-anchored heparin-binding EGF like growth factor and subcellular localization of the C-terminal fragment in the cell cycle. J Cell Physiol. 2005 Mar;202(3):83948 Sagmeister S, Drucker C, Losert A, Grusch M, Daryabeigi A, Parzefall W, Rohr-Udilova N, Bichler C, Smedsrød B, Kandioler D, Grünberger T, Wrba F, Schulte-Hermann R, Grasl-Kraupp B. HB-EGF is a paracrine growth stimulator for early tumor prestages in inflammation-associated hepatocarcinogenesis. J Hepatol. 2008 Dec;49(6):955-64 Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level. Nat Rev Mol Cell Biol. 2006 Jul;7(7):505-16 Shimura T, Kataoka H, Ogasawara N, Kubota E, Sasaki M, Tanida S, Joh T. Suppression of proHB-EGF carboxy-terminal fragment nuclear translocation: a new molecular target therapy for gastric cancer. Clin Cancer Res. 2008 Jun 15;14(12):395665 Slovin SF. Neuroendocrine differentiation in prostate cancer: a sheep in wolf's clothing? Nat Clin Pract Urol. 2006 Mar;3(3):138-44 Sorensen BS, Ornskov D, Nexo E. The chemotherapeutic agent VP16 increases the stability of HB-EGF mRNA by a mechanism involving the 3'-UTR. Exp Cell Res. 2006 Nov 1;312(18):3651-8 Yagi H, Yotsumoto F, Miyamoto S. Heparin-binding epidermal growth factor-like growth factor promotes transcoelomic metastasis in ovarian cancer through epithelial-mesenchymal transition. Mol Cancer Ther. 2008 Oct;7(10):3441-51 Fridman JS, Caulder E, Hansbury M, Liu X, Yang G, Wang Q, Lo Y, Zhou BB, Pan M, Thomas SM, Grandis JR, Zhuo J, Yao W, Newton RC, Friedman SM, Scherle PA, Vaddi K. Selective inhibition of ADAM metalloproteases as a novel approach for modulating ErbB pathways in cancer. Clin Cancer Res. 2007 Mar 15;13(6):1892-902 Bishop-Bailey D. Tumour vascularisation: a druggable target. Curr Opin Pharmacol. 2009 Apr;9(2):96-101 Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009 Jul;30(7):1073-81 Kageyama T, Ohishi M, Miyamoto S, Mizushima H, Iwamoto R, Mekada E. Diphtheria toxin mutant CRM197 possesses weak EF2-ADP-ribosyl activity that potentiates its anti-tumorigenic activity. J Biochem. 2007 Jul;142(1):95-104 Hirata Y, Ogasawara N, Sasaki M, Mizushima T, Shimura T, Mizoshita T, Mori Y, Kubota E, Wada T, Tanida S, Kataoka H, Kamiya T, Higashiyama S, Joh T. BCL6 degradation caused by the interaction with the C-terminus of pro-HB-EGF induces cyclin D2 expression in gastric cancers. Br J Cancer. 2009 Apr 21;100(8):1320-9 Kinugasa Y, Hieda M, Hori M, Higashiyama S. The carboxylterminal fragment of pro-HB-EGF reverses Bcl6-mediated gene repression. J Biol Chem. 2007 May 18;282(20):14797806 Martarelli D, Pompei P, Mazzoni G. Inhibition of adrenocortical carcinoma by diphtheria toxin mutant CRM197. Chemotherapy. 2009;55(6):425-32 Kramer C, Klasmeyer K, Bojar H, Schulz WA, Ackermann R, Grimm MO. Heparin-binding epidermal growth factor-like growth factor isoforms and epidermal growth factor receptor/ErbB1 expression in bladder cancer and their relation to clinical outcome. Cancer. 2007 May 15;109(10):2016-24 Ray KC, Blaine SA, Washington MK, Braun AH, Singh AB, Harris RC, Harding PA, Coffey RJ, Means AL. Transmembrane and soluble isoforms of heparin-binding epidermal growth factor-like growth factor regulate distinct processes in the pancreas. Gastroenterology. 2009 Nov;137(5):1785-94 Rocourt DV, Mehta VB, Besner GE. Heparin-binding EGF-like growth factor decreases inflammatory cytokine expression after intestinal ischemia/reperfusion injury. J Surg Res. 2007 May 15;139(2):269-73 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 618 Role of HB-EGF in cancer Adam RM Wilson KJ, Gilmore JL, Foley J, Lemmon MA, Riese DJ 2nd. Functional selectivity of EGF family peptide growth factors: implications for cancer. Pharmacol Ther. 2009 Apr;122(1):1-8 promote cancer growth via a GM-CSF/HB-EGF paracrine loop that is enhanced by CXCL12. Mol Cancer. 2010 Oct 14;9:273 Sanui A, Yotsumoto F, Tsujioka H, Fukami T, Horiuchi S, Shirota K, Yoshizato T, Kawarabayashi T, Kuroki M, Miyamoto S. HB-EGF inhibition in combination with various anticancer agents enhances its antitumor effects in gastric cancer. Anticancer Res. 2010 Aug;30(8):3143-9 Yagi H, Yotsumoto F, Sonoda K, Kuroki M, Mekada E, Miyamoto S. Synergistic anti-tumor effect of paclitaxel with CRM197, an inhibitor of HB-EGF, in ovarian cancer. Int J Cancer. 2009 Mar 15;124(6):1429-39 Ebi M, Kataoka H, Shimura T, Kubota E, Hirata Y, Mizushima T, Mizoshita T, Tanaka M, Mabuchi M, Tsukamoto H, Tanida S, Kamiya T, Higashiyama S, Joh T. TGFβ induces proHBEGF shedding and EGFR transactivation through ADAM activation in gastric cancer cells. Biochem Biophys Res Commun. 2010 Nov 19;402(3):449-54 Yin Y, Grabowska AM, Clarke PA, Whelband E, Robinson K, Argent RH, Tobias A, Kumari R, Atherton JC, Watson SA. Helicobacter pylori potentiates epithelial:mesenchymal transition in gastric cancer: links to soluble HB-EGF, gastrin and matrix metalloproteinase-7. Gut. 2010 Aug;59(8):1037-45 Yotsumoto F, Oki E, Tokunaga E, Maehara Y, Kuroki M, Miyamoto S. HB-EGF orchestrates the complex signals involved in triple-negative and trastuzumab-resistant breast cancer. Int J Cancer. 2010 Dec 1;127(11):2707-17 Nolan-Stevaux O, Truitt MC, Pahler JC, Olson P, Guinto C, Lee DC, Hanahan D. Differential Contribution to Neuroendocrine Tumorigenesis of Parallel Egfr Signaling in Cancer Cells and Pericytes. Genes Cancer. 2010 Apr;1(2):125-141 This article should be referenced as such: Rigo A, Gottardi M, Zamò A, Mauri P, Bonifacio M, Krampera M, Damiani E, Pizzolo G, Vinante F. Macrophages may Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) Adam RM. Role of HB-EGF in cancer. Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7):610-619. 619 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 Role of HB-EGF in cancer Adam RM © ATLAS - ISSN 1768-3262 Atlas Genet Cytogenet Oncol Haematol. 2011; 15(7) 621
Similar documents
always the best profiles
avec une grande porte pour faciliter l’installation et accessoires injéctées en bi matériel pour une fermeture hermétique. À utiliser avec les congés d’angle standards. COAF-COPF 100/65 COAF-COPF 40
More information