Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
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, and also more traditional review articles (“deep insights”) on the above subjects and on surrounding topics. It also present 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 jlhuret@AtlasGeneticsOncology.org or Editorial@AtlasGeneticsOncology.org The Atlas of Genetics and Cytogenetics in Oncology and Haematology is published 4 times a year by ARMGHM, a non profit organisation. Philippe Dessen is the Database Director, and Alain Bernheim the Chairman of the on-line version (Gustave Roussy Institute – Villejuif – France). http://AtlasGeneticsOncology.org © ATLAS - ISSN 1768-3262 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, and also more traditional review articles (“deep insights”) on the above subjects and on surrounding topics. It also present 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 jlhuret@AtlasGeneticsOncology.org or Editorial@AtlasGeneticsOncology.org The Atlas of Genetics and Cytogenetics in Oncology and Haematology is published 4 times a year by ARMGHM, a non profit organisation. Philippe Dessen is the Database Director, and Alain Bernheim the Chairman of the on-line version (Gustave Roussy Institute – Villejuif – France). 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) Volume 5, Number 1, January - March 2001 Table of contents Gene Section GRAF (GTPase activating protein for Rho associated with FAK) Stig Bojesen, Arndt Borkhardt 1 HMGIY (high mobility group protein (non histone chromosomal) isoform I and Y) Gilka JF Gattas, Florence Pedeutour 3 PU.1 (hematopoietic transcription factor PU.1) Françoise Moreau-Gachelin 7 FGFR1 (Fibroblast Growth Factor Receptor 1) Marie-Josèphe Pébusque 9 MLL (myeloid/lymphoid or mixed lineage leukemia) Jay L Hess, Jean-Loup Huret 11 PRDX1 (peroxiredoxin 1) Jean-Loup Huret 14 PTCH1 patched homolog 1 (Drosophila) Erika Lindström, Rune Toftgård 16 TFF2 (TreFoil Factor 2) Catherine Tomasetto 19 FGFR1OP (FGFR1 oncogene partner) Marie-Josèphe Pébusque 21 MST1R (Macrophage stimulating 1 receptor) Debora Angeloni, Michael I Lerman 23 FIM (fused in myeloproliferative disorders) Marie-Josèphe Pébusque 27 Leukaemia Section t(1;3)(p36;q21) Pascale Cornillet-Lefebvre, Sylvie Daliphard, Stéphanie Struski 29 del(17p) in non-Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi 31 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS del(20q) in myeloid malignancies Chrystèle Bilhou-Nabera 33 Juvenile Chronic Myelogenous Leukemia (JCML) Jay L Hess 35 t(6;8)(q27;p12) Marie-Joséphe Pébusque 37 t(8;13)(p12;q12) Marie-Josèphe Pébusque, Nicholas CP Cross 39 t(9;12)(q34;p13) Nyla A Heerema 42 +3 or trisomy 3 in non Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi 44 11q23 rearrangements in leukaemia Jean-Loup Huret 46 del(11q) in non-Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi 50 del(7q) in non-Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi 52 t(1;14)(p22;q32) in non Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi 54 t(5;10)(q33;q21) Cristina Mecucci 55 -Y, Y loss in leukemia Daniel L, Van Dyke 56 Solid Tumour Section Nervous system: Astrocytic tumors Anne-Marie Capodano 58 Cancer Prone Disease Section Beckwith-Wiedemann syndrome Marcel Mannens 62 Hereditary breast cancer Kaija Holli 66 Variegated aneuploidy related to premature centromere division (PCD) Alberto Plaja 69 Hereditary pancreatic cancer Ralph H Hruban, Scott E Kern 72 Li-Fraumeni syndrome Jenny M Varley 76 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review GRAF (GTPase activating associated with FAK) protein for Rho Stig Bojesen, Arndt Borkhardt Department of Clinical Biochemistry, Herlev University Hospital, Herlev Ringvej 75, Herlev DK-2730, Denmark (SEB, AB) Published in Atlas Database: November 2000 Online updated version: http://AtlasGeneticsOncology.org/Genes/GRAFID291.html DOI: 10.4267/2042/37688 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Protein Other names: GRAF (GTPase activating protein for Rho associated with FAK); KIAA0621; OPHN1L HGNC (Hugo): ARHGAP26 Location: 5q31 Local order: Just centromeric of GRL. Description Isoform A: 759 amino acids, 86 kDa; isoform B: 814 amino acids, 92 kDa. Expression At least 24 exons. Highly expressed in epithelial tissues i.e. pancreas islet beta-cells, testicles, prostate, mammary gland, GI glands, squamous layer of skin epithelium; highly expressed in nervous tissues including enteric ganglia; expressed in cardiomyocytes, erythropoiesis cells and liver. Transcription Localisation Two isoforms of 2277 bp (leukocytes) and 2442 bp (brain); transcripts of 4,4 and 9,5 kb. Mainly cytoplasmatic. DNA/RNA Description Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 1 GRAF (GTPase activating protein for Rho associated with FAK) Bojesen SE, Borkhardt A Function Implicated in Interacts with FAK and RhoA both in vivo and in vitro; acts as GTPase activating protein (GAP) for the active GTP-bound RhoA; negative regulator of RhoA. t(5;11)(q31;q23) / acute non lymphocytic leukemia --> MLL - GRAF Homology Prognosis Unknown; only a few cases. Hybrid/Mutated gene 5' MLL 3' GRAF Abnormal protein MLL-GRAF Oligophrenin-1, Beta-chimerin, BCR. Mutations Germinal Not known. Acute non lymphocytic leukemia and myelodysplastic syndrome with del(5q) Somatic Deletion of four bases (251-254, A in ATG=nt1) Prognosis Unknown. Cytogenetics del(5q). Oncogenesis Basically unknown; a bi-allelic loss of GRAF has been documented in three cases of ANLL. Insert 1158. GRAF-base 1144, 5' 1 TA GAG ACA GGA TTT CAT CAT GTT GGC CAG GTT GGT TTT GAA 42 TTC CTG ACC TCA AGT GAT CCA CCT GCC TCG GCC TCC CAA AGT 84 GGT GGG ATT TTG G 3' ......GRAF-base 1145 References Insert 1299 GRAF-base 1285, 5' 1 TC ATC GTT GTC ATA TAA ATC GGC GAG GTA ATA TTC CAT CAG 42 GTA GAC ATA CG 3' ...GRAF-base 1286. Predicted STOP codon underlined. Borkhardt A, Bojesen S, Haas OA, Fuchs U, Bartelheimer D, Loncarevic IF, Bohle RM, Harbott J, Repp R, Jaeger U, Viehmann S, Henn T, Korth P, Scharr D, Lampert F. The human GRAF gene is fused to MLL in a unique t(5;11)(q31;q23) and both alleles are disrupted in three cases of myelodysplastic syndrome/acute myeloid leukemia with a deletion 5q. Proc Natl Acad Sci U S A. 2000 Aug 1;97(16):9168-73 Insert 2002 GRAF-base 1988 5' 1 G GTT CAT GCG AGT TCA GCA AGC AGT TAC CAT GTC TAC GGC 41 ATG CCA GGA TAC TGT TGG GAA GGT AGT ATT CCG T 3' ...GRAF-base 1989 This article should be referenced as such: Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Bojesen SE, Borkhardt A. GRAF (GTPase activating protein for Rho associated with FAK). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):1-2. 2 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review HMGIY (high mobility group protein (non histone chromosomal) isoform I and Y) Gilka JF Gattas, Florence Pedeutour UF Recherche Clinique 952, Laboratoire de Génétique, Université de Nice-Sophia Antipolis, CHU de Nice, 06202 Nice, France (GJFG, FP) Published in Atlas Database: November 2000 Online updated version: http://AtlasGeneticsOncology.org/Genes/HMGIYID221.html DOI: 10.4267/2042/37689 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology not transcribed, exons 5-7 encode three DNA binding domains; exon 5 contains a 33 bp segment subject to alternative splicing; exon 8 encodes the acidic carboxy-terminal end; RNA length: 1.85 kb. Identity Other names:HMG-I(Y), HMGI/Y HGNC (Hugo): HMGA1 Location: 6p21.3 Local order: centromeric to HLA-A, telomeric to D6S19. Protein Description 107 amino acids; three DNA binding domains (AT hooks). Expression Expressed in embryonal cells; expressed in a variety of normal human adult tissues such as heart, brain, lung, skeletal muscle, kidney, pancreas, spleen, thymus, testis, ovary, small intestine, submandibular gland and leukocytes; expressed in transformed cells with a malignant phenotype and in human malignant tumors such as prostate, thyroid carcinoma and colorectal carcinomas and a subset of benign lipomas. Probe(s) - Courtesy Mariano Rocchi, Resources for Molecular Cytogenetics. DNA/RNA Description 10 144 bp; 8 exons, 7 introns. Localisation Transcription Nuclear. HMGI and HMGY are encoded by the same gene and are generated trough alternative splicing; exons 1-4 are Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 3 HMGIY (high mobility group protein (non histone chromosomal) isoform I and Y) translocations might be more complex than shown by conventional cytogenetics, with the presence of additional cryptic rearrangements; translocations involving partner chromosomes other than chromosome 14, such as chromosomes 1, 3, 4, 5, 10, 12, 17 have also been reported; inversions inv(6)(p21q21) or inv(6)(p21.3q26) have been described. Hybrid/Mutated gene In most cases, the breakpoint was extragenic, located within a 80 kb region 3' of HMGIY; aberrant transcripts with truncation of sequences from the 3' UTR have been described; in only one case with inversion inv(6)(p21q21), a hybrid intragenic fusion has been reported: HMGIY was fused to the LAMA4 (laminin a4 chain) gene. Abnormal protein The HMGIY-LAMA4 resulted from the fusion of the three HMGIY DNA-binding domains with the LAMA4 EGF-like domain. Oncogenesis The exact role of HMGI(Y)-LAMA4 fusion is not established yet. Function Architectural transcription, non histone, factor that binds to the minor groove of AT-rich DNA; alters DNA conformation by introducing bends and supercoils; HMGIY was shown to be an essential component of enhanceosome (higher order transcription enhancer complex); positive induction of several genes including IFN-b, E-selectin, interleukin-2 receptor a-chain, the chemokine MGSA/GRO, and the class II major histocompatibility complex gene HLADRA; negative regulation by binding the promoter regions of interleukin-4 and GP91-PHOX. The precise function remains to be elucidated; probable role in regulation of chromatin structure and gene expression, and transcriptional regulation; potential oncogenic role. Homology Member of the HMGI protein family, structural (but not expression pattern) homology with HMGIC. Lipomas Disease Benign adipocyte tumors. Prognosis Good. Cytogenetics A small subset (5-8%) of ordinary lipomas is characterized by 6p21 rearrangements, the most frequent of which being a reciprocal translocation t(3;6)(q28;p21); in contrast to other benign mesenchymal tumors with 6p21 rearrangement, there is no evidence of HMGIY rearrangements in ordinary lipomas yet; however, to be noticed, the breakpoint on 6p21 was shown to be located whithin a 80 kb region surrounding HMGIY in one lipoma case and HMGIY expression was correlated with 6p rearrangements in two ordinary lipomas and two spindle cell lipomas Mutations Somatic HMGIY is found rearrangements in mesenchymal tumors. involved in chromosome benign tumours, mainly Implicated in Pulmonary chondroid hamartoma Disease Benign tumor of the lung. Prognosis Good. Cytogenetics The most frequent rearrangement is a reciprocal balanced translocation t(6;14) (p21.3; q24); the rearrangement between chromosomes 6 and 14 can sometimes be complex, identifiable by FISH; molecular results also suggest that the Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Gattas GJF, Pedeutour F Uterine leiomyoma Disease Benign mesenchymal tumors. Prognosis Good. 4 HMGIY (high mobility group protein (non histone chromosomal) isoform I and Y) Cytogenetics One case with a t(1;6)(p21;21), involving the HMGIY gene has been described. Cytogenetics Approximately 40% of uterine leiomyomas present structural chromosomal rearrangements, 5% of which involve 6p abnormalities; they include t(1;6)(q23;p21), t(6;14)(p21;q24) and t(6;10)(p21;q22) as well as inversions and translocations involving other chromosomal partners; the rearrangements are sometimes complex, only identifiable by FISH analysis. Hybrid/Mutated gene No hybrid gene has been described yet; as for other mesenchymal tumors, the breakpoint was extragenic, located within a 80 kb region 3' of HMGIY; one case of aberrant transcript with truncation of 1295 bp from the 3' UTR has been described. Abnormal protein HMGIY mRNA and protein levels do not always correlate, suggesting that post-transcriptional mechanisms are involved in the regulation of HMGIY. Microfollicular adenoma of the thyroid Disease Epithelial tumors. Prognosis Favourable. Cytogenetics One case with a t(1;6)(p35;21) correlated with an overexpression of HMGIY has been described. References Friedmann M, Holth LT, Zoghbi HY, Reeves R. Organization, inducible-expression and chromosome localization of the human HMG-I(Y) nonhistone protein gene. Nucleic Acids Res. 1993 Sep 11;21(18):4259-67 Chiappetta G, Avantaggiato V, Visconti R, Fedele M, Battista S, Trapasso F, Merciai BM, Fidanza V, Giancotti V, Santoro M, Simeone A, Fusco A. High level expression of the HMGI (Y) gene during embryonic development. Oncogene. 1996 Dec 5;13(11):2439-46 Endometrial polyps Disease Uterine benign tumors. Prognosis Good. Cytogenetics Several chromosomal abnormalities involving the 6p21.3 region, including translocations, deletions, inversions have been described; various chromosomal partner regions, such as 14q24, 20q13, 2q35, 10q22, 8q12, 1p32, 7p15, 15q21, have been described to be associated with 6p21.3 in reciprocal translocations. Hybrid/Mutated gene No hybrid gene has been described yet; as for other mesenchymal tumors, the breakpoint is extragenic, located within a 80 kb region 3' of HMGIY. Dal Cin P, Wanschura S, Christiaens MR, Van den Berghe I, Moerman P, Polito P, Kazmierczak B, Bullerdiek J, Van den Berghe H. Hamartoma of the breast with involvement of 6p21 and rearrangement of HMGIY. Genes Chromosomes Cancer. 1997 Sep;20(1):90-2 Tallini G, Dal Cin P, Rhoden KJ, Chiapetta G, Manfioletti G, Giancotti V, Fusco A, Van den Berghe H, Sciot R. Expression of HMGI-C and HMGI(Y) in ordinary lipoma and atypical lipomatous tumors: immunohistochemical reactivity correlates with karyotypic alterations. Am J Pathol. 1997 Jul;151(1):37-43 Tkachenko A, Ashar HR, Meloni AM, Sandberg AA, Chada KK. Misexpression of disrupted HMGI architectural factors activates alternative pathways of tumorigenesis. Cancer Res. 1997 Jun 1;57(11):2276-80 Williams AJ, Powell WL, Collins T, Morton CC. HMGI(Y) expression in human uterine leiomyomata. Involvement of another high-mobility group architectural factor in a benign neoplasm. Am J Pathol. 1997 Mar;150(3):911-8 Hamartoma of the breast Disease Benign tumor-like nodule of the breast, also called adenolipoma. Prognosis Good. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Gattas GJF, Pedeutour F Xiao S, Lux ML, Reeves R, Hudson TJ, Fletcher JA. HMGI(Y) activation by chromosome 6p21 rearrangements in multilineage mesenchymal cells from pulmonary hamartoma. Am J Pathol. 1997 Mar;150(3):901-10 5 HMGIY (high mobility group protein (non histone chromosomal) isoform I and Y) Bandiera A, Bonifacio D, Manfioletti G, Mantovani F, Rustighi A, Zanconati F, Fusco A, Di Bonito L, Giancotti V. Expression of HMGI(Y) proteins in squamous intraepithelial and invasive lesions of the uterine cervix. Cancer Res. 1998 Feb 1;58(3):426-31 Klotzbücher M, Wasserfall A, Fuhrmann U. Misexpression of wild-type and truncated isoforms of the high-mobility group I proteins HMGI-C and HMGI(Y) in uterine leiomyomas. Am J Pathol. 1999 Nov;155(5):1535-42 Pedeutour F, Ligon AH, Morton CC. [Genetics of uterine leiomyomata]. Bull Cancer. 1999 Nov;86(11):920-8 Hess JL. Chromosomal translocations in benign tumors: the HMGI proteins. Am J Clin Pathol. 1998 Mar;109(3):251-61 Sornberger KS, Weremowicz S, Williams AJ, Quade BJ, Ligon AH, Pedeutour F, Vanni R, Morton CC. Expression of HMGIY in three uterine leiomyomata with complex rearrangements of chromosome 6. Cancer Genet Cytogenet. 1999 Oct 1;114(1):9-16 Kazmierczak B, Dal Cin P, Wanschura S, Borrmann L, Fusco A, Van den Berghe H, Bullerdiek J. HMGIY is the target of 6p21.3 rearrangements in various benign mesenchymal tumors. Genes Chromosomes Cancer. 1998 Dec;23(4):279-85 Martelli AM, Riccio M, Bareggi R, Manfioletti G, Tabellini G, Baldini G, Narducci P, Giancotti V. Intranuclear distribution of HMGI/Y proteins. An immunocytochemical study. J Histochem Cytochem. 1998 Jul;46(7):863-4 Yie J, Merika M, Munshi N, Chen G, Thanos D. The role of HMG I(Y) in the assembly and function of the IFN-beta enhanceosome. EMBO J. 1999 Jun 1;18(11):3074-89 Banks GC, Li Y, Reeves R. Differential in vivo modifications of the HMGI(Y) nonhistone chromatin proteins modulate nucleosome and DNA interactions. Biochemistry. 2000 Jul 18;39(28):8333-46 Shannon MF, Himes SR, Attema J. A role for the architectural transcription factors HMGI(Y) in cytokine gene transcription in T cells. Immunol Cell Biol. 1998 Oct;76(5):461-6 Dal Cin P, Fusco A, Belge G, Chiappetta G, Fedele M, Pauwels P, Bullerdiek J, Van den Berghe H. Involvement of the HMGI(Y) gene in a microfollicular adenoma of the thyroid. Genes Chromosomes Cancer. 1999 Mar;24(3):286-9 Tallini G, Vanni R, Manfioletti G, Kazmierczak B, Faa G, Pauwels P, Bullerdiek J, Giancotti V, Van Den Berghe H, Dal Cin P. HMGI-C and HMGI(Y) immunoreactivity correlates with cytogenetic abnormalities in lipomas, pulmonary chondroid hamartomas, endometrial polyps, and uterine leiomyomas and is compatible with rearrangement of the HMGI-C and HMGI(Y) genes. Lab Invest. 2000 Mar;80(3):359-69 Kazmierczak B, Meyer-Bolte K, Tran KH, Wöckel W, Breightman I, Rosigkeit J, Bartnitzke S, Bullerdiek J. A high frequency of tumors with rearrangements of genes of the HMGI(Y) family in a series of 191 pulmonary chondroid hamartomas. Genes Chromosomes Cancer. 1999 Oct;26(2):125-33 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Gattas GJF, Pedeutour F This article should be referenced as such: Gattas GJF, Pedeutour F. HMGIY (high mobility group protein (non histone chromosomal) isoform I and Y). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):3-6. 6 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Short Communication PU.1 (hematopoietic transcription factor PU.1) Françoise Moreau-Gachelin INSERM U528, Laboratoire de Transduction du Signal et Oncogenèse, Section de Recherche -Institut Curie, 26, rue d'Ulm, 75 248 Paris cedex 05, France (FMG) Published in Atlas Database: November 2000 Online updated version : http://AtlasGeneticsOncology.org/Genes/SPI1ID269.html DOI: 10.4267/2042/37690 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Expression Identity Hematopoietic specific; mainly in B lymphocytes and myeloid lineages; faintly expressed in erythroid progenitors. Other names: PU.1; SPI-1 HGNC (Hugo): SPI1 Location: 11p11-22 Localisation Nuclear. DNA/RNA Function Description Transcriptional regulator; specific DNA binding sites with a G/AGAA minimal element in promoter and enhancers of myeloid and B lymphoid genes; involved both in transcriptional regulation of genes and in splicing regulation of pre-mRNAs. Genomic locus around 50kb; 5 exons. Transcription 1,4kb. open reading frame (ORF): 991bp. Protein Homology ETS genes family. Description 264 amino acids. DNA binding protein with a ETS consensus motif. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 7 PU.1 (hematopoietic transcription factor PU.1) Moreau-Gachelin F Mutations References Germinal Moreau-Gachelin F, Tavitian A, Tambourin P. Spi-1 is a putative oncogene in virally induced murine erythroleukaemias. Nature. 1988 Jan 21;331(6153):277-80 Unknown. Somatic Klemsz MJ, McKercher SR, Celada A, Van Beveren C, Maki RA. The macrophage and B cell-specific transcription factor PU.1 is related to the ets oncogene. Cell. 1990 Apr 6;61(1):113-24 Unknown in human pathologies and cancers; insertional mutagenesis in murine erythroleukemia. Implicated in McKercher SR, Torbett BE, Anderson KL, Henkel GW, Vestal DJ, Baribault H, Klemsz M, Feeney AJ, Wu GE, Paige CJ, Maki RA. Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities. EMBO J. 1996 Oct 15;15(20):5647-58 Oncogenesis in mouse; mouse gene name: Spi-1 (SFFV Proviral Integration 1) Moreau-Gachelin F, Wendling F, Molina T, Denis N, Titeux M, Grimber G, Briand P, Vainchenker W, Tavitian A. Spi-1/PU.1 transgenic mice develop multistep erythroleukemias. Mol Cell Biol. 1996 May;16(5):2453-63 Disease Murine acute erythroleukemia induced by the Friend retrovirus SFFV (Spleen Focus Forming Virus). Prognosis 100%. Hybrid/Mutated gene No. Abnormal protein No; overexpression of the normal protein in the proerythroblast. Scott EW, Fisher RC, Olson MC, Kehrli EW, Simon MC, Singh H. PU.1 functions in a cell-autonomous manner to control the differentiation of multipotential lymphoid-myeloid progenitors. Immunity. 1997 Apr;6(4):437-47 Hallier M, Lerga A, Barnache S, Tavitian A, Moreau-Gachelin F. The transcription factor Spi-1/PU.1 interacts with the potential splicing factor TLS. J Biol Chem. 1998 Feb 27;273(9):4838-42 This article should be referenced as such: Moreau-Gachelin F. PU.1 (hematopoietic transcription factor PU.1). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):7-8. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 8 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review FGFR1 (Fibroblast Growth Factor Receptor 1) Marie-Josèphe Pébusque INSERM U119, IFR 57, 27 Blvd Lei Roure, 13009 Marseille, France (MJP) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Genes/FGFR1113.html DOI: 10.4267/2042/37691 This article is an update of: Huret JL. FGFR1 (fibroblast growth factor receptor 1). Atlas Genet Cytogenet Oncol Haematol.1998;2(2):35. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Other names: BFGFR (basic fibroblast growth factor receptor); FLT2 (FMS-like tyrosine kinase 2); FLG (FMS-like gene); CEK; FGFBR; N-SAM Location: 8p11 DNA/RNA FGFR1 (8p12) - Courtesy Mariano Rocchi, Resources for Molecular Cytogenetics. Transcription 2.7 mRNA. Protein Description 822 amino acids; 100-135 kDa glycoprotein from a 90-115 kDa Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 9 FGFR1 (Fibroblast Growth Factor Receptor 1) Pébusque MJ CEP110-FGFR1, and FIM-FGFR1; they encode large proteins containing the N-term of either FOP or CEP110, or FIM, and the catalytic domain of FGFR1 at their C-term: -N-term leucine-rich region from FOP fused to the catalytic domain of FGFR1; -N-term leucine zipper motifs from CEP110 fused to the catalytic domain of FGFR1; -N-term zinc fingers from FIM fused to the Tyrosine kinase domain of FGFR1in C-term. Oncogenesis Constitutive activation of FGFR1. protein core; tyrosine kinase receptor; contains four major domains: an extracellular domain with 2 or 3 Iglike loops, a transmembrane domain and an intracellular domain, a juxtamembrane domain, and an intracellular domain composed of the tyrosine kinase domain (two kinase domains interrupted by a short kinase insert), and a C-terminal tail. Localisation Plasma membrane. Function FGF receptor with tyrosine kinase activity; binding of ligand (FGF) in association with heparan sulfate proteoglycans induces receptor dimerization, autophosphorylation and signal transduction. Pfeiffer syndrome (inborn disease) Disease One form of Pfeiffer syndrome, an autosomal dominant craniosynostosis syndrome with broad thumbs and usually no mental deficiency, is due to a mutation in amino acid 252 (Pro252Arg substitution) of FGFR1. Homology With other FGFR (FGFR2, FGFR3, and FGFR4). Implicated in Breast cancer Stem-cell myeloproliferative disorder associated with chromosomal translocations involving 8p12; to date, seven FGFR1 partners have been described (see below) Disease Stem-cell myeloproliferative disorder characterized by T- or B-cell lymphoblastic leukemia/lymphoma, myeloid hyperplasia, and peripheral blood eosinophilia, and it generally progresses to acute myeloid leukemia; specific to the 8p12 chromosomal region. Prognosis Very poor (median survival: 12 months). Cytogenetics The 7 translocations are: -t(6;8)(q27; p12) involving FOP (FGFR1 Oncogene Partner); -t(8;9)(p12;q33) involving CEP110 (Centrosome protein 110); -t(8;11)(p12;p15); -t(8;12)(p12;q15); -t(8;13)(p12;q12) involving FIM (Fused In Myeloproliferative disorder also called ZNF198 or RAMP); -t(8;17)(p12;q25); -t(8;19)(p12;q13.3); additional anomalies: in the t(8;9)(p12;q33): +der(9), +21; in the t(8;13)(p12;q12): +8, +der(13), +21. Hybrid/Mutated gene -5' FOP - 3' FGFR1 in the t(6;8), -5' CEP110 - FGFR1 in the t(8;9), -5' FIM/ZNF198 - 3' FGFR1 in the t(8;13). Abnormal protein Three fusion transcripts are identified: FOP-FGFR1, Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Disease Gene amplification and overexpression in sporadic breast tumors. References Lee PL, Johnson DE, Cousens LS, Fried VA, Williams LT. Purification and complementary DNA cloning of a receptor for basic fibroblast growth factor. Science. 1989 Jul 7;245(4913):57-60 Itoh N, Terachi T, Ohta M, Seo MK. The complete amino acid sequence of the shorter form of human basic fibroblast growth factor receptor deduced from its cDNA. Biochem Biophys Res Commun. 1990 Jun 15;169(2):680-5 Johnson DE, Lu J, Chen H, Werner S, Williams LT. The human fibroblast growth factor receptor genes: a common structural arrangement underlies the mechanisms for generating receptor forms that differ in their third immunoglobulin domain. Mol Cell Biol. 1991 Sep;11(9):4627-34 Wennström S, Sandström C, Claesson-Welsh L. cDNA cloning and expression of a human FGF receptor which binds acidic and basic FGF. Growth Factors. 1991;4(3):197-208 Johnson DE, Williams LT. Structural and functional diversity in the FGF receptor multigene family. Adv Cancer Res. 1993;60:1-41 Webster MK, Donoghue DJ. FGFR activation in skeletal disorders: too much of a good thing. Trends Genet. 1997 May;13(5):178-82 Ugolini F, Adélaïde J, Charafe-Jauffret E, Nguyen C, Jacquemier J, Jordan B, Birnbaum D, Pébusque MJ. Differential expression assay of chromosome arm 8p genes identifies Frizzled-related (FRP1/FRZB) and Fibroblast Growth Factor Receptor 1 (FGFR1) as candidate breast cancer genes. Oncogene. 1999 Mar 11;18(10):1903-10 This article should be referenced as such: Pébusque MJ. FGFR1 (Fibroblast Growth Factor Receptor 1). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):9-10. 10 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review MLL (myeloid/lymphoid leukemia) or mixed lineage Jay L Hess, Jean-Loup Huret Department of Pathology, The University of Michigan, M5240 Medical Science I, 1301 Catherine Avenue, Ann Arbor, MI 48109-0602, USA (JLH), Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (J-LH) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Genes/MLL.html DOI: 10.4267/2042/37692 This article is an update of: Huret JL. MLL (myeloid/lymphoid or mixed lineage leukemia). Atlas Genet Cytogenet Oncol Haematol.1997;1(2):68-69. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Expression Identity Wide; especially in: brain, kidney, thyroid; expressed in Taned B lymphocytes and myeloid cells. Other names: ALL1; HRX; Htrx (human trithorax); TRX1 HGNC (Hugo): MLL Location: 11q23 Local order: Telomeric to PLZF, centromeric from RCK. Localisation Nuclear, in punctate spots. Function Transcriptional regulatory factor, maintenance of Hox gene expression. DNA/RNA involved in Homology Description Trithorax (Drosophila), ALR (human), MLL2 (human). 37 exons, spanning over 100 kb. Mutations Transcription Note MLL is implicated in at least 10 % of acute leukemias (AL) of various types: acute lymphoblastic leukemias (ALL), acute non lymphocytic leukemias (ANLL), biphenotypic ALs, treatment related leukemias, infant leukemias; the prognosis is poor. In a centromeric to telomeric direction; 13 and 15 kb; coding sequence: 11.9 kb. Protein Description 3969 amino acids; 431 kDa; contains two DNA binding motifs: a AT hook homologous to high mobility group proteins HMGI-(Y) and HMGI(C) that binds to the minor groove of DNA, and zinc fingers, a DNA methyl transferase motif, a bromodomain, and segments of homology with trithorax, in particular in the C-terminal SET domain. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Implicated in t(4;11)(q21;q23)/acute leukaemias --> MLL/AF4 Disease Typically CD19+ CD10-precursor B-ALL, biphenotypic AL, at times ANLL (M4/M5); common in 11 MLL (myeloid/lymphoid or mixed lineage leukemia) Hess JL, Huret JL infants may be congenital; treatment related leukaemia (secondary to epipodophyllotoxins). Prognosis Median survival < 1 year. Cytogenetics Additional chromosome anomalies are found in 1/4 of cases, one of which is the i(7q). Hybrid/Mutated gene 5' MLL - 3' AF4; 12 kb. Abnormal protein 240 kDa protein with about 1400 amino acids from NH2 MLL and 850 from COOH AF4 (variable breakpoints); the reciprocal may or may not be expressed. Abnormal protein AT hook and DNA methyltransferase from MLL fused to most of ELL. Oncogenesis Potential transcription factor. t(11;19)(q23;p13.3)/acute leukaemias --> MLL/ENL Disease ALL (CD19+), biphenotypic AL, ANLL (M4/M5); mainly congenital; treatment-related leukaemia. Prognosis Very poor, except in rare T-cell cases. Cytogenetics Detected with G banding. Hybrid/Mutated gene 5' MLL - 3' ENL Abnormal protein AT hook and DNA methyltransferase from MLL fused to, most often, the nearly entire ENL. t(6;11)(q27;q23)/ANLL --> MLL/AF6 Disease M5/M4 de novo and therapy related ANLL, T-cell ALL. Prognosis Poor. trisomy 11/ANLL --> MLL tandem duplication t(X;11)(q13;q23)/ANLL, T-ALL --> MLL/AFX1 t(X;11)(q22;q23)/ANLL --> MLL/Septin2 t(1;11)(p32;q23)/ALL --> MLL/AF1p t(1;11)(q21;q23)/ANLL --> MLL/AF1q t(2;11)(q11;q23)/MDS --> MLL/LAF4 t(3;11)(p21;q23)/t-ANLL --> MLL/AF3p21 t(3;11)(q25;q23)/t-ANLL --> MLL/GMPS ins(5;11)(q31;q13q23)/ALL --> MLL/AF5q31 t(5;11)(q31;q23)/ANLL --> MLL/GRAF t(6;11)(q21;q23)/ANLL --> MLL/AF6q21 t(9;11)(q34;q23)/ALL --> MLL/AF9q34 t(11;14)(q23;q24)/ANLL --> MLL/h-gephyrin t(11;15)(q23;q14)/ANLL --> MLL/AF15q14 t(11;16)(q23;p13)/t-ANLL --> MLL/CBP t(11;17)(q23;p13)/t-ANLL --> MLL/GAS7 t(11;17)(q23;q12)/ANLL --> MLL/RARa t(11;17)(q23;q21)/ANLL --> MLL/AF17 t(11;17)(q23;q25)/ANLL --> MLL/MSF/AF17q25 t(11;19)(q23;p13)/ANLL --> MLL/EEN t(9;11)(p22;q23)/ANLL --> MLL/AF9 Disease M5/M4 de novo and therapy related ANLL. Prognosis The prognosis may not be as poor as in other 11q23 leukaemias in de novo cases; very poor prognosis in secondary ANLL cases. Cytogenetics May be overlooked; often as a sole anomaly. Hybrid/Mutated gene Variable breakpoints on both genes. Abnormal protein N-term -- AT hook and DNA methyltransferase from MLL fused to the 192 C-term amino acids from AF9 (as breakpoints are variable, this is only an example). t(10;11)(p12;q23)/ANLL --> MLL/AF10 Disease M4 or M5 ANLL; ALL at times; therapy related ANLL. Prognosis Poor. t(11;19)(q23;p13.1)/ANLL --> MLL/ELL Disease Mainly M4/M5; treatment related leukemia; all ages. Prognosis Very poor. Cytogenetics Detected with R banding. Hybrid/Mutated gene 5' MLL - 3' ELL Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) t(11;22)(q23;q11.2)/ANLL --> MLL/hCDCRel t(11;22)(q23;q13)/ANLL --> MLL/P300 12 MLL (myeloid/lymphoid or mixed lineage leukemia) Hess JL, Huret JL Breakpoints Note Spanning a 8 kb genomic region; between exons 5 to 11; highly variable on the partner, ranging from close to the NH2term in ENL, to near the COOH-term in AF9. References Bernard OA, Berger R. Molecular basis of 11q23 rearrangements in hematopoietic malignant proliferations. Genes Chromosomes Cancer. 1995 Jun;13(2):75-85 Waring PM, Cleary ML. Disruption of a homolog of trithorax by 11q23 translocations: leukemogenic and transcriptional implications. Curr Top Microbiol Immunol. 1997;220:1-23 Schichman SA, Canaani E, Croce CM. Self-fusion of the ALL1 gene. A new genetic mechanism for acute leukemia. JAMA. 1995 Feb 15;273(7):571-6 Dimartino JF, Cleary ML. Mll rearrangements in haematological malignancies: lessons from clinical and biological studies. Br J Haematol. 1999 Sep;106(3):614-26 Rubnitz JE, Behm FG, Downing JR. 11q23 rearrangements in acute leukemia. Leukemia. 1996 Jan;10(1):74-82 Huntsman DG, Chin SF, Muleris M, Batley SJ, Collins VP, Wiedemann LM, Aparicio S, Caldas C. MLL2, the second human homolog of the Drosophila trithorax gene, maps to 19q13.1 and is amplified in solid tumor cell lines. Oncogene. 1999 Dec 23;18(56):7975-84 Young BD, Saha V. Chromosome abnormalities in leukaemia: the 11q23 paradigm. Cancer Surv. 1996;28:225-45 Prasad R, Zhadanov AB, Sedkov Y, Bullrich F, Druck T, Rallapalli R, Yano T, Alder H, Croce CM, Huebner K, Mazo A, Canaani E. Structure and expression pattern of human ALR, a novel gene with strong homology to ALL-1 involved in acute leukemia and to Drosophila trithorax. Oncogene. 1997 Jul 31;15(5):549-60 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Hess JL, Huret JL. MLL (myeloid/lymphoid or mixed lineage leukemia). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):11-13. 13 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review PRDX1 (peroxiredoxin 1) Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Genes/PAGID266.html DOI: 10.4267/2042/37693 This article is an update of: Prosperi MP, Ferbus D, Goubin G. PAG (Proliferation Associated Gene). Atlas Genet Cytogenet Oncol Haematol 2000;4(4):192 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Expression Identity Widely expressed, in particular in the various cell types of the central nervous system and in red blood cells; overexpressed following induction of proliferation and oxidative stress. Other names: PAGA (Proliferation Associated Gene A); PAG; PRX1; Hs.1163; NKEFA (natural killerenhancing factor A); Prx-1 (peroxiredoxin 1); HBP23 (Heme-binding protein 23 kDa); MSP23 (macrophage 23-kD stress protein) HGNC (Hugo): PRDX1 Location: 1p34.1 Note: PAGA/NKEFA/PRDX1/peroxiredoxin 1, located in 1p34, is often confused in databases and elsewhere with PAGB/TDPX2, a pseudogene located in 9p22; PAGB is not either NKEFB/PRDX2/peroxiredoxin 2, located in 13q12; the other peroxiredoxins, PRDX3 and PRDX5, are located in 10q25-26 and in 11q13 respectively. Localisation Cytosolic. Function Antioxidant, against oxidative stress; Abl SH3-binding protein; inhibitor of c-Abl tyrosine kinase activity; also binds to heme. Homology Thioperoxiredoxines. Implicated in DNA/RNA Disease Correlations between the expression level and the stage of tumor progrssion in squamous cell carcinoma of the oral cavity; high expression in follicular thyroid tumors, but not in papillary carcinoma of the thyroid. Description 6 exons, 13 kb. Transcription 937 bp mRNA; 599 bp coding sequence. References Pseudogene Prospéri MT, Ferbus D, Karczinski I, Goubin G. A human cDNA corresponding to a gene overexpressed during cell proliferation encodes a product sharing homology with amoebic and bacterial proteins. J Biol Chem. 1993 May 25;268(15):11050-6 Pseudogene in 9p22. Protein Description Prospéri MT, Apiou F, Dutrillaux B, Goubin G. Organization and chromosomal assignment of two human PAG gene loci: PAGA encoding a functional gene and PAGB a processed pseudogene. Genomics. 1994 Jan 15;19(2):236-41 199 amino acids; 22 kDa; form dimers through a disulfide bridge. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 14 PRDX1 (peroxiredoxin 1) Huret JL Shau H, Butterfield LH, Chiu R, Kim A. Cloning and sequence analysis of candidate human natural killer-enhancing factor genes. Immunogenetics. 1994;40(2):129-34 Hirotsu S, Abe Y, Okada K, Nagahara N, Hori H, Nishino T, Hakoshima T. Crystal structure of a multifunctional 2-Cys peroxiredoxin heme-binding protein 23 kDa/proliferationassociated gene product. Proc Natl Acad Sci U S A. 1999 Oct 26;96(22):12333-8 Shau H, Kim A. Identification of natural killer enhancing factor as a major antioxidant in human red blood cells. Biochem Biophys Res Commun. 1994 Feb 28;199(1):83-8 Sarafian TA, Verity MA, Vinters HV, Shih CC, Shi L, Ji XD, Dong L, Shau H. Differential expression of peroxiredoxin subtypes in human brain cell types. J Neurosci Res. 1999 Apr 15;56(2):206-12 Sauri H, Butterfield L, Kim A, Shau H. Antioxidant function of recombinant human natural killer enhancing factor. Biochem Biophys Res Commun. 1995 Mar 28;208(3):964-9 Sauri H, Ashjian PH, Kim AT, Shau H. Recombinant natural killer enhancing factor augments natural killer cytotoxicity. J Leukoc Biol. 1996 Jun;59(6):925-31 Yanagawa T, Ishikawa T, Ishii T, Tabuchi K, Iwasa S, Bannai S, Omura K, Suzuki H, Yoshida H. Peroxiredoxin I expression in human thyroid tumors. Cancer Lett. 1999 Oct 18;145(12):127-32 Wen ST, Van Etten RA. The PAG gene product, a stressinduced protein with antioxidant properties, is an Abl SH3binding protein and a physiological inhibitor of c-Abl tyrosine kinase activity. Genes Dev. 1997 Oct 1;11(19):2456-67 Mizusawa H, Ishii T, Bannai S. Peroxiredoxin I (macrophage 23 kDa stress protein) is highly and widely expressed in the rat nervous system. Neurosci Lett. 2000 Mar 31;283(1):57-60 Kang SW, Baines IC, Rhee SG. Characterization of a mammalian peroxiredoxin that contains one conserved cysteine. J Biol Chem. 1998 Mar 13;273(11):6303-11 Yanagawa T, Iwasa S, Ishii T, Tabuchi K, Yusa H, Onizawa K, Omura K, Harada H, Suzuki H, Yoshida H. Peroxiredoxin I expression in oral cancer: a potential new tumor marker. Cancer Lett. 2000 Aug 1;156(1):27-35 Prospéri MT, Ferbus D, Rouillard D, Goubin G. The pag gene product, a physiological inhibitor of c-abl tyrosine kinase, is overexpressed in cells entering S phase and by contact with agents inducing oxidative stress. FEBS Lett. 1998 Feb 13;423(1):39-44 This article should be referenced as such: Huret JL. PRDX1 (peroxiredoxin 1). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):14-15. Sarafian TA, Huang C, Kim A, de Vellis J, Shau H. Expression of the antioxidant gene NKEF in the central nervous system. Mol Chem Neuropathol. 1998 May;34(1):39-51 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 15 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review PTCH1 patched homolog 1 (Drosophila) Erika Lindström, Rune Toftgård Karolinska Institute, Department of Biosciences, NOVUM, Huddinge, Sweden (EL, RT) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Genes/PTCH100.html DOI: 10.4267/2042/37694 This article is an update of: Huret JL. PTCH (patched homolog). Atlas Genet Cytogenet Oncol Haematol.1999;3(2):57-58. Huret JL. PTCH (patched homolog). Atlas Genet Cytogenet Oncol Haematol.1997;1(1):1-2. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Function Identity Part of a signalling pathway; opposed by the gene products of hedgehog genes; transmembrane protein; is thought to have a repressive activity on cell proliferation; the recent demonstration of NBCCS syndrome (see below) as a chromosome instability syndrome suggests that this protein has a role in DNA maintenance, repair and/or replication. Other names: PTC, but this term was confusing with PTC/PKA; PTCH; patched HGNC (Hugo): PTCH1 Location: 9q22.3 Local order: (between FACC and XPAC PTCH1 is flanked by the microsatellite markers D9S196 and D9S287; a microsatellite marker, 1AJL, is located inside the gene. Homology Patched (drosophila segment polarity gene), PTCH2 (human gene with unknown function). DNA/RNA Mutations Description 24 exons, exon 24 is non-coding; 34 kb. Germinal Transcription Germ-line mutations lead to protein truncation in naevoid basal cell carcinoma syndrome (NBCCS) patients (see below); mutations types are variable: nucleotide substitutions (missense/nonsense), small deletions, or small insertions mainly, leading to protein truncation; these mutations have been observed in most exons; there is, so far, no hot-spot. Alternate splicing: 3 different 5' termini; 6.5 kb mRNA; coding sequence: CDS 1... 4344. Protein Description Somatic Glycoprotein; 12 transmembrane domains, 2 extra cellular loops, intracellular N-term and C-term and sterol-sensing domain (SSD). Mutation and allele loss events in basal cell carcinoma, in NBCCS and in sporadic basal cell carcinoma are, so far, in accordance with the two-hit model for neoplasia, as is found in retinoblastoma; mutation and allele loss have also been found in sporadic primitive neuroectodermal tumours (PNETs), sporadic medulloblastomas and in a few cases of esophageal squamous cell carcinoma and invasive transitional cell carcinoma of the bladder; mutations have also been reported in a low frequency of sporadic Expression Widely expressed at low levels; increased levels in cells receiving a hedgehog signal. Localisation Transmembrane protein, intracellular vesicles. cellular membrane, Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 16 PTCH1 patched homolog 1 (Drosophila) trichoepitheliomas keratocysts. and in sporadic Lindström E, Toftgård R Drosophila patched in sporadic basal cell carcinomas. Nat Genet. 1996 Sep;14(1):78-81 odontogenic Hahn H, Christiansen J, Wicking C, Zaphiropoulos PG, Chidambaram A, Gerrard B, Vorechovsky I, Bale AE, Toftgard R, Dean M, Wainwright B. A mammalian patched homolog is expressed in target tissues of sonic hedgehog and maps to a region associated with developmental abnormalities. J Biol Chem. 1996 May 24;271(21):12125-8 Implicated in Naevoid basal cell carcinoma syndrome (NBCCS) or Gorlin syndrome Disease Autosomal dominant condition; cancer prone disease (multiple basal cell carcinomas, medulloblastomas); malformations; it is also a chromosome instability syndrome. Cytogenetics Spontaneous and induced chromosome instability. Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley S, Chidambaram A, Vorechovsky I, Holmberg E, Unden AB, Gillies S, Negus K, Smyth I, Pressman C, Leffell DJ, Gerrard B, Goldstein AM, Dean M, Toftgard R, Chenevix-Trench G, Wainwright B, Bale AE. Mutations of the human homolog of Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell. 1996 Jun 14;85(6):841-51 Johnson RL, Rothman AL, Xie J, Goodrich LV, Bare JW, Bonifas JM, Quinn AG, Myers RM, Cox DR, Epstein EH Jr, Scott MP. Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science. 1996 Jun 14;272(5268):1668-71 Skin cancers Disease Sporadic basal cell carcinoma, but also in the benign trichoepithelioma, a tumor often associated with basal cell carcinomas. Sporadic basal cell carcinoma from xeroderma pigmentosum patients have a high frequency of typical UV-induced mutations in PTCH1. Lench NJ, Telford EA, High AS, Markham AF, Wicking C, Wainwright BJ. Characterisation of human patched germ line mutations in naevoid basal cell carcinoma syndrome. Hum Genet. 1997 Oct;100(5-6):497-502 Raffel C, Jenkins RB, Frederick L, Hebrink D, Alderete B, Fults DW, James CD. Sporadic medulloblastomas contain PTCH mutations. Cancer Res. 1997 Mar 1;57(5):842-5 Brain diseases Vorechovský I, Undén AB, Sandstedt B, Toftgård R, StåhleBäckdahl M. Trichoepitheliomas contain somatic mutations in the overexpressed PTCH gene: support for a gatekeeper mechanism in skin tumorigenesis. Cancer Res. 1997 Nov 1;57(21):4677-81 Disease In a subset of sporadic primitive neuroectodermal tumours (PNETs)of the central nervous system (cerebral PNETs, medulloblastomas, and desmoplastic medulloblastomas); note: NBCCS patients have a predisposition for the development of PNETs, while, herein mentioned are sporadic PNETs. PTCH1 have also been found mutated in both familiar and sporadic cases of Holoprosencephaly (HPE). Wicking C, Shanley S, Smyth I, Gillies S, Negus K, Graham S, Suthers G, Haites N, Edwards M, Wainwright B, ChenevixTrench G. Most germ-line mutations in the nevoid basal cell carcinoma syndrome lead to a premature termination of the PATCHED protein, and no genotype-phenotype correlations are evident. Am J Hum Genet. 1997 Jan;60(1):21-6 Wolter M, Reifenberger J, Sommer C, Ruzicka T, Reifenberger G. Mutations in the human homologue of the Drosophila segment polarity gene patched (PTCH) in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Res. 1997 Jul 1;57(13):2581-5 Various cancers and benign tumors Disease Invasive transitional cell carcinoma of the bladder: PTCH1 has been found mutated in rare cases. Sporadic esophageal squamous cell carcinoma Jaws: in sporadic odontogenic keratocysts and in odontogenic keratocysts from NBCCS patients. Xie J, Johnson RL, Zhang X, Bare JW, Waldman FM, Cogen PH, Menon AG, Warren RS, Chen LC, Scott MP, Epstein EH Jr. Mutations of the PATCHED gene in several types of sporadic extracutaneous tumors. Cancer Res. 1997 Jun 15;57(12):2369-72 References Tabata T, Eaton S, Kornberg TB. The Drosophila hedgehog gene is expressed specifically in posterior compartment cells and is a target of engrailed regulation. Genes Dev. 1992 Dec;6(12B):2635-45 Aszterbaum M, Rothman A, Johnson RL, Fisher M, Xie J, Bonifas JM, Zhang X, Scott MP, Epstein EH Jr. Identification of mutations in the human PATCHED gene in sporadic basal cell carcinomas and in patients with the basal cell nevus syndrome. J Invest Dermatol. 1998 Jun;110(6):885-8 Basler K, Struhl G. Compartment boundaries and the control of Drosophila limb pattern by hedgehog protein. Nature. 1994 Mar 17;368(6468):208-14 Bale SJ, Falk RT, Rogers GR. Patching together the genetics of Gorlin syndrome. J Cutan Med Surg. 1998 Jul;3(1):31-4 Louhelainen J, Lindström E, Hemminki K, Toftgård R. Dinucleotide repeat polymorphism within the tumor suppressor gene PTCH at 9q22. Clin Genet. 1998 Sep;54(3):239-41 Capdevila J, Estrada MP, Sánchez-Herrero E, Guerrero I. The Drosophila segment polarity gene patched interacts with decapentaplegic in wing development. EMBO J. 1994 Jan 1;13(1):71-82 Maesawa C, Tamura G, Iwaya T, Ogasawara S, Ishida K, Sato N, Nishizuka S, Suzuki Y, Ikeda K, Aoki K, Saito K, Satodate R. Mutations in the human homologue of the Drosophila patched gene in esophageal squamous cell carcinoma. Genes Chromosomes Cancer. 1998 Mar;21(3):276-9 Gailani MR, Ståhle-Bäckdahl M, Leffell DJ, Glynn M, Zaphiropoulos PG, Pressman C, Undén AB, Dean M, Brash DE, Bale AE, Toftgård R. The role of the human homologue of Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 17 PTCH1 patched homolog 1 (Drosophila) Lindström E, Toftgård R McGarvey TW, Maruta Y, Tomaszewski JE, Linnenbach AJ, Malkowicz SB. PTCH gene mutations in invasive transitional cell carcinoma of the bladder. Oncogene. 1998 Sep 3;17(9):1167-72 Daya-Grosjean L, Sarasin A. UV-specific mutations of the human patched gene in basal cell carcinomas from normal individuals and xeroderma pigmentosum patients. Mutat Res. 2000 May 30;450(1-2):193-9 Shafei-Benaissa E, Savage JR, Babin P, Larrègue M, Papworth D, Tanzer J, Bonnetblanc JM, Huret JL. The naevoid basal-cell carcinoma syndrome (Gorlin syndrome) is a chromosomal instability syndrome. Mutat Res. 1998 Feb 2;397(2):287-92 D'Errico M, Calcagnile A, Canzona F, Didona B, Posteraro P, Cavalieri R, Corona R, Vorechovsky I, Nardo T, Stefanini M, Dogliotti E. UV mutation signature in tumor suppressor genes involved in skin carcinogenesis in xeroderma pigmentosum patients. Oncogene. 2000 Jan 20;19(3):463-7 Bodak N, Queille S, Avril MF, Bouadjar B, Drougard C, Sarasin A, Daya-Grosjean L. High levels of patched gene mutations in basal-cell carcinomas from patients with xeroderma pigmentosum. Proc Natl Acad Sci U S A. 1999 Apr 27;96(9):5117-22 Dong J, Gailani MR, Pomeroy SL, Reardon D, Bale AE. Identification of PATCHED mutations in medulloblastomas by direct sequencing. Hum Mutat. 2000 Jul;16(1):89-90 Toftgård R. Hedgehog signalling in cancer. Cell Mol Life Sci. 2000 Nov;57(12):1720-31 Booth DR. The hedgehog signalling pathway and its role in basal cell carcinoma. Cancer Metastasis Rev. 1999;18(2):26184 This article should be referenced as such: Lindström E, Toftgård R. PTCH1 patched homolog 1 (Drosophila). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):16-18. Bailey EC, Scott MP, Johnson RL. Hedgehog signaling in animal development and human disease. Ernst Schering Res Found Workshop. 2000;(29):211-35 Barreto DC, Gomez RS, Bale AE, Boson WL, De Marco L. PTCH gene mutations in odontogenic keratocysts. J Dent Res. 2000 Jun;79(6):1418-22 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 18 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review TFF2 (TreFoil Factor 2) Catherine Tomasetto I.G.B.M.C., BP 163, 1 rue Laurent Fries, 67404 Illkirch, France (CT) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Genes/TFF2ID264.html DOI: 10.4267/2042/37695 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Homology Other names: SP (Spasmolytic Polypeptide) HGNC (Hugo): TFF2 Location: 21q22.3 Local order: Belongs to the TFF cluster TFF2 belongs to the Trefoil peptide Family (TFF) and possesses two tandemly duplicated TFF motifs each being homologous to the TFF motif of TFF1 and TFF3. The TFF motif spans about 40 amino acids and is formed by 6 conserved residues involved in specific disulfides bridges. DNA/RNA Implicated in Description 600 bp. TFF2 was found implicated in inflamatory bowel diseases, cancers of gastrointestinal organs such as stomach and pancreas; in constrast to TFF1 and TFF3, TFF2 expression was not found in breast carcinomas Protein References 5.1 kb gene, 4 exons. Transcription Description Thim L, Thomsen J, Christensen M, Jørgensen KH. The amino acid sequence of pancreatic spasmolytic polypeptide. Biochim Biophys Acta. 1985 Mar 1;827(3):410-8 Precursor: 129 amino acids; mature peptide: 106 amino acids; 3-dimentional structure was solved; the129 amino acids TFF2 protein contains a signal peptide; the mature secreted peptide of 106 amino acids contains two TFF (TreFoil Factor) domains and one acidic Cterminal domain. Rio MC, Bellocq JP, Daniel JY, Tomasetto C, Lathe R, Chenard MP, Batzenschlager A, Chambon P. Breast cancerassociated pS2 protein: synthesis and secretion by normal stomach mucosa. Science. 1988 Aug 5;241(4866):705-8 Thim L. A new family of growth factor-like peptides. 'Trefoil' disulphide loop structures as a common feature in breast cancer associated peptide (pS2), pancreatic spasmolytic polypeptide (PSP), and frog skin peptides (spasmolysins). FEBS Lett. 1989 Jun 19;250(1):85-90 Expression Under normal condition, TFF2 is expressed in mucus neck cells of the fundus, basal cells of the antral and pyloric glands and by the Brunner's glands of the duodenum. Tomasetto C, Rio MC, Gautier C, Wolf C, Hareuveni M, Chambon P, Lathe R. hSP, the domain-duplicated homolog of pS2 protein, is co-expressed with pS2 in stomach but not in breast carcinoma. EMBO J. 1990 Feb;9(2):407-14 Localisation Secreted in gastric fluid. Rio MC, Chenard MP, Wolf C, Marcellin L, Tomasetto C, Lathe R, Bellocq JP, Chambon P. Induction of pS2 and hSP genes as markers of mucosal ulceration of the digestive tract. Gastroenterology. 1991 Feb;100(2):375-9 Function In repair and epithelial restitution of the gastrointestinal mucosa. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 19 TFF2 (TreFoil Factor 2) Tomasetto C Theisinger B, Welter C, Seitz G, Rio MC, Lathe R, Chambon P, Blin N. Expression of the breast cancer associated gene pS2 and the pancreatic spasmolytic polypeptide gene (hSP) in diffuse type of stomach carcinoma. Eur J Cancer. 1991;27(6):770-3 inflammatory bowel Jan;104(1):12-20 Gastroenterology. 1993 Playford RJ, Marchbank T, Chinery R, Evison R, Pignatelli M, Boulton RA, Thim L, Hanby AM. Human spasmolytic polypeptide is a cytoprotective agent that stimulates cell migration. Gastroenterology. 1995 Jan;108(1):108-16 Welter C, Theisinger B, Seitz G, Tomasetto C, Rio MC, Chambon P, Blin N. Association of the human spasmolytic polypeptide and an estrogen-induced breast cancer protein (pS2) with human pancreatic carcinoma. Lab Invest. 1992 Feb;66(2):187-92 Babyatsky MW, deBeaumont M, Thim L, Podolsky DK. Oral trefoil peptides protect against ethanol- and indomethacininduced gastric injury in rats. Gastroenterology. 1996 Feb;110(2):489-97 Gajhede M, Petersen TN, Henriksen A, Petersen JF, Dauter Z, Wilson KS, Thim L. Pancreatic spasmolytic polypeptide: first three-dimensional structure of a member of the mammalian trefoil family of peptides. Structure. 1993 Dec 15;1(4):253-62 Seib T, Blin N, Hilgert K, Seifert M, Theisinger B, Engel M, Dooley S, Zang KD, Welter C. The three human trefoil genes TFF1, TFF2, and TFF3 are located within a region of 55 kb on chromosome 21q22.3. Genomics. 1997 Feb 15;40(1):200-2 Lefebvre O, Wolf C, Kédinger M, Chenard MP, Tomasetto C, Chambon P, Rio MC. The mouse one P-domain (pS2) and two P-domain (mSP) genes exhibit distinct patterns of expression. J Cell Biol. 1993 Jul;122(1):191-8 This article should be referenced as such: Tomasetto C. TFF2 (TreFoil Factor 2). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):19-20. Wright NA, Poulsom R, Stamp G, Van Noorden S, Sarraf C, Elia G, Ahnen D, Jeffery R, Longcroft J, Pike C. Trefoil peptide gene expression in gastrointestinal epithelial cells in Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) disease. 20 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Short Communication FGFR1OP (FGFR1 oncogene partner) Marie-Josèphe Pébusque INSERM U119, IFR 57, 27 Blvd Lei Roure, 13009 Marseille, France (MJP) Published in Atlas Database: January 2001 Online updated version: http://AtlasGeneticsOncology.org/Genes/FOPID140.html DOI: 10.4267/2042/37696 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Transcription Identity A single open reading frame of 1 197 bp mRNA; putative ATG: bp 85; stop codon at bp 1 282; alternative splicing: multiple FOP transcript variants resulting from exon 7 or exon 11 splices. Other names: FOP (Fibroblast Growth Factor Receptor 1 Oncogene Partner) HGNC (Hugo): FGFR1OP Location: 6q27 Protein Description 399 amino acids; predicted molecular mass: 44.3 kDa; Hydrophobic protein containing in its N- and C-termini several regions folding in a-helices with leucine-rich repeats with the consensus sequence L-X2-L-X3-5-LX3-5-L, in one-third of which the leucine is substituted by either a valine or an isoleucine. Expression Ubiquitous expression. Localisation Cell cytoplasm. FOP (6q27) - Courtesy Mariano Rocchi, Resources for Molecular Cytogenetics. Function DNA/RNA Unknown. Description Full length cDNA: 1 627 bp. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 21 FGFR1OP (FGFR1 oncogene partner) Pébusque MJ Implicated in Abnormal protein N-term leucine-rich region from FOP fused to the catalytic domain of FGFR1 (FGFR1 intracellular region minus the major part of the juxtamembrane domain). Oncogenesis Constitutive kinase activity of FGFR1 through constitutive activation of FGFR1 signal transduction pathways via putative constitutive dimerization capability mediated by the FOP N-term LRR sequences t(6;8)(q27; p12) myeloproliferative disorder --> FOP - FGFR1; stem-cell myeloproliferative disorder associated with the 8p12 chromosomal translocations with fusions to the catalytic domain of FGFR1. Disease Stem-cell myeloproliferative disorder characterized by myeloid hyperplasia, T -cell lymphoblastic leukemia/lymphoma and peripheral blood eosinophilia, and it generally progresses to acute myeloid leukemia; specific to the 8p12 chromosomal region. Prognosis Very poor (median survival: 12 mths). Cytogenetics Additional abnormalities: 2q+ and +21. Hybrid/Mutated gene 5' FOP - 3' FGFR1; localisation: der(6). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) References Popovici C, Zhang B, Grégoire MJ, Jonveaux P, LafagePochitaloff M, Birnbaum D, Pébusque MJ. The t(6;8)(q27;p11) translocation in a stem cell myeloproliferative disorder fuses a novel gene, FOP, to fibroblast growth factor receptor 1. Blood. 1999 Feb 15;93(4):1381-9 This article should be referenced as such: Pébusque MJ. FGFR1OP (FGFR1 oncogene partner). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):21-22. 22 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review MST1R (Macrophage stimulating 1 receptor) Debora Angeloni, Michael I Lerman Laboratory of Immunobiology, National Cancer Institute, Frederick Cancer Research Facility Bldg. 560 Rm. 12 26 Frederick, MD 21702, USA (DA, MIL) Published in Atlas Database: January 2001 Online updated version: http://AtlasGeneticsOncology.org/Genes/RONID287.html DOI: 10.4267/2042/37697 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology in intracellular transcription factors where they are involved in DNA binding. Part of exon 12 codes for the transmembrane domain, (pink). Exons 14 to 20 codes for the kinase domain (blue).Four-digit numbers refer to splice sites location, based on RON cDNA sequence. Identity Other names: C-MET-related tyrosine kinase (RON); RON protein tyrosine kinase (RON);; Macrophage stimulating protein receptor (MSP-receptor) HGNC (Hugo): MST1R Location: 3p21.31 Local order: Between LIMD1 and CCXCR1; between D3S1568 and D3S3822 Transcription Two major transcripts are detected, respectively 4.5 kb and 2 kb. ORF: 4204 bp. Protein DNA/RNA Description Description The RON protein is a glycosilated heterodimeric protein composed of one a- (35 kD) and one b-chain (150 kD) linked by an unknown number of disulfide bonds. The two chains derive from a single-chain precursor of about 185 kD that undergoes proteolytic cleavage at the basic amino acid site KRRRR. The achain is extracellular. The b-chain has an extracellular part, a one-pass transmembrane helix and an intracellular part containing the tyrosine kinase domain. The first 24 amino acids made the putative signal peptide (green). Twenty coding exons. All exons are small in size, ranging from 93 bp to 253 bp, with the exception of exon 1 (>1 kb). Exon 1, 2 and 3 code for the SEMA domain of the RON protein (red). Exons 4 codes for a PSI domain (orange), a modular structure about 50 amino acid long containing eight conserved Cys residues, putatively involved in protein-protein interactions. The sequence between exon 4 and 12 codes for four repeated modular structures called IPT (yellow); these domains are found in cell surface receptors such as MET and RON as well as Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 23 MST1R (Macrophage stimulating 1 receptor) Angeloni D, Lerman MI proposed that plexins, MET RTK family and VESPR (virus-encoded semaphorin receptor) are classified as semaphorins. RON orthologs have been identified in mouse (STK), chicken (c-sea) and Xenopus. The SEMA domain (consisting of most of a- and part of b- chain) contains the ligand (MSP) binding pocket (unpublished data). Tyrosine residues 1238 and 1239 (upward arrowheads in the figure) are essential for upregulation of RON catalytic activity. Tyrosine residues1353 and 1360 (downward arrowheads, in the figure) make a docking site that mediates high affinity interactions with multiple SH2-containing signal transducers. Mutations Germinal Several Single Nucleotide Polymorphisms (SNPs) were found in healthy CEPH individuals: A993G:Gln322Arg (index of heterozygosity: 0.28); C4024T (same-sense variant, index of heterozygosity: 0.03); A4031G: Arg1344Gly (index of heterozygosity: 0.46). Expression RON is expressed in human keratinocytes (it was initially cloned from a keratinocytes cDNA library). By Northern blot was found expressed in the following normal human tissues: skin, lung, bone marrow, small intestin, heart, pancreas, thyroid, prostate, testis (unpublished data), colonic mucosa and in a variety of cell types: granulocytes and monocytes, hematopoietic cells such as erythroid and myeloid progenitor cells, macrophages, osteoclasts, bone marrow megakaryocytes, epithelial and neuroendocrine cells. Somatic T915C: Leu296Pro was found in the tumor DNA of one single patient affected with adenocarcinoma of the lung. The mutated protein is not constitutively activated. The mutation has no causative role in the disease. Experimental introduction in the RON kinase domain of amino acid substitutions D1232V and M1254T - initially found in the oncogenes KIT, RET and MET, involved respectively in mastocytosis, Multiple Endocrine Neoplasia type 2B and renal papillary carcinoma - results in activation of oncogenic capacity and triggers a strong metastatic activity of RON. Expression of these RON mutants causes cellular accumulation of b-catenin via inhibition of its association with the axin/GSK complex and subsequent protection from proteasomal degradation (Danilkovitch-Miagkova, personal communication). Localisation Transmembrane protein. Function The ligand for RON is MSP. Originally, MSP was described as a serum factor enhancing the chemotactic response of murine peritoneal macrophage to the C5a fraction of complement, but RON/MSP complex has a much broader spectrum of activity. Ligand-stimulated RON activates the pathways regulating cell adhesion and motility, growth and survival. STK (the mouse ortholog) is essential for peri-implantation development during gestation, as STK-deficient mice (STK-/-) are viable only through the blastocyst stage. Hemizygous mice (STK+/-) grow to adulthood; however, they are highly susceptible to endotoxic shock and appear to be compromised in their ability to down-regulate nitric oxide production. These results suggest STK has a limiting role not only in the inflammatory response but also in early mouse development. Implicated in RON was found over-expressed in infiltrating breast carcinomas. A constitutively activated splicing variant of RON (lacking exon 11) was found in the gastric carcinoma cell line KATO-III. This variant induces activation of cell dissociation, motility and invasion of extracellular matrices. The same variant was found in malignant colonic mucosa. Another splicing variant, lacking exons 5 and 6, was found in the human colon carcinoma cell line HT-29.Truncated STK - the mouse RON ortholog - confers susceptibility to Friend virusinduced erythroleukemia in mice, and c-sea, the avian ortholog, causes erythroblastosis in chickens. Homology RON belongs to the MET receptor tyrosine kinase (RTK) family. On the basis of the presence of multiple PSI domains and a SEMA domain, it has been Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 24 MST1R (Macrophage stimulating 1 receptor) Angeloni D, Lerman MI References of liver progenitor cells in vitro. Mol Biol Cell. 1996 Apr;7(4):495-504 Leonard EJ, Skeel A. A serum protein that stimulates macrophage movement, chemotaxis and spreading. Exp Cell Res. 1976 Oct 15;102(2):434-8 Nakamura T, Aoki S, Takahashi T, Matsumoto K, Kiyohara T, Nakamura T. Cloning and expression of Xenopus HGF-like protein (HLP) and Ron/HLP receptor implicate their involvement in early neural development. Biochem Biophys Res Commun. 1996 Jul 16;224(2):564-73 Smith DR, Vogt PK, Hayman MJ. The v-sea oncogene of avian erythroblastosis retrovirus S13: another member of the proteintyrosine kinase gene family. Proc Natl Acad Sci U S A. 1989 Jul;86(14):5291-5 Tsujimura T. Role of c-kit receptor tyrosine kinase in the development, survival and neoplastic transformation of mast cells. Pathol Int. 1996 Dec;46(12):933-8 Dausset J, Cann H, Cohen D, Lathrop M, Lalouel JM, White R. Centre d'etude du polymorphisme humain (CEPH): collaborative genetic mapping of the human genome. Genomics. 1990 Mar;6(3):575-7 Wang MH, Dlugosz AA, Sun Y, Suda T, Skeel A, Leonard EJ. Macrophage-stimulating protein induces proliferation and migration of murine keratinocytes. Exp Cell Res. 1996 Jul 10;226(1):39-46 Ronsin C, Muscatelli F, Mattei MG, Breathnach R. A novel putative receptor protein tyrosine kinase of the met family. Oncogene. 1993 May;8(5):1195-202 Wang MH, Montero-Julian FA, Dauny I, Leonard EJ. Requirement of phosphatidylinositol-3 kinase for epithelial cell migration activated by human macrophage stimulating protein. Oncogene. 1996 Nov 21;13(10):2167-75 Gaudino G, Follenzi A, Naldini L, Collesi C, Santoro M, Gallo KA, Godowski PJ, Comoglio PM. RON is a heterodimeric tyrosine kinase receptor activated by the HGF homologue MSP. EMBO J. 1994 Aug 1;13(15):3524-32 Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, Scherer SW, Zhuang Z, Lubensky I, Dean M, Allikmets R, Chidambaram A, Bergerheim UR, Feltis JT, Casadevall C, Zamarron A, Bernues M, Richard S, Lips CJ, Walther MM, Tsui LC, Geil L, Orcutt ML, Stackhouse T, Lipan J, Slife L, Brauch H, Decker J, Niehans G, Hughson MD, Moch H, Storkel S, Lerman MI, Linehan WM, Zbar B. Germline and somatic mutations in the tyrosine kinase domain of the MET protooncogene in papillary renal carcinomas. Nat Genet. 1997 May;16(1):68-73 Hofstra RM, Landsvater RM, Ceccherini I, Stulp RP, Stelwagen T, Luo Y, Pasini B, Höppener JW, van Amstel HK, Romeo G. A mutation in the RET proto-oncogene associated with multiple endocrine neoplasia type 2B and sporadic medullary thyroid carcinoma. Nature. 1994 Jan 27;367(6461):375-6 Iwama A, Okano K, Sudo T, Matsuda Y, Suda T. Molecular cloning of a novel receptor tyrosine kinase gene, STK, derived from enriched hematopoietic stem cells. Blood. 1994 Jun 1;83(11):3160-9 Maggiora P, Marchio S, Stella MC, Giai M, Belfiore A, De Bortoli M, Di Renzo MF, Costantino A, Sismondi P, Comoglio PM. Overexpression of the RON gene in human breast carcinoma. Oncogene. 1998 Jun 4;16(22):2927-33 Skeel A, Leonard EJ. Action and target cell specificity of human macrophage-stimulating protein (MSP). J Immunol. 1994 May 1;152(9):4618-23 Santoro MM, Penengo L, Minetto M, Orecchia S, Cilli M, Gaudino G. Point mutations in the tyrosine kinase domain release the oncogenic and metastatic potential of the Ron receptor. Oncogene. 1998 Aug 13;17(6):741-9 Wang MH, Ronsin C, Gesnel MC, Coupey L, Skeel A, Leonard EJ, Breathnach R. Identification of the ron gene product as the receptor for the human macrophage stimulating protein. Science. 1994 Oct 7;266(5182):117-9 Waltz SE, Toms CL, McDowell SA, Clay LA, Muraoka RS, Air EL, Sun WY, Thomas MB, Degen SJ. Characterization of the mouse Ron/Stk receptor tyrosine kinase gene. Oncogene. 1998 Jan 8;16(1):27-42 Gaudino G, Avantaggiato V, Follenzi A, Acampora D, Simeone A, Comoglio PM. The proto-oncogene RON is involved in development of epithelial, bone and neuro-endocrine tissues. Oncogene. 1995 Dec 21;11(12):2627-37 Willett CG, Wang MH, Emanuel RL, Graham SA, Smith DI, Shridhar V, Sugarbaker DJ, Sunday ME. Macrophagestimulating protein and its receptor in non-small-cell lung tumors: induction of receptor tyrosine phosphorylation and cell migration. Am J Respir Cell Mol Biol. 1998 Apr;18(4):489-96 Iwama A, Wang MH, Yamaguchi N, Ohno N, Okano K, Sudo T, Takeya M, Gervais F, Morissette C, Leonard EJ, Suda T. Terminal differentiation of murine resident peritoneal macrophages is characterized by expression of the STK protein tyrosine kinase, a receptor for macrophage-stimulating protein. Blood. 1995 Nov 1;86(9):3394-403 Bork P, Doerks T, Springer TA, Snel B. Domains in plexins: links to integrins and transcription factors. Trends Biochem Sci. 1999 Jul;24(7):261-3 Banu N, Price DJ, London R, Deng B, Mark M, Godowski PJ, Avraham H. Modulation of megakaryocytopoiesis by human macrophage-stimulating protein, the ligand for the RON receptor. J Immunol. 1996 Apr 15;156(8):2933-40 Danilkovitch A, Skeel A, Leonard EJ. Macrophage stimulating protein-induced epithelial cell adhesion is mediated by a PI3-Kdependent, but FAK-independent mechanism. Exp Cell Res. 1999 May 1;248(2):575-82 Collesi C, Santoro MM, Gaudino G, Comoglio PM. A splicing variant of the RON transcript induces constitutive tyrosine kinase activity and an invasive phenotype. Mol Cell Biol. 1996 Oct;16(10):5518-26 Muraoka RS, Sun WY, Colbert MC, Waltz SE, Witte DP, Degen JL, Friezner Degen SJ. The Ron/STK receptor tyrosine kinase is essential for peri-implantation development in the mouse. J Clin Invest. 1999 May;103(9):1277-85 Kurihara N, Iwama A, Tatsumi J, Ikeda K, Suda T. Macrophage-stimulating protein activates STK receptor tyrosine kinase on osteoclasts and facilitates bone resorption by osteoclast-like cells. Blood. 1996 May 1;87(9):3704-10 Okino T, Egami H, Ohmachi H, Takai E, Tamori Y, Nakagawa K, Nakano S, Akagi J, Sakamoto O, Suda T, Ogawa M. Presence of RON receptor tyrosine kinase and its splicing variant in malignant and non-malignant human colonic mucosa. Int J Oncol. 1999 Oct;15(4):709-14 Medico E, Mongiovi AM, Huff J, Jelinek MA, Follenzi A, Gaudino G, Parsons JT, Comoglio PM. The tyrosine kinase receptors Ron and Sea control "scattering" and morphogenesis Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 25 MST1R (Macrophage stimulating 1 receptor) Angeloni D, Lerman MI Persons DA, Paulson RF, Loyd MR, Herley MT, Bodner SM, Bernstein A, Correll PH, Ney PA. Fv2 encodes a truncated form of the Stk receptor tyrosine kinase. Nat Genet. 1999 Oct;23(2):159-65 Chen YQ, Zhou YQ, Angeloni D, Kurtz AL, Qiang XZ, Wang MH. Overexpression and activation of the RON receptor tyrosine kinase in a panel of human colorectal carcinoma cell lines. Exp Cell Res. 2000 Nov 25;261(1):229-38 Wahl RC, Hsu RY, Huff JL, Jelinek P, Patterson SD, Parsons JT, macrophage stimulating protein is protein-tyrosine kinase Sea. J 10;274(37):26361-8 Danilkovitch A, Donley S, Skeel A, Leonard EJ. Two independent signaling pathways mediate the antiapoptotic action of macrophage-stimulating protein on epithelial cells. Mol Cell Biol. 2000 Mar;20(6):2218-27 MA, Chen K, Courchesne Welcher AA. Chicken a ligand of the receptor Biol Chem. 1999 Sep This article should be referenced as such: Angeloni D, Danilkovitch-Miagkova A, Ivanov SV, Breathnach R, Johnson BE, Leonard EJ, Lerman MI. Gene structure of the human receptor tyrosine kinase RON and mutation analysis in lung cancer samples. Genes Chromosomes Cancer. 2000 Oct;29(2):147-56 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Angeloni D, Lerman MI. MST1R (Macrophage stimulating 1 receptor). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):23-26. 26 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Mini Review FIM (fused in myeloproliferative disorders) Marie-Josèphe Pébusque INSERM U119, IFR 57, 27 Blvd Lei Roure, 13009 Marseille, France (MJP) Published in Atlas Database: January 2001 Online updated version : http://AtlasGeneticsOncology.org/Genes/ZNF198ID114.html DOI: 10.4267/2042/37698 This article is an update of : Huret JL, Leroux D. ZNF198 (zinc finger protein 198). Atlas Genet Cytogenet Oncol Haematol 1998;2(2):52-53 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity DNA/RNA Other names: FIM (fused in myeloproliferative disorders).; ZNF198 (zinc finger protein 198).; RAMP HGNC (Hugo): ZMYM2 Location: 13q12 Local order: Proximal from FLT1 and FLT3. Description Full length cDNA: 5,016 bp; a single open reading frame of 4,137 bp; alternative spliced cDNA variant. Transcription Main transcripts: 5.0 and 7.5 kb. Protein Description 1 379 amino acids; hydrophobic protein containing several motifs: a N-terminal cystein-rich region containing ten repeats with the consensus sequence CX2-C-X18-24-F/Y-C-X3-C, which correspond to a novel zinc finger motifs, a highly hydrophobic prolinerich stretch, and a bipartite nuclear localization signal. Expression Wide. FIM (13q12) - Courtesy Mariano Rocchi, Resources for Molecular Cytogenetics. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 27 FIM (fused in myeloproliferative disorders) Pébusque MJ DNA Diagram. Localisation Abnormal protein Aberrant tyrosine kinase composed of the N-term twothirds of FIM (retaining the 10 putative zinc finger motifs), and the FGFR1 intracellular region minus the major part of the juxtamembrane domain. Oncogenesis Constitutive kinase activity of FGFR1 through constitutive activation of FGFR1 signal transduction pathways via constitutive dimerization capability mediated by the FIM N-term zinc finger sequences. Cell nucleus and nucleolus; within the nucleolus, colocalizes with UBF (Upstream Binding Factor). Function May be involved in the regulation of rRNA transcription. Homology FIM is related to DXS6673E, a gene which may be related with mental retardation. References Implicated in Popovici C, Adélaïde J, Ollendorff V, Chaffanet M, Guasch G, Jacrot M, Leroux D, Birnbaum D, Pébusque MJ. Fibroblast growth factor receptor 1 is fused to FIM in stem-cell myeloproliferative disorder with t(8;13). Proc Natl Acad Sci U S A. 1998 May 12;95(10):5712-7 t(8;13)(p12;q12)/ANLL-NHL --> 5' FIM - 3' FGFR1; stem-cell myeloproliferative disorder associated with the 8p12 chromosomal translocations; fused to the catalytic domain of FGFR1 Disease stem-cell myeloproliferative disorder characterized by myeloid hyperplasia, T -cell lymphoblastic leukemia/lymphoma and peripheral blood eosinophilia, and it generally progresses to acute myeloid leukemia; specific to the 8p12 chromosomal region. Prognosis Very poor (median survival: 12 mths). Cytogenetics Usually, t(8;13)(p12;q12) occurs as a single anomaly; duplication of the der(13) was found during disease progression, suggesting that the crucial event might lie on this derivative chromosome; additional abnormalities:+8, +21. Hybrid/Mutated gene 5' FIM - 3' FGFR1; localisation: der(13) Smedley D, Hamoudi R, Clark J, Warren W, Abdul-Rauf M, Somers G, Venter D, Fagan K, Cooper C, Shipley J. The t(8;13)(p11;q11-12) rearrangement associated with an atypical myeloproliferative disorder fuses the fibroblast growth factor receptor 1 gene to a novel gene RAMP. Hum Mol Genet. 1998 Apr;7(4):637-42 Xiao S, Nalabolu SR, Aster JC, Ma J, Abruzzo L, Jaffe ES, Stone R, Weissman SM, Hudson TJ, Fletcher JA. FGFR1 is fused with a novel zinc-finger gene, ZNF198, in the t(8;13) leukaemia/lymphoma syndrome. Nat Genet. 1998 Jan;18(1):84-7 Ollendorff V, Guasch G, Isnardon D, Galindo R, Birnbaum D, Pébusque MJ. Characterization of FIM-FGFR1, the fusion product of the myeloproliferative disorder-associated t(8;13) translocation. J Biol Chem. 1999 Sep 17;274(38):26922-30 Xiao S, McCarthy JG, Aster JC, Fletcher JA. ZNF198-FGFR1 transforming activity depends on a novel proline-rich ZNF198 oligomerization domain. Blood. 2000 Jul 15;96(2):699-704 This article should be referenced as such: Pébusque MJ. FIM (fused in myeloproliferative disorders). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):27-28. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 28 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication t(1;3)(p36;q21) Pascale Cornillet-Lefebvre, Sylvie Daliphard, Stéphanie Struski Laboratory of Hematology, Robert Debré(PCL), Hospital and Medical Faculty (UPRES EA 20-70-IFR 53 Biomolecules), 51092, Reims Cedex, France (SD, SS) Published in Atlas Database: November 2000 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/t0103.html DOI: 10.4267/2042/37699 This article is an update of: Huret JL. t(1;3)(p36;q21). Atlas Genet Cytogenet Oncol Haematol.1997;1(1):16. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity t(1;3)(p36;q21) G-banding (left) - Courtesy Diane H. Norback, Eric B. Johnson, and Sara Morrison-Delap, Cytogenetics at the Waisman Center; R-banding (right) -Courtesy Pascale Cornillet-Lefebvre and Stéphanie Struski. Clinics and pathology chronic myelogenous leukemia (CML), 1 multiple myeloma, 1 waldenstrom's macroglobulinemia. Disease Epidemiology Myeloid lineage (MDS, ANLL, therapy related ANLL, CML, MPD); very rarely in lymphoid lineage. Patients are aged: 30-80 years. Clinics Phenotype/cell stem origin Blood data: frequent thrombocytosis or normal platelet count. Of 39 available cases, there were: 22 myelodysplastic syndromes (MDS) (17/22 transformed into refractory acute non lymphoblastic leukemia (ANLL) of -M1 or M4 type), 8 de novo ANLL, 3 therapy-related MDS, 2 polycythemia vera, 1 essential thrombocythemia, 1 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Cytology Frequently characterized by dysmegakaryocytopoiesis. 29 t(1;3)(p36;q21) Cornillet-Lefebvre P et al. molecular mechanism may explain the clinical and morphologic similarities seen in these malignancies. Pathology Trilineage dysplasia. References Prognosis Very poor so far: from 16 cases, median survival was 6 months in ANLL, 20 months in MDS. Welborn JL, Lewis JP, Jenks H, Walling P. Diagnostic and prognostic significance of t(1;3)(p36;q21) in the disorders of hematopoiesis. Cancer Genet Cytogenet. 1987 Oct;28(2):27785 Cytogenetics Grigg AP, Gascoyne RD, Phillips GL, Horsman DE. Clinical, haematological and cytogenetic features in 24 patients with structural rearrangements of the Q arm of chromosome 3. Br J Haematol. 1993 Jan;83(1):158-65 Cytogenetics morphological del(5q) in 5 of 20 cases (1/4). Genes involved and proteins Secker-Walker LM, Mehta A, Bain B. Abnormalities of 3q21 and 3q26 in myeloid malignancy: a United Kingdom Cancer Cytogenetic Group study. Br J Haematol. 1995 Oct;91(2):490501 Note Genes involved are yet unknown. Shimizu S, Suzukawa K, Kodera T, Nagasawa T, Abe T, Taniwaki M, Yagasaki F, Tanaka H, Fujisawa S, Johansson B, Ahlgren T, Yokota J, Morishita K. Identification of breakpoint cluster regions at 1p36.3 and 3q21 in hematologic malignancies with t(1;3)(p36;q21). Genes Chromosomes Cancer. 2000 Mar;27(3):229-38 To be noted Note It has been hypothesized that the chromosomal breakpoints at the 3q21 in ANLL/MDS with t(1;3) overlap with the breakpoints of the 3q21q26 syndrome(inv(3)(q21q26), t(3;3)(q21;q26)); common Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Cornillet-Lefebvre P, Daliphard S, Struski S. t(1;3)(p36;q21). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):29-30. 30 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication del(17p) in non-Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi Hematology Section, Department of Biomedical Sciences, University of Ferrara, Corso Giovecca 203, Ferrara, Italy (AC, GLC) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/del17pNHLID2083.html DOI: 10.4267/2042/37700 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Prognosis Note the 17p- chromosome is a secondary change in most cases of NHL The 17p- chromosome was reported to predict for a poor prognosis in low grade lymphomas; any abnormality of chromosome 17 was also reported to negatively affect survival in lymphomas of all histologic grades Cytogenetics Cytogenetics morphological The deleted segment may vary in size and many cases with sub-microscopic deletions involving the 17p13 band were reported by FISH; cases with unbalanced 17p translocations leading to 17p loss were also described; these cases may be associated with dicentric rearrangements. The 17p- is usually associated with transformation of a low-grade FCCL with t(14;18) into a high grade lymphoma; likewise, there is a higher incidence of 17pin the blastoid variant of MCL with t(11,14) than in the typical form. del(17p) in non-Hodgkin's lymphoma (NHL) G- banding Courtesy Melanie Zenger and Claudia Haferlach. Clinics and pathology Cytogenetics molecular Disease The deletion may be detected by G or R-banding; FISH using a 17p13/p53 probe is recommended, this technique being more sensitive than conventional cytogenetics. Virtually all histologic subsets of NHL may harbour a 17p- chromosome; there is variation in the reported incidence due to heterogeneity of histologic classification and to the different sensitivity of the detection methods. 10 to 15% of follicle centre cell lymphoma (FCCL) and mantle cell lymphomas (MCL) may carry a 17pchromosome; minority of marginal zone B-cell lymphomas may be associated with 17p deletion. This anomaly is rarely found in T-cell NHL. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Genes involved and proteins Note The majority of cases with 17p- carry a p53 gene deletion, associated with mutation of the remaining allele; there may be a small fraction of cases with a more distal deletion involving an as yet unidentified locus. 31 del(17p) in non-Hodgkin's lymphoma (NHL) Cuneo A, Castoldi GL Guillermo A, Piris MA, Cardesa A, Montserrat E, Miró R, Campo E. Increased number of chromosomal imbalances and high-level DNA amplifications in mantle cell lymphoma are associated with blastoid variants. Blood. 1999 Jun 15;93(12):4365-74 References Cabanillas F, Pathak S, Grant G, Hagemeister FB, McLaughlin P, Swan F, Rodriguez MA, Trujillo J, Cork A, Butler JJ. Refractoriness to chemotherapy and poor survival related to abnormalities of chromosomes 17 and 7 in lymphoma. Am J Med. 1989 Aug;87(2):167-72 Callet-Bauchu E, Salles G, Gazzo S, Poncet C, Morel D, Pagès J, Coiffier B, Coeur P, Felman P. Translocations involving the short arm of chromosome 17 in chronic Blymphoid disorders: frequent occurrence of dicentric rearrangements and possible association with adverse outcome. Leukemia. 1999 Mar;13(3):460-8 Schlegelberger B, Himmler A, Gödde E, Grote W, Feller AC, Lennert K. Cytogenetic findings in peripheral T-cell lymphomas as a basis for distinguishing low-grade and high-grade lymphomas. Blood. 1994 Jan 15;83(2):505-11 Cuneo A, Bigoni R, Rigolin GM, Roberti MG, Bardi A, Piva N, Milani R, Bullrich F, Veronese ML, Croce C, Birg F, Döhner H, Hagemeijer A, Castoldi G. Cytogenetic profile of lymphoma of follicle mantle lineage: correlation with clinicobiologic features. Blood. 1999 Feb 15;93(4):1372-80 Tilly H, Rossi A, Stamatoullas A, Lenormand B, Bigorgne C, Kunlin A, Monconduit M, Bastard C. Prognostic value of chromosomal abnormalities in follicular lymphoma. Blood. 1994 Aug 15;84(4):1043-9 Clodi K, Younes A, Goodacre A, Roberts M, Palmer J, Younes M, Cabanillas F, Andreeff M. Analysis of p53 gene deletions in patients with non-Hodgkin's lymphoma by dual-colour fluorescence in-situ hybridization. Br J Haematol. 1997 Sep;98(4):913-21 Cuneo A, Bigoni R, Roberti MG, Milani R, Agostini P, Cavazzini F, Minotto C, De Angeli C, Bardi A, Tammiso E, Negrini M, Cavazzini P, Castoldi G. Molecular cytogenetic characterization of marginal zone B-cell lymphoma: correlation with clinicopathologic findings in 14 cases. Haematologica. 2001 Jan;86(1):64-70 Sankar M, Tanaka K, Kumaravel TS, Arif M, Shintani T, Yagi S, Kyo T, Dohy H, Kamada N. Identification of a commonly deleted region at 17p13.3 in leukemia and lymphoma associated with 17p abnormality. Leukemia. 1998 Apr;12(4):510-6 This article should be referenced as such: Cuneo A, Castoldi GL. del(17p) in non-Hodgkin's lymphoma (NHL). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):3132. Beà S, Ribas M, Hernández JM, Bosch F, Pinyol M, Hernández L, García JL, Flores T, González M, López- Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 32 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication del(20q) in myeloid malignancies Chrystèle Bilhou-Nabera Laboratoire d'Hématologie, Hôpital du Haut-Lévêque, CHU de Bordeaux, Ave de Magellan, 33 604 Pessac, France (CBN) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/del20qID1040.html DOI: 10.4267/2042/37701 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Epidemiology Identity An interstitial or terminal deletion of the long arm of chromosome 20 (20q-) has been described as the second most frequent sole clonal structural abnormality (5 %) behind t(9.22). Prognosis In MDS, 20q- alone is associated with a good prognosis regarding survival and potential for AML evolution, as defined by the International Prognostic Scoring System (IPSS) for MDS prognosis. In de novo acute leukemia, a poor response to treatment and a reduced survival is observed. In myeloproliferative disorders, the presence of 20q does not appear to adversely affect survival. Cytogenetics del(20q) G- banding (left) - Courtesy Diane H. Norback, Eric B. Johnson, Sara Morrison-Delap Cytogenetics at theWaisman Center; R-banding (right) - top: Courtesy Jean-Luc Lai; bottom: Editor Cytogenetics morphological The breakpoint on chromosome 20 is not constant; 20qis frequently associated with other cytogenetic abnormalities as del(5q), trisomy 8, trisomy 21, deletions or translocations involving the long arm of chromosome 13; a newly described translocation t(11;20)(p15;q11) resulting in a NUP98- TOP1 fusion gene was described in therapy-related myelodysplastic syndrome (RAEB); t(11;20)(p15;q11) is a rare recurrent translocation reported in patients with MDS, ANLL and polycythemia vera. Clinics and pathology Disease A very large spectrum of hematological malignancies as myelodysplastic syndromes (MDS), acute non lymphocytic leukemias (ANLL), polycythemia vera, chronic neutrophilic leukemia. Phenotype/cell stem origin As described in various types of hematological disorders, 20q- appears as a primary karyotypic abnormality occurring in a pluripotential hematopoietic stem cell; the pathogenic mechanism by which 20qalters the hematopoietic stem cells in hematological disorders remains unknown; 20q- may confer a proliferative advantage to myeloid cells through deletion of a tumor suppressor gene. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Cytogenetics molecular A small fragment (around 8 Mb), proximally flanked by D20S206 and distally by D20S119 and UT 654 was identified using FISH. Additional anomalies del(5q), trisomy 8, deletions or translocations involving 13q and trisomy 21. 33 del(20q) in myeloid malignancies Bilhou-Nabera C Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, Ohyashiki K, Toyama K, Aul C, Mufti G, Bennett J. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997 Mar 15;89(6):2079-88 Genes involved and proteins Note Genes remaining within this deleted region are topoisomerase 1 (TPO1-OMIN 126420), phospholipase C (PLC1), hepatocyte factor nuclear 4 (HNF4) and adenosine desaminase (ADA); recently, a new gene KRML transcriptional regulator was mapped in the smallest commonly deleted region in malignant myeloid leukemias. Wang PW, Iannantuoni K, Davis EM, Espinosa R 3rd, Stoffel M, Le Beau MM. Refinement of the commonly deleted segment in myeloid leukemias with a del(20q). Genes Chromosomes Cancer. 1998 Feb;21(2):75-81 Ahuja HG, Felix CA, Aplan PD. The t(11;20)(p15;q11) chromosomal translocation associated with therapy-related myelodysplastic syndrome results in an NUP98-TOP1 fusion. Blood. 1999 Nov 1;94(9):3258-61 References Wang PW, Eisenbart JD, Cordes SP, Barsh GS, Stoffel M, Le Beau MM. Human KRML (MAFB): cDNA cloning, genomic structure, and evaluation as a candidate tumor suppressor gene in myeloid leukemias. Genomics. 1999 Aug 1;59(3):27581 Mitelman F, Kaneko Y, Trent J. Report of the committee on chromosome changes in neoplasia. Cytogenetic Cell Genet 1990; 55(1-4):358-86. Campbell LJ, Garson OM. The prognostic significance of deletion of the long arm of chromosome 20 in myeloid disorders. Leukemia. 1994 Jan;8(1):67-71 This article should be referenced as such: Bilhou-Nabera C. del(20q) in myeloid malignancies. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):33-34. Kurtin PJ, Dewald GW, Shields DJ, Hanson CA. Hematologic disorders associated with deletions of chromosome 20q: a clinicopathologic study of 107 patients. Am J Clin Pathol. 1996 Nov;106(5):680-8 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 34 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Mini Review Juvenile Chronic Myelogenous Leukemia (JCML) Jay L Hess Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 413b Stellar Chance Laboratories, Philadelphia, PA 19104, USA (JLH) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/JCMLID1099.html DOI: 10.4267/2042/37702 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology of left shift; myeloblasts average about 5% of total nucleated cells; elevation of fetal hemoglobin (hbF) very common; absence of the Philadelphia chromosome in all cases. Proposed clinical criteria from the International Juvenile Myelomonocytic Leukemia Working Group includes: 1. White blood cell count > 13 x 109/L (corrected for nucleated red blood cells). 2. Absolute monocyte count >1 x 109/L (corrected). 3. Presence of immature myeloid precursors (myelocytes, promyelocytes, and myeloblasts) in the peripheral blood. 4. Bone marrow aspirate revealing < 30% blasts 5. No Ph chromosome on cytogenetic assessment. About 15% of cases are associated with neurofibromatosis type 1 (NF-1 mutation). Identity Alias Juvenile myelomonocytic leukemia (JMML) Juvenile myelomonocytic leukemia syndrome (JMML syndrome) Note The proper terminology of this disorder is controversial; many authors, including the European Working Group on MDS in Childhood favor the term JMML; another working group suggests using the term JMML syndrome with a qualifier with or without monosomy 7 or 7q-. Clinics and pathology Disease Pathology JCML is a chronic myeloproliferative disorder that typically affects young children: more than 95% of cases are diagnosed before age 4. Blood: leukocytosis, monocytosis, left shift in myeloid maturation, circulating mucleated red blood cells. Bone marrow: hypercellular marrow with mildly increased M:E ratio (typically 5:1), dispersed erythroid elements, and decreased numbers of megakaryocytes; dyplasia is usually not prominent. Phenotype/cell stem origin Evidence exists for leukemic involvement of CD34positive stem cells and monocyte-macrophage, erythroid, and B-lymphoid lineages in cases with cytogenetic abnormalities. Treatment Annual incidence is estimated to be roughly 4/million; median age 1-4 yrs; sex ratio: 1.4M/1F. Intensive chemotherapy and all trans retinoic have not been shown to induce durable remissions; complete remissions have been achieved with stem cell transplantation. Clinics Prognosis Splenomegaly, lymphadenopathy, and skin rash are common; typical peripheral blood findings include leukocytosis (usually less than 100 x 109/L), monocytosis, and thrombocytosis with variable degree The disease is uniformly fatal when treated with conventional chemotherapy; among those who undergo bone marrow transplantations, the majority ultimately relapse, with an overall survival rate of 25%. Epidemiology Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 35 Juvenile Chronic Myelogenous Leukemia (JCML) Hess JL Lutz P, Zix-Kieffer I, Souillet G, Bertrand Y, Dhooge C, Rubie C, Mazingue F, Marguerite F, Machinaud-Lacroix F, Rialland X, Plouvier E, Behar C, Vilmer E, Philippe N, Otten J. Juvenile myelomonocytic leukemia: analyses of treatment results in the EORTC Children's Leukemia Cooperative Group (CLCG). Bone Marrow Transplant. 1996 Dec;18(6):1111-6 Cytogenetics Cytogenetics morphological Other than the frequent association with monosomy 7, no consistent cytogenetic abnormalities have been identified; whether the infantile monosomy 7 syndrome is distinct from JCML is controversial. Kai S, Sumita H, Fujioka K, Takahashi H, Hanzawa N, Funabiki T, Ikuta K, Sasaki H. Loss of heterozygosity of NF1 gene in juvenile chronic myelogenous leukemia with neurofibromatosis type 1. Int J Hematol. 1998 Jul;68(1):53-60 Genes involved and proteins Zhang YY, Vik TA, Ryder JW, Srour EF, Jacks T, Shannon K, Clapp DW. Nf1 regulates hematopoietic progenitor cell growth and ras signaling in response to multiple cytokines. J Exp Med. 1998 Jun 1;187(11):1893-902 Note Mechanisms of Oncogenesis: JCML patients show spontaneous growth of granulocyte-macrophage colony forming units (CFUGM) from peripheral blood, which appears to be the result of hypersensitivity to GM-CSF, IL-3, or SCF; cases associated with NF-1 are likely to be the result of constitutive activation of the Ras pathway as a result of decreased GT Pase activity although there is also evidence of a GAP independent function; up to 30% of cases show mutations in K-ras and N-ras; the importance of the RAS pathway has been confirmed in mouse models with targeted disrupted of Nf-1; recently data suggest that TNFa produced by neoplastic cells may prevent expansion of hematopoietic progenitors. Hasle H, Aricò M, Basso G, Biondi A, Cantù Rajnoldi A, Creutzig U, Fenu S, Fonatsch C, Haas OA, Harbott J, Kardos G, Kerndrup G, Mann G, Niemeyer CM, Ptoszkova H, Ritter J, Slater R, Starý J, Stollmann-Gibbels B, Testi AM, van Wering ER, Zimmermann M. Myelodysplastic syndrome, juvenile myelomonocytic leukemia, and acute myeloid leukemia associated with complete or partial monosomy 7. European Working Group on MDS in Childhood (EWOG-MDS). Leukemia. 1999 Mar;13(3):376-85 Smith FO, Sanders JE. Juvenile myelomonocytic leukemia: what we don't know. J Pediatr Hematol Oncol. 1999 NovDec;21(6):461-3 Birnbaum RA, O'Marcaigh A, Wardak Z, Zhang YY, Dranoff G, Jacks T, Clapp DW, Shannon KM. Nf1 and Gmcsf interact in myeloid leukemogenesis. Mol Cell. 2000 Jan;5(1):189-95 References Cooper LJ, Shannon KM, Loken MR, Weaver M, Stephens K, Sievers EL. Evidence that juvenile myelomonocytic leukemia can arise from a pluripotential stem cell. Blood. 2000 Sep 15;96(6):2310-3 Bollag G, Clapp DW, Shih S, Adler F, Zhang YY, Thompson P, Lange BJ, Freedman MH, McCormick F, Jacks T, Shannon K. Loss of NF1 results in activation of the Ras signaling pathway and leads to aberrant growth in haematopoietic cells. Nat Genet. 1996 Feb;12(2):144-8 Novitzky N. Myelodysplastic syndromes in children. A critical review of the clinical manifestations and management. Am J Hematol. 2000 Apr;63(4):212-22 Hess JL, Zutter MM, Castleberry RP, Emanuel PD. Juvenile chronic myelogenous leukemia. Am J Clin Pathol. 1996 Feb;105(2):238-48 This article should be referenced as such: Hess JL. Juvenile Chronic Myelogenous Leukemia (JCML). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):35-36. Largaespada DA, Brannan CI, Jenkins NA, Copeland NG. Nf1 deficiency causes Ras-mediated granulocyte/macrophage colony stimulating factor hypersensitivity and chronic myeloid leukaemia. Nat Genet. 1996 Feb;12(2):137-43 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 36 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication t(6;8)(q27;p12) Marie-Josèphe Pébusque INSERM U119, IFR 57, 27 Blvd Lei Roure, 13009 Marseille, France (MJP) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/t68ID1090.html DOI: 10.4267/2042/37703 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Clinics and pathology Genes involved and proteins Disease FGFR1 Multilineage disorder with combined occurrence of myeloid malignancy and T- cell NHL, or myeloid metaplasia. Location: 8p12 Protein FGF receptor with tyrosine kinase activity. Phenotype/cell stem origin FOP (FGFR1 Oncogene Partner) The same t(6;8)(q27;p12) is found both in the bone marrow and in the lymph node: the multilineage involvement suggests the malignant transformation of a primitive hematopoietic stem cell. Location: 6q27 Protein Hydrophobic protein containing alpha-helices in the Nand C-termini with leucine-rich repeats. Epidemiology Result of the chromosomal anomaly 4 cases are described; median age 29 years (range 2348); sex ratio: 2M/2F. Clinics Aggressive disease; complex picture of myeloid hyperplasia progressing to myelodysplasia and Tlymphoma, and acute non lymphocytic leukemia; enlarged lymph node infiltrated by myeloid blast cells; blood data: high WBC (median 40 X 109/l); myelemia; monocytosis and eosinophilia. Hybrid gene Evolution Description Aberrant tyrosine kinase composed of the putative leucine-rich N-terminal region of FOP, and the FGFR1 intracellular region minus the major part of the juxtamembrane domain. Oncogenesis Through constitutive activation of FGFR1 signal transduction pathways, via putative dimerization of the fusion protein via the FOP leucine-rich repeats. Description Breakpoint in FGFR1 intron 8 which encodes the juxtamembrane domain, breakpoint in FOP intron 6. Fusion protein CR is obtained, but is promptly followed by relapse progressing rapidly to acute non lymphocytic leukemia. Prognosis Median survival: 6 months. Cytogenetics Cytogenetics morphological References Occurs as a single anomaly. Cytogenetics molecular Vannier JP, Bizet M, Bastard C, Bernard A, Ducastelle T, Tron P. Simultaneous occurrence of a T-cell lymphoma and a chronic myelogenous leukemia with an unusual karyotype. Leuk Res. 1984;8(4):647-57 Mega YAC 959-A -4 (1260 kb) from CEPH; FGFR1specific cosmid 134.8. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 37 t(6;8)(q27;p12) Pébusque MJ Elsner S, Martin H, Rode C, Wassman B, Ganser A, Hoelzer D. An uncommon chromosomal translocation t(6;8) associated with atypical myelogenous leukemia/myeloproliferative disease detected by fluorescence in situ hybridisation. Br J Haematol, 1994;87:124 disorders have close or identical breakpoints in chromosome region 8p11-12. Oncogene. 1998 Feb 19;16(7):945-9 Popovici C, Zhang B, Grégoire MJ, Jonveaux P, LafagePochitaloff M, Birnbaum D, Pébusque MJ. The t(6;8)(q27;p11) translocation in a stem cell myeloproliferative disorder fuses a novel gene, FOP, to fibroblast growth factor receptor 1. Blood. 1999 Feb 15;93(4):1381-9 Macdonald D, Aguiar RC, Mason PJ, Goldman JM, Cross NC. A new myeloproliferative disorder associated with chromosomal translocations involving 8p11: a review. Leukemia. 1995 Oct;9(10):1628-30 This article should be referenced as such: Chaffanet M, Popovici C, Leroux D, Jacrot M, Adélaïde J, Dastugue N, Grégoire MJ, Hagemeijer A, Lafage-Pochitaloff M, Birnbaum D, Pébusque MJ. t(6;8), t(8;9) and t(8;13) translocations associated with stem cell myeloproliferative Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Pébusque MJ. t(6;8)(q27;p12). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):37-38. 38 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication t(8;13)(p12;q12) Marie-Josèphe Pébusque, Nicholas CP Cross INSERM U119, IFR 57, 27 Blvd Leï Roure, 13009 Marseille, France (MJP); Wessex Regional Genetics Laboratory, Salisbury District Hospital, Salisbury, SP2 8BJ, UK (NCPC) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/t813ID1094.html DOI: 10.4267/2042/37704 This article is an update of: Huret JL, Leroux D, Bernheim A. t(8;13)(p12;q12). Atlas Genet Cytogenet Oncol Haematol.1998;2(3):95-96. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity t(8;13)(p12;q12) G- banding - Top: Courtesy Melanie Zenger and Claudia Haferlach; Middle and bottom: Courtesy Charles Bangs and Patty Jones. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 39 t(8;13)(p12;q12) Pébusque MJ, Cross NCP der(13) was found during disease progression, suggesting that the crucial event might lie on this derivative chromosome; +8, +21 are also recurrently found. Clinics and pathology Disease A myeloproliferative disorder that is frequently associated with T cell, or less commonly, B-cell non Hodgkin lymphoma. Genes involved and proteins Phenotype/cell stem origin FGFR1 May involve a stem cell involving both myeloid, T lineage, and B-cell lineage. Location: 8p12 Epidemiology Location: 13q12 Protein zinc finger protein (ten repeats in the N-terminal region with the consensus sequence C-X2-C-X18-24-(F/Y)-CX3-C that corresponds to a novel type of zing finger motifs), a hydrophobic repeat (proline-rich), and potentially two putative nuclear localisation signals. ZNF198 (also called FIM or ID_P) 14 cases are described; median age 43 yrs (range 1868); sex ratio: 6M/8F. Clinics Aggressive disease; complex picture of myeloid hyperplasia progressing to myelodysplasia and Tor -Bcell lymphoma; enlarged lymph node; blood data: high WBC (median 40 X 109/l); myelemia; monocytosis and eosinophilia. Result of the chromosomal anomaly Evolution Hybrid gene The disease transforms to ANLL, or occasionally ALL, in a median of 6 months. Prognosis Description Breakpoint in FGFR1 intron 8. Median survival: 12 months. Fusion protein Cytogenetics Description Aberrant tyrosine kinase composed of the N-term twothirds of FIM (retaining the 10 putative zinc finger motifs), and the FGFR1 intracellular region minus the major part of the juxtamembrane domain (and deleting the N-term immunoglobulin-like and central transmembrane domains of FGFR1). Expression / Localisation Cytoplasmic. Oncogenesis Through constitutive activation of FGFR1 signal transduction pathways, possibly via dimerization capability mediated by the FIM N-term sequences of the fusion protein. Cytogenetics morphological The same t(8;13) is found both in the bone marrow and in the lymph node, ruling out the hypothesis of a leukemoid reaction caused by a lymphoma; the multilineage involvement suggests the malignant transformation of a primitive hematopoietic stem cell. Probes Megac Yacs 770-c-2 (1390 kb) and 959-a-4 (1260kb), 856-b-6, 967; 899e2 - (CEPH); BAC 7M15; PAC RPCI 20-G12; FGFR1-specific cosmid 134.8; Additional anomalies Usually occurs as a single anomaly; duplication of the Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 40 t(8;13)(p12;q12) Pébusque MJ, Cross NCP To be noted receptor 1 gene to a novel gene RAMP. Hum Mol Genet. 1998 Apr;7(4):637-42 Case Report t(8;13)(p12;q12) in an atypical chronic myeloid leukaemia case. Xiao S, Nalabolu SR, Aster JC, Ma J, Abruzzo L, Jaffe ES, Stone R, Weissman SM, Hudson TJ, Fletcher JA. FGFR1 is fused with a novel zinc-finger gene, ZNF198, in the t(8;13) leukaemia/lymphoma syndrome. Nat Genet. 1998 Jan;18(1):84-7 References Kulkarni S, Reiter A, Smedley D, Goldman JM, Cross NC. The genomic structure of ZNF198 and location of breakpoints in the t(8;13) myeloproliferative syndrome. Genomics. 1999 Jan 1;55(1):118-21 Aguiar RC, Chase A, Coulthard S, Macdonald DH, Carapeti M, Reiter A, Sohal J, Lennard A, Goldman JM, Cross NC. Abnormalities of chromosome band 8p11 in leukemia: two clinical syndromes can be distinguished on the basis of MOZ involvement. Blood. 1997 Oct 15;90(8):3130-5 Ollendorff V, Guasch G, Isnardon D, Galindo R, Birnbaum D, Pébusque MJ. Characterization of FIM-FGFR1, the fusion product of the myeloproliferative disorder-associated t(8;13) translocation. J Biol Chem. 1999 Sep 17;274(38):26922-30 Popovici C, Adélaïde J, Ollendorff V, Chaffanet M, Guasch G, Jacrot M, Leroux D, Birnbaum D, Pébusque MJ. Fibroblast growth factor receptor 1 is fused to FIM in stem-cell myeloproliferative disorder with t(8;13). Proc Natl Acad Sci U S A. 1998 May 12;95(10):5712-7 Smedley D, Demiroglu A, Abdul-Rauf M, Heath C, Cooper C, Shipley J, Cross NC. ZNF198-FGFR1 transforms Ba/F3 cells to growth factor independence and results in high level tyrosine phosphorylation of STATS 1 and 5. Neoplasia. 1999 Oct;1(4):349-55 Reiter A, Sohal J, Kulkarni S, Chase A, Macdonald DH, Aguiar RC, Gonçalves C, Hernandez JM, Jennings BA, Goldman JM, Cross NC. Consistent fusion of ZNF198 to the fibroblast growth factor receptor-1 in the t(8;13)(p11;q12) myeloproliferative syndrome. Blood. 1998 Sep 1;92(5):1735-42 Xiao S, McCarthy JG, Aster JC, Fletcher JA. ZNF198-FGFR1 transforming activity depends on a novel proline-rich ZNF198 oligomerization domain. Blood. 2000 Jul 15;96(2):699-704 Smedley D, Hamoudi R, Clark J, Warren W, Abdul-Rauf M, Somers G, Venter D, Fagan K, Cooper C, Shipley J. The t(8;13)(p11;q11-12) rearrangement associated with an atypical myeloproliferative disorder fuses the fibroblast growth factor Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Pébusque MJ, Cross NCP. t(8;13)(p12;q12). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):39-41. 41 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication t(9;12)(q34;p13) Nyla A Heerema The Ohio State University, Division of Clinical Pathology, Department of Pathology, 167 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, USA (NAH) Published in Atlas Database: December 2000 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/t912ID1080.html DOI: 10.4267/2042/37705 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Protein Contains Helix-Loop-Helix (HLH) at N-terminal end and ETS DNA binding domain at C-terminal end; wide expression; nuclear localization; ETS- related transcription factor. Clinics and pathology Disease Described in only 6 cases; acute lymphoblastic leukemia (ALL), acute non lymphocytic leukemia (ANLL) and chronic myeloid leukemia (CML). Result of the chromosomal anomaly Prognosis Numbers small, but one CML case had allogeneic BMT and is in complete remission, the remaining cases had rapid disease progression and died of shortly after diagnosis. Hybrid gene Description 5’ ETV6–3’ ABL; two different fusion breakpoints have been described; ETV6 exon 4 fused in frame to ABL exon 2 (Type A) and ETV6 exon 5 fused in frame to ABL exon 2 (Type B); ETV6 maintains the HLH domain and ABL the tyrosine kinase domain. Cytogenetics Cytogenetics morphological t(9;12)(q34;p13), cryptic at the cytogenetic level. Fusion protein Variants Description a 155 kDa protein in Type A, 180 kDa protein in Type B; has elevated tyrosine kinase activity, localized in the cytoplasm and co-localizes with the actin filaments of the cells. Oncogenesis The HLH domain of ETV6 induces oligomerization, which results in the constitutive activation of the kinase domain of ABL; this is thought to result in phosphorylation of JAK2 and activation of the STAT pathway. Biological activity very similar to BCR-ABL. t(9;12;14)(q34;p13;q22) and complex insertions of ETV6 into ABL. Genes involved and proteins ABL Location: 9q34 DNA/RNA ETV6 is fused to exon 2 of ABL in the three cases described. Protein Tyrosine kinase, localized primarily to the nucleus. References ETV6 Papadopoulos P, Ridge SA, Boucher CA, Stocking C, Wiedemann LM. The novel activation of ABL by fusion to an ets-related gene, TEL. Cancer Res. 1995 Jan 1;55(1):34-8 Location: 12p13 DNA/RNA 9 exons; alternate splicing. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 42 t(9;12)(q34;p13) Heerema NA Golub TR, Goga A, Barker GF, Afar DE, McLaughlin J, Bohlander SK, Rowley JD, Witte ON, Gilliland DG. Oligomerization of the ABL tyrosine kinase by the Ets protein TEL in human leukemia. Mol Cell Biol. 1996 Aug;16(8):410716 Lacronique V, Boureux A, Monni R, Dumon S, Mauchauffé M, Mayeux P, Gouilleux F, Berger R, Gisselbrecht S, Ghysdael J, Bernard OA. Transforming properties of chimeric TEL-JAK proteins in Ba/F3 cells. Blood. 2000 Mar 15;95(6):2076-83 Van Limbergen H, Beverloo HB, van Drunen E, Janssens A, Hählen K, Poppe B, Van Roy N, Marynen P, De Paepe A, Slater R, Speleman F. Molecular cytogenetic and clinical findings in ETV6/ABL1-positive leukemia. Genes Chromosomes Cancer. 2001 Mar;30(3):274-82 Andreasson P, Johansson B, Carlsson M, Jarlsfelt I, Fioretos T, Mitelman F, Höglund M. BCR/ABL-negative chronic myeloid leukemia with ETV6/ABL fusion. Genes Chromosomes Cancer. 1997 Nov;20(3):299-304 Hannemann JR, McManus DM, Kabarowski JH, Wiedemann LM. Haemopoietic transformation by the TEL/ABL oncogene. Br J Haematol. 1998 Jul;102(2):475-85 This article should be referenced as such: Heerema NA. t(9;12)(q34;p13). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):42-43. Gesbert F, Griffin JD. Bcr/Abl activates transcription of the BclX gene through STAT5. Blood. 2000 Sep 15;96(6):2269-76 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 43 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication +3 or trisomy 3 in non Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi Hematology Section, Department of Biomedical Sciences, University of Ferrara, Corso Giovecca 203, Ferrara, Italy (AC) Published in Atlas Database: January 2001 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/tri3NHLID2008.html DOI: 10.4267/2042/37711 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity +3 (right) and partial trisomy 3 due to i(3)(q10) (left) FISH - Courtesy Hossein Mossafa. exception of marginal zone lymphomas (MZL) and mantle cell lymphoma (MCL); in MZL, total or partial trisomy 3 may occur in 50-70% of cytogenetically abnormal cases, with a reported incidence by interphase FISH in the 50-85% range; the incidence does not appear to vary according to the clinicopathologic features, with similar frequency in the extra-nodal MALT lymphoma, in the nodal and the splenic form of MZL; trisomy 3/3q was reported in 10-15% of MCL with an higher incidence (up to 40%) by molecular cytogenetic techniques; sporadically, other low-grade and high grade B-lymphoid tumors may carry trisomy 3/3q. Clinics and pathology Disease Trisomy 3 occurs more frequently in T-cell lymphomas than in B-cell lymphomas. Globally, 20-30% of T-NHL may carry trisomy 3, the highest incidence having been noted in lymphoepithelioid lymphoma, in low-grade peripheral T-cell lymphoma, in angioimmonoblastic lymphadenopathy and in adult T-cell leukemialymphoma. Trisomy 3 is relatively rare in B-NHL, with the Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 44 +3 or trisomy 3 in non Hodgkin's lymphoma (NHL) Cuneo A, Castoldi GL on Chromosomes in Leukemia-Lymphoma. Blood. 1987 Nov;70(5):1554-64 Prognosis The prognostic significance of trisomy 3 in T-cell and B-cell lymphomas is unknown; there does not appear to be a role for trisomy 3 in tumor progression from lowgrade MALT lymphoma to the high grade form, whereas gains of 3q may be associated with the aggressive blastoid variant of MCL. Schlegelberger B, Himmler A, Gödde E, Grote W, Feller AC, Lennert K. Cytogenetic findings in peripheral T-cell lymphomas as a basis for distinguishing low-grade and high-grade lymphomas. Blood. 1994 Jan 15;83(2):505-11 Dierlamm J, Pittaluga S, Wlodarska I, Stul M, Thomas J, Boogaerts M, Michaux L, Driessen A, Mecucci C, Cassiman JJ, De Wolf-Peeters C, Van den Berghe H. Marginal zone Bcell lymphomas of different sites share similar cytogenetic and morphologic features. Blood. 1996 Jan 1;87(1):299-307 Cytogenetics Cytogenetics morphological Michaux L, Dierlamm J, Wlodarska L, Criel A, Louwagie A, Ferrant A, Hagemeijer A, Van den Berghe H. Trisomy 3q11q29 is recurrently observed in B-cell non-Hodgkin's lymphomas associated with cold agglutinin syndrome. Ann Hematol. 1998 May;76(5):201-4 Trisomy 3 may be total or partial; commonly overrepresented segments in partial trisomy 3 include the q21-23 region and the q25-29 region; total/partial trisomy 3 may occur as an isolated anomaly in a minority of cases. Beà S, Ribas M, Hernández JM, Bosch F, Pinyol M, Hernández L, García JL, Flores T, González M, LópezGuillermo A, Piris MA, Cardesa A, Montserrat E, Miró R, Campo E. Increased number of chromosomal imbalances and high-level DNA amplifications in mantle cell lymphoma are associated with blastoid variants. Blood. 1999 Jun 15;93(12):4365-74 Cytogenetics molecular The anomaly is readily detectable by G- and R-banding in most cases; however, FISH using a centromeric probe is more sensitive than conventional cytogenetics, allowing for the study of non-dividing cells and for the detection of partial trisomy in complex karyotypes with marker chromosomes. Hoeve MA, Gisbertz IA, Schouten HC, Schuuring E, Bot FJ, Hermans J, Hopman A, Kluin PM, Arends JW, van Krieken JH. Gastric low-grade MALT lymphoma, high-grade MALT lymphoma and diffuse large B cell lymphoma show different frequencies of trisomy. Leukemia. 1999 May;13(5):799-807 Genes involved and proteins Bigoni R, Cuneo A, Milani R, Roberti MG, Bardi A, Rigolin GM, Cavazzini F, Agostini P, Castoldi G. Secondary chromosome changes in mantle cell lymphoma: cytogenetic and fluorescence in situ hybridization studies. Leuk Lymphoma. 2001 Feb;40(5-6):581-90 Note The gene(s) involved in the transformation process by gene dosage effect or by other mechanisms are not known. This article should be referenced as such: Cuneo A, Castoldi GL. +3 or trisomy 3 in non Hodgkin's lymphoma (NHL). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):44-45. References . Correlation of chromosome abnormalities with histologic and immunologic characteristics in non-Hodgkin's lymphoma and adult T cell leukemia-lymphoma. Fifth International Workshop Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 45 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Mini Review 11q23 rearrangements in leukaemia Jean-Loup Huret Genetics, Dept Medical Information, UMR 8125 CNRS, University of Poitiers, CHU Poitiers Hospital, F86021 Poitiers, France (JLH) Published in Atlas Database: January 2001 Online updated version: http://AtlasGeneticsOncology.org/Anomalies/11q23ID1030.html DOI: 10.4267/2042/37706 This article is an update of: Huret JL. 11q23 rearrangements in leukaemia. Atlas Genet Cytogenet Oncol Haematol.1998;2(4):137-139. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Clinics and pathology Cytogenetics Disease Cytogenetics morphological De novo and therapy related leukaemias; acute non lymphocytic leukaemia (ANLL) and acute lymphocytic leukaemia (ALL) grossly represent half cases each; myelodysplasia (MDS) in the remaining 5%; biphenotypic leukaemia at times (likely to be more frequent with more investigations); 11q23 rearrangements in treatment related leukaemias (5-10% of 11q23 cases) are found mainly following a treatment with anti-topoisomerase II, or an intercalating topoisomerase II inhibitor, but also after alkylating agents treatment and/or radiotherapy; the prior cancer is variable. I-The most frequent are: -Normal karyotype: a partial tandem duplication (in situ) of MLL is present in a percentage of ANLL with a normal karyotype; LARG, in 11q23, has been found fused to MLL; -+11: 1% of ANLL and MDS as well; M1, M2, and M4 ANLL; therapy related ANLL; MDS evolving towards ANLL; partial tandem duplication (in situ) of MLL; visible dup(11q) also occur; -t(4;11)(q21;q23): represent 1/3 of cases; found mainly (95%) in B-ALL (CD19+ in 75%, CD10+ in 15%); treatment related ALL in 5%; unbalanced sex ratio t(6;11)(q27;q23): 5% of cases; mostly; children and young adults; male predominance; the gene involved in 6q27 is AF6; role in signal transduction; -t(9;11)(p23;q23): represent 1/4 of cases; found in ANLL mainly in M5a (70%), or M4 (10%); in ALL in 10%; de novo and therapy related AL; children represent half cases (infants (- t(10;11)(p12;q23): 5% of cases; M4 or M5 ANLL; ALL at times; from infants and children to (rare) adult cases; the gene involved in 10p12 is AF10, a transcription activator; -t(11;19)(q23;p13.1): 5% of cases; M4 or M5 ANLL most often; de novo and therapy related AL; adults mainly; the gene involved in 19p13.1 is ELL, a transcription activator; -t(11;19)(q23;p13.3): 5% of cases; ALL, biphenotypic AL and ANLL (M4/M5 mainly); therapy related AL; T-cell ALL at times, these T-cell cases are the only cases of t(11;19) with an excellent prognosis, a rather Phenotype/cell stem origin ANLL: M5a in half cases, M4 (20%), M1 or M5b (10% each), M2 (5%); ALL: B-cell mostly, L1 or L2, CD19+ in 60% of B-ALL cases, CD10+ 35%; T-ALL in rare cases (<1%); MDS: most often RA or RAEB1T. Epidemiology 25% are infant (<1 yr) cases; children and adults each represent 50% of cases; M/F=0.9 (NS). Clinics Organomegaly; frequent CNS involvement (5%); high WBC (>50 X 109/l in 40%). Prognosis Very poor in general; variable according to the translocation, the phenotype, the age, and whether the leukaemia is de novo or treatment related. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 46 11q23 rearrangements in leukaemia Huret JL -t(11;19)(q23;p13): ANLL; the gene in 19p13 is EEN; -t(11;21)(q23;q11); -t(11;22)(q23;q13): ANLL; the gene in 22q13 is P300; -t(11;22)(q23;q11.2): ANLL; the gene in 22q11.2 is hCDCRel. III-Finally, various other breakpoints with 11q23 have been described, but without gene ascertainment: Xq24, 1q32, 2q37, 7q22, 7q32, 8q11, 9p11, 9q33, 12p13, 12q24, 14q11, 14q32, 17q11, 18q12, 20q13... rare feature in this page!!; mostly found in infants (half cases), and other children (altogether: 70%), or young adults (cases> 40 yrs are 4%; 23 unpublished cases and a review of 90 cases); the gene involved in 19p13.3 is ENL, a transcription activator. II-Various other 11q23 rearrangements have be described; these are rare, some are even poorly known, but the ones listed below are recurrent and/or with ascertainement of a partner gene to MLL: -inv(11)(p15q23): ANLL and MDS; -del(11q): one case (t-ANLL) showed involvement of GAS7, a gene sitting in 17p13; del(11q) with MLL rearrangement is likely to be heterogeneous, as MLL shows multiple possible partners, and, not rarely, complex translocations; -t(X;11)(q13;q23): ANLL; the gene involved in Xq13 is AFX1, a transcription regulator; -t(X;11)(q22;q23): the gene in Xq22 is Septin2; -t(1;11)(p32;q23): ALL and ANLL; the gene involved in 1p32 is AF1P; -t(1;11)(q21;q23): mostly M4 ANLL; the gene involved in 1q21 is AF1q; -t(2;11)(p21;q23): ANLL and MDS; may be found associated with del(5q); -t(2;11)(q11;q23): the gene in 2q11 is LAF4; -t(3;11)(p21;q23): the gene involved in 3p21 is AF3p21; -t(3;11)(q25;q23): the gene in 3q25 is GMPS; -t(5;11)(q31;q23) and ins(5;11)(q31;q13q23): the latter involve AF5q31 in 5q31; very rare; -t(5;11)(q31;q23): the gene in 5q31 is GRAF; -t(6;11)(q21;q23): ANLL; the gene in 6q21 is AF6q21, a transcription regulator; -t(9;11)(q34;q23): the gene in 9q34 is AF9q34; -t(10;11)(p11.2;q23): the gene in 10p11.2 is ABI1; -t(10;11)(q22;q23); -t(11;11)(q13;q23); -t(11;12)(q23;q13); -t(11;14)(q23;q24): the gene in 14q24 is h-gephyrin; -t(11;15)(q23;q14): the gene in 15q14 is AF15q14; -t(11;15)(q23;q15); -t(11;16)(q23;p13): treatment related ANLL/MDS; most cases are children cases; the gene involved in 16p13 is CBP, a transcriptional adaptor/coactivator; - t(11;17)(q23;p13): the gene in 17p13 is GAS7; - t(11;17)(q23;q12): the gene in 17q12 is RARa; - t(11;17)(q23;q21): ANLL; the gene involved in 17q21 is AF17; not to be confused with the in M3 ANLL variant, with involvement of PLZF in 11q23 and RARA in 17q21; -t(11;17)(q23;q25): ANLL and MDS; the gene in 17q25 is MSF/AF17q25; -t(11;18)(q23;q23); Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Additional anomalies +X and i(7q) in the t(4;11); +8, +19, +21 in the t(6;11); +8 and +19 in the t(9;11); inv(11) in the t(10;11); +X, +6 and +8 in the 19p13.3; +8 in the 19p13.1. Genes involved and proteins MLL Location: 11q23 DNA/RNA 21 exons, spanning over 100 kb; 13-15 kb mRNA; coding sequence: 11.9 kb. Protein 431 kDa; contains two DNA binding motifs (a AT hook, and Zinc fingers), a DNA methyl transferase motif, a bromodomain; transcriptional regulatory factor; nuclear localisation; wide expression; homology with trithorax (drosophila). Variable gene, from a variable chromosome partner (see below) DNA/RNA These genes appear to have, in most cases, no apparent homology to each other; for DNA and protein description of each, refer to their gene entry. Result of the chromosomal anomaly Hybrid gene Description 5' MLL-3' partner; highly variable breakpoints on the partner. Fusion protein Description N-term AT hook and DNA methyltransferase from MLL fused to (little or most of) the partner C-term part; the reciprocal (partner-MLL) may or may not be expressed. 47 11q23 rearrangements in leukaemia Huret JL Huret JL, Brizard A, Slater R, Charrin C, Bertheas MF, Guilhot F, Hählen K, Kroes W, van Leeuwen E, Schoot EV. Cytogenetic heterogeneity in t(11;19) acute leukemia: clinical, hematological and cytogenetic analyses of 48 patients-updated published cases and 16 new observations. Leukemia. 1993 Feb;7(2):152-60 References Lampert F, Harbott J, Ludwig WD, Bartram CR, Ritter J, Gerein V, Neidhardt M, Mertens R, Graf N, Riehm H. Acute leukemia with chromosome translocation (4;11): 7 new patients and analysis of 71 cases. Blut. 1987 Jun;54(6):325-35 Pui CH, Carroll LA, Raimondi SC, Shuster JJ, Crist WM, Pullen DJ. Childhood acute lymphoblastic leukemia with the t(4;11)(q21;q23): an update. Blood. 1994 Apr 15;83(8):2384-5 Ingram L, Raimondi SC, Mirro J Jr, Rivera GK, Ragsdale ST, Behm F. Characteristics of trisomy 11 in childhood acute leukemia with review of the literature. Leukemia. 1989 Oct;3(10):695-8 Bernard OA, Berger R. Molecular basis of 11q23 rearrangements in hematopoietic malignant proliferations. Genes Chromosomes Cancer. 1995 Jun;13(2):75-85 Albain KS, Le Beau MM, Ullirsch R, Schumacher H. Implication of prior treatment with drug combinations including inhibitors of topoisomerase II in therapy-related monocytic leukemia with a 9;11 translocation. Genes Chromosomes Cancer. 1990 May;2(1):53-8 Schichman SA, Canaani E, Croce CM. Self-fusion of the ALL1 gene. A new genetic mechanism for acute leukemia. JAMA. 1995 Feb 15;273(7):571-6 Pui CH, Frankel LS, Carroll AJ, Raimondi SC, Shuster JJ, Head DR, Crist WM, Land VJ, Pullen DJ, Steuber CP. Clinical characteristics and treatment outcome of childhood acute lymphoblastic leukemia with the t(4;11)(q21;q23): a collaborative study of 40 cases. Blood. 1991 Feb 1;77(3):440-7 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Rubnitz JE, Behm FG, Downing JR. 11q23 rearrangements in acute leukemia. Leukemia. 1996 Jan;10(1):74-82 Young BD, Saha V. Chromosome abnormalities in leukaemia: the 11q23 paradigm. Cancer Surv. 1996;28:225-45 48 11q23 rearrangements in leukaemia Huret JL Ida K, Kitabayashi I, Taki T, Taniwaki M, Noro K, Yamamoto M, Ohki M, Hayashi Y. Adenoviral E1A-associated protein p300 is involved in acute myeloid leukemia with t(11;22)(q23;q13). Blood. 1997 Dec 15;90(12):4699-704 Swansbury GJ, Slater R, Bain BJ, Moorman AV, SeckerWalker LM. Hematological malignancies with t(9;11)(p2122;q23)--a laboratory and clinical study of 125 cases. European 11q23 Workshop participants. Leukemia. 1998 May;12(5):792-800 Rowley JD, Reshmi S, Sobulo O, Musvee T, Anastasi J, Raimondi S, Schneider NR, Barredo JC, Cantu ES, Schlegelberger B, Behm F, Doggett NA, Borrow J, Zeleznik-Le N. All patients with the T(11;16)(q23;p13.3) that involves MLL and CBP have treatment-related hematologic disorders. Blood. 1997 Jul 15;90(2):535-41 Taki T, Shibuya N, Taniwaki M, Hanada R, Morishita K, Bessho F, Yanagisawa M, Hayashi Y. ABI-1, a human homolog to mouse Abl-interactor 1, fuses the MLL gene in acute myeloid leukemia with t(10;11)(p11.2;q23). Blood. 1998 Aug 15;92(4):1125-30 Anderson MJ, Viars CS, Czekay S, Cavenee WK, Arden KC. Cloning and characterization of three human forkhead genes that comprise an FKHR-like gene subfamily. Genomics. 1998 Jan 15;47(2):187-99 Fleischman EW, Reshmi S, Frenkel MA, Konovalova WI, Guleva GP, Kulagina OE, Konstantinova LN, Tupitsyn NN, Rowley JD. MLL is involved in a t(2;11)(p21;q23) in a patient with acute myeloblastic leukemia. Genes Chromosomes Cancer. 1999 Feb;24(2):151-5 Harrison CJ, Cuneo A, Clark R, Johansson B, LafagePochitaloff M, Mugneret F, Moorman AV, Secker-Walker LM. Ten novel 11q23 chromosomal partner sites. European 11q23 Workshop participants. Leukemia. 1998 May;12(5):811-22 Pegram LD, Megonigal MD, Lange BJ, Nowell PC, Rappaport EF, Felix CA.. t(3;11)(q25;q23) fuses MLL with the GMPS (guanosine 5'-monophosphate synthetase) gene in treatmentrelated acute myeloid leukemia (AML). Blood. 1999; 94 (numero Suppl 1). Johansson B, Moorman AV, Haas OA, Watmore AE, Cheung KL, Swanton S, Secker-Walker LM. Hematologic malignancies with t(4;11)(q21;q23)--a cytogenetic, morphologic, immunophenotypic and clinical study of 183 cases. European 11q23 Workshop participants. Leukemia. 1998 May;12(5):77987 Robert L. Redner, Susanne M. Gollin, Sandra S. Kaplan, Sofia Shekhter-Levin. Involvement of the MLL and RARA genes in a patient with acute monocytic leukemia with t(11;17)(q23;q12). Blood 1999;94 Suppl 1:Abst 3978 Johansson B, Moorman AV, Secker-Walker LM. Derivative chromosomes of 11q23-translocations in hematologic malignancies. European 11q23 Workshop participants. Leukemia. 1998 May;12(5):828-33 Taki T, Kano H, Taniwaki M, Sako M, Yanagisawa M, Hayashi Y. AF5q31, a newly identified AF4-related gene, is fused to MLL in infant acute lymphoblastic leukemia with ins(5;11)(q31;q13q23). Proc Natl Acad Sci U S A. 1999 Dec 7;96(25):14535-40 Lillington DM, Young BD, Berger R, Martineau M, Moorman AV, Secker-Walker LM. The t(10;11)(p12;q23) translocation in acute leukaemia: a cytogenetic and clinical study of 20 patients. European 11q23 Workshop participants. Leukemia. 1998 May;12(5):801-4 Borkhardt A, Bojesen S, Haas OA, Fuchs U, Bartelheimer D, Loncarevic IF, Bohle RM, Harbott J, Repp R, Jaeger U, Viehmann S, Henn T, Korth P, Scharr D, Lampert F. The human GRAF gene is fused to MLL in a unique t(5;11)(q31;q23) and both alleles are disrupted in three cases of myelodysplastic syndrome/acute myeloid leukemia with a deletion 5q. Proc Natl Acad Sci U S A. 2000 Aug 1;97(16):9168-73 Martineau M, Berger R, Lillington DM, Moorman AV, SeckerWalker LM. The t(6;11)(q27;q23) translocation in acute leukemia: a laboratory and clinical study of 30 cases. EU Concerted Action 11q23 Workshop participants. Leukemia. 1998 May;12(5):788-91 Hayette S, Tigaud I, Vanier A, Martel S, Corbo L, Charrin C, Beillard E, Deleage G, Magaud JP, Rimokh R. AF15q14, a novel partner gene fused to the MLL gene in an acute myeloid leukaemia with a t(11;15)(q23;q14). Oncogene. 2000 Sep 7;19(38):4446-50 Megonigal MD, Rappaport EF, Jones DH, Williams TM, Lovett BD, Kelly KM, Lerou PH, Moulton T, Budarf ML, Felix CA. t(11;22)(q23;q11.2) In acute myeloid leukemia of infant twins fuses MLL with hCDCrel, a cell division cycle gene in the genomic region of deletion in DiGeorge and velocardiofacial syndromes. Proc Natl Acad Sci U S A. 1998 May 26;95(11):6413-8 Kuwada N, Kimura F, Matsumura T, Yamashita T, Nakamura Y, Ikeda T, Sato K, Motoyoshi K.. A human homologue of the rat gephyrin gene is fused to MLL in a de novo leukemia with t(11;14)(q23;q24). Blood. 2000; 96 (numero Suppl 1). Moorman AV, Hagemeijer A, Charrin C, Rieder H, SeckerWalker LM. The translocations, t(11;19)(q23;p13.1) and t(11;19)(q23;p13.3): a cytogenetic and clinical profile of 53 patients. European 11q23 Workshop participants. Leukemia. 1998 May;12(5):805-10 Slater DJ, Hilgenfeld E, Rappaport EF, Shah NR, Megonigal MD, Ried T, Felix CA.. Identification of septin 2 as a new partner gene of MLL in infant aml with a complex translocation. Blood. 2000; 96 (numero Suppl 1). Secker-Walker LM. General Report on the European Union Concerted Action Workshop on 11q23, London, UK, May 1997. Leukemia. 1998 May;12(5):776-8 von Bergh A, Beverloo B, Slater R, Groot A, Rombout P, Kluin P, Schuuring E.. Cloning of unknown MLL fusion transcripts identifies two novel MLL fusion partners. Blood. 2000; 96 (numero Suppl 1). Secker-Walker LM, Moorman AV, Bain BJ, Mehta AB. Secondary acute leukemia and myelodysplastic syndrome with 11q23 abnormalities. EU Concerted Action 11q23 Workshop. Leukemia. 1998 May;12(5):840-4 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Huret JL. 11q23 rearrangements in leukaemia. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):46-49. 49 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication del(11q) in non-Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi Hematology Section, Department of Biomedical Sciences, University of Ferrara, Corso Giovecca 203, Ferrara, Italy (AC, GLC) Published in Atlas Database: January 2001 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/del11qNHLID2020.html DOI: 10.4267/2042/37707 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity del(11q) G- banding (the 3 left partial karyotypes) - Courtesy Diane H. Norback, Eric B. Johnson, Sara Morrison-Delap UW Cytogenetic Services; R-banding (right) - Editor Clinics and pathology Cytogenetics Disease Cytogenetics morphological The overall incidence in NHL is 4-5%, the highest incidence having been reported in mantle cell lymphoma, where up to 70% of the cases studied by FISH may harbour a cryptic deletion in association with the classical t(11;14) translocation; FISH detects an approximate 10% incidence of 11q deletion among other histologic subsets of B-NHL; among diffuse large B-cell lymphoma the 11q- chromosome shows a preferential association with the immunoblastic variant; sensitive molecular cytogenetic methods may show 5070% of T-cell prolymphocytic leukemia to carry an 11q deletion involving the ATM gene. The chromosome 11q deletion occurring in NHL most frequently affects the q22-23 bands; the 11q- anomaly occurs as a secondary change in the majority of cases. Prognosis Note The region of minimal deletion was narrowed down to a 2-3 Mb pair segment where the ataxia teleangiectasia (ATM) gene is located; sequencing studies showed mutation in the remaining ATM allele in a significant fraction of cases. Cytogenetics molecular Because the size of the deleted segment may be beyond the resolution power of conventional banding analysis, many cases can only be detected by interphase FISH or other genetic methods using probes targeting the 11q22.3-q23.1 region. Genes involved and proteins A possible association between 11q-/ATM- and poor prognosis in B-cell NHL was reported. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 50 del(11q) in non-Hodgkin's lymphoma (NHL) Cuneo A, Castoldi GL Stoppa-Lyonnet D, Soulier J, Laugé A, Dastot H, Garand R, Sigaux F, Stern MH. Inactivation of the ATM gene in T-cell prolymphocytic leukemias. Blood. 1998 May 15;91(10):3920-6 References . Correlation of chromosome abnormalities with histologic and immunologic characteristics in non-Hodgkin's lymphoma and adult T cell leukemia-lymphoma. Fifth International Workshop on Chromosomes in Leukemia-Lymphoma. Blood. 1987 Nov;70(5):1554-64 Schlegelberger B, Zwingers T, Harder L, Nowotny H, Siebert R, Vesely M, Bartels H, Sonnen R, Hopfinger G, Nader A, Ott G, Müller-Hermelink K, Feller A, Heinz R. Clinicopathogenetic significance of chromosomal abnormalities in patients with blastic peripheral B-cell lymphoma. Kiel-Wien-Lymphoma Study Group. Blood. 1999 Nov 1;94(9):3114-20 Vandenberghe E, De Wolf Peeters C, Wlodarska I, Stul M, Louwagie A, Verhoef G, Thomas J, Criel A, Cassiman JJ, Mecucci C. Chromosome 11q rearrangements in B non Hodgkin's lymphoma. Br J Haematol. 1992 Jun;81(2):212-7 Stilgenbauer S, Winkler D, Ott G, Schaffner C, Leupolt E, Bentz M, Möller P, Müller-Hermelink HK, James MR, Lichter P, Döhner H. Molecular characterization of 11q deletions points to a pathogenic role of the ATM gene in mantle cell lymphoma. Blood. 1999 Nov 1;94(9):3262-4 Stilgenbauer S, Liebisch P, James MR, Schröder M, Schlegelberger B, Fischer K, Bentz M, Lichter P, Döhner H. Molecular cytogenetic delineation of a novel critical genomic region in chromosome bands 11q22.3-923.1 in lymphoproliferative disorders. Proc Natl Acad Sci U S A. 1996 Oct 15;93(21):11837-41 Cuneo A, Bigoni R, Rigolin GM, Roberti MG, Milani R, Bardi A, Minotto C, Agostini P, De Angeli C, Narducci MG, Sabbioni S, Russo G, Negrini M, Castoldi G. Acquired chromosome 11q deletion involving the ataxia teleangiectasia locus in B-cell nonHodgkin's lymphoma: correlation with clinicobiologic features. J Clin Oncol. 2000 Jul;18(13):2607-14 Cuneo A, Bigoni R, Negrini M, Bullrich F, Veronese ML, Roberti MG, Bardi A, Rigolin GM, Cavazzini P, Croce CM, Castoldi G. Cytogenetic and interphase cytogenetic characterization of atypical chronic lymphocytic leukemia carrying BCL1 translocation. Cancer Res. 1997 Mar 15;57(6):1144-50 This article should be referenced as such: Cuneo A, Castoldi GL. del(11q) in non-Hodgkin's lymphoma (NHL). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):5051. Stilgenbauer S, Schaffner C, Litterst A, Liebisch P, Gilad S, Bar-Shira A, James MR, Lichter P, Döhner H. Biallelic mutations in the ATM gene in T-prolymphocytic leukemia. Nat Med. 1997 Oct;3(10):1155-9 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 51 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication del(7q) in non-Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi Hematology Section, Department of Biomedical Sciences, University of Ferrara, Corso Giovecca 203, Ferrara, Italy (AC, GLC) Published in Atlas Database: January 2001 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/del7qNHLID2082.html DOI: 10.4267/2042/37708 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology usually involve a relatively large segment, usually centered around the 7q22-q32 region Identity Cytogenetics molecular conventional G- or R-banded preparations detect the majority of cases; however some patients with submicroscopic deletion were detected by FISH or loss-of-heterozigosity studies Genes involved and proteins Note the involved gene(s) are unknown; the minimal region of deletion in MZBCL carrying a 7q- chromosome was narrowed down to a 5cM segment defined by the D7S685 and D7S514 markers; homozygous deletion of the D7S685 was reported, suggesting that a tumor suppressor gene relevant to lymphomagenesis may be located in this region; a recurrent 7q21 translocation involving a small 3.6 Kb segment upstream of the cyclin-dependent kinase 6 gene (CDK6), with resultant CDK6 overexpression, was described del(7q) in non-Hodgkin's lymphoma (NHL) G- banding Courtesy Melanie Zenger and Claudia Haferlach. Clinics and pathology Disease The frequency of 7q deletions in unseletced NHL is less than 5%; an association with splenic marginal zone B-cell lymphomas (MZBCL) was established, with a 20-30% incidence; sensitive molecular genetic studies found a 40% incidence in splenic MZBCL, as against a 7% incidence in other forms of NHL References Oscier DG, Matutes E, Gardiner A, Glide S, Mould S, BritoBabapulle V, Ellis J, Catovsky D. Cytogenetic studies in splenic lymphoma with villous lymphocytes. Br J Haematol. 1993 Nov;85(3):487-91 Prognosis There may be an association of 7q- with tumor progression or transformation into a high-grade MZBCL Corcoran MM, Mould SJ, Orchard JA, Ibbotson RE, Chapman RM, Boright AP, Platt C, Tsui LC, Scherer SW, Oscier DG. Dysregulation of cyclin dependent kinase 6 expression in splenic marginal zone lymphoma through chromosome 7q translocations. Oncogene. 1999 Nov 4;18(46):6271-7 Cytogenetics Cytogenetics morphological Mateo M, Mollejo M, Villuendas R, Algara P, Sanchez-Beato M, Martínez P, Piris MA. 7q31-32 allelic loss is a frequent finding in splenic marginal zone lymphoma. Am J Pathol. 1999 May;154(5):1583-9 7q deletions or unbalanced 7q translocations in NHL Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 52 del(7q) in non-Hodgkin's lymphoma (NHL) Cuneo A, Castoldi GL Cuneo A, Bigoni R, Roberti MG, Milani R, Agostini P, Cavazzini F, Minotto C, De Angeli C, Bardi A, Tammiso E, Negrini M, Cavazzini P, Castoldi G. Molecular cytogenetic characterization of marginal zone B-cell lymphoma: correlation with clinicopathologic findings in 14 cases. Haematologica. 2001 Jan;86(1):64-70 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Cuneo A, Castoldi GL. del(7q) in non-Hodgkin's lymphoma (NHL). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):5253. 53 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication t(1;14)(p22;q32) in non Hodgkin's lymphoma (NHL) Antonio Cuneo, Gianluigi Castoldi Hematology Section, Department of Biomedical Sciences, University of Ferrara, Corso Giovecca 203, Ferrara, Italy (AC) Published in Atlas Database: January 2001 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t114ID2044.html DOI: 10.4267/2042/37709 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Protein 322 amino acids; contains a caspase recruitment domain; role in the apoptosis. Clinics and pathology Disease IgH The translocation is cytogenetically detectable in a minority of extranodal MALT lymphomas; irrespective of the presence of the 1;14 translocation, mutation or deletion of the BCL10 gene located at 1p22 can be detected by molecular genetic methods in 5-10% of extra-nodal MALT lymphomas, follicle centre cell lymphoma and diffuse large B-cell lymphoma; among MALT lymphoma a preferential association was noted with high-grade histology. Location: 14q32 References Wotherspoon AC, Pan LX, Diss TC, Isaacson PG. Cytogenetic study of B-cell lymphoma of mucosa-associated lymphoid tissue. Cancer Genet Cytogenet. 1992 Jan;58(1):35-8 Willis TG, Jadayel DM, Du MQ, Peng H, Perry AR, Abdul-Rauf M, Price H, Karran L, Majekodunmi O, Wlodarska I, Pan L, Crook T, Hamoudi R, Isaacson PG, Dyer MJ. Bcl10 is involved in t(1;14)(p22;q32) of MALT B cell lymphoma and mutated in multiple tumor types. Cell. 1999 Jan 8;96(1):35-45 Prognosis In MALT lymphoma there may be an association with aggressive histology and antibiotic-unresponsive forms. Zhang Q, Siebert R, Yan M, Hinzmann B, Cui X, Xue L, Rakestraw KM, Naeve CW, Beckmann G, Weisenburger DD, Sanger WG, Nowotny H, Vesely M, Callet-Bauchu E, Salles G, Dixit VM, Rosenthal A, Schlegelberger B, Morris SW. Inactivating mutations and overexpression of BCL10, a caspase recruitment domain-containing gene, in MALT lymphoma with t(1;14)(p22;q32). Nat Genet. 1999 May;22(1):63-8 Cytogenetics Cytogenetics morphological The translocation is readily detectable by conventional karyotyping. Du MQ, Peng H, Liu H, Hamoudi RA, Diss TC, Willis TG, Ye H, Dogan A, Wotherspoon AC, Dyer MJ, Isaacson PG. BCL10 gene mutation in lymphoma. Blood. 2000 Jun 15;95(12):388590 Genes involved and proteins Note The breakpoints on chromosome 1p22 are located upstream of the promoter of the BCL10 gene, which shows inactivating mutations or deletions. This article should be referenced as such: Cuneo A, Castoldi GL. t(1;14)(p22;q32) in non Hodgkin's lymphoma (NHL). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):54. BCL10 Location: 1p22 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 54 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Short Communication t(5;10)(q33;q21) Cristina Mecucci Hematology, Policlinico Monteluce, Via Brunamonti, 06123 Perugia, Italy (CM) Published in Atlas Database: January 2001 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t510ID1166.html DOI: 10.4267/2042/37710 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Clinics and pathology Genes involved and proteins Disease PDGFRB (Platelet Derived Growth Factor Myeloid lineage. Receptor Beta) Location: 5q33 Protein Transmembrane and tyrosine kinase domains. Phenotype/cell stem origin Atypical chronic myeloid leukemia (CML), Philadelphia negative, bcr-abl negative with eosinophilia. H4(D10S170) Epidemiology Location: 10q21 Protein Carboxyterminal putative cytoskeletal protein? Only two cases available. Clinics Massive splenomegaly in one case; some clinical features of accelerated CML. SH3 binding site; Result of the chromosomal anomaly Cytology Peripheral blood leukoerythroblastosis; bone marrow granulocytic hyperplasia, bone marrow fibrosis (grade III-IV reticulin), dry tap Hybrid gene Transcript H4-PDGFBR chimeric RNA constantly present. Pathology Extramedullary hemopoiesis in the spleen. References Treatment Siena S, Sammarelli G, Grimoldi MG, Schiavo R, Nozza A, Roncalli M, Mecucci C, Santoro A, Carlo-Stella C. New reciprocal translocation t(5;10)(q33;q22) associated with atypical chronic myeloid leukemia. Haematologica. 1999 Apr;84(4):369-72 Control of disease by hydroxyurea in both cases. Cytogenetics Cytogenetics morphological Kulkarni S, Heath C, Parker S, Chase A, Iqbal S, Pocock CF, Kaeda J, Cwynarski K, Goldman JM, Cross NC. Fusion of H4/D10S170 to the platelet-derived growth factor receptor beta in BCR-ABL-negative myeloproliferative disorders with a t(5;10)(q33;q21). Cancer Res. 2000 Jul 1;60(13):3592-8 Cytogenetic result: add(5q), del(10q). Probes cosB for PDGFBR (5q33); PAC29F6 for H4/D10S170 (10q21) This article should be referenced as such: Mecucci C. t(5;10)(q33;q21). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):55. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 55 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Leukaemia Section Mini Review -Y, Y loss in leukemia Daniel L Van Dyke Department of Medical Genetics, Henry Ford Health System, 2799 West Grand Boulevard, Clara Ford Pavillion, Detroit, MI 48202, USA (DLV) Published in Atlas Database: January 2001 Online updated version : http://AtlasGeneticsOncology.org/Anomalies/YlossID1089.html DOI: 10.4267/2042/37712 This article is an update of: Desangles F. Y loss in leukemia. Atlas Genet Cytogenet Oncol Haematol.1999;3(2):80-81. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology In MDS, the proportion of -Y cells has been observed to increase, decrease, remain stable, or fluctuate up and down on follow-up studies. In four cases of Hodgkin disease, simultaneous fluorescence immunophenotyping and FISH showed that the -Y cell population was probably independent of the Hodgkin disease in at least two of the patients. It is notable that the -Y cells represented fewer than 10-15% of the metaphase cells in all four cases. Identity Note Loss of the Y chromosome from individual metaphases is common in metaphase cells from both PHAstimulated lymphocytes and spontaneously dividing bone marrow cells. The frequency of Y loss is greater in older men, and the size of the 45,X,-Y cell population probably increases gradually with advancing age. (In females, a corollary loss of one X chromosome also occurs with advancing age.) This natural phenomenon challenges our ability to distinguish between a normal and a disease-associated 45,X,-Y clone. Cytology No known association. Prognosis In ANLL, a 45,X,-Y karyotype is believed to have an intermediate prognosis. In MDS, the prognosis appears to be neutral or favorable. There are insufficient data for MPD or lymphoproliferative disease. Clinics and pathology Disease Cytogenetics -Y is frenquently observed in myeloproliferative diseases (MPD), myelodysplasic syndromes (MDS), acute non lymphocytic leukemias (ANLL), and can also be seen in lymphoproliferations. Cytogenetics morphological In PHA-stimulated lymphocyte karyotype studies of males, about 2% have one or more cells with loss of the Y chromosome. Cells with -Y are observed more often in males over age 55 than in younger males. In all age groups, the proportion of -Y cells is usually under 10%. The pattern of Y loss is more striking in bone marrow aspirate karyotype studies. Here, clonal Y chromosome loss as a sole abnormality in the karyotype is a common finding. A 45,X,-Y karyotype is observed in about 6% of bone marrow karyotype studies from males, and it represents 15-20% of abnormal karyotypes. The frequency of -Y cells increases with advancing age and is significantly greater in cases with MDS, MPD, ANLL, or lymphoproliferative disease than in subjects Epidemiology In CML with t(9;22) and in ANLL with a t(8;21), loss of the Y chromosome tends to occurs at a younger age than in the general population. Clinics Partial or complete reappearance of the Y chromosome has been described in several cases of ANLL in remission. In most or all of these ANLL cases, the 45,X,-Y cell population represented 80-100% of preremission metaphases. These observations support the interpretation that the leukemia cell karyotype is 45,X,Y. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 56 -Y, Y loss in leukemia Van Dyke DL XXXV. The missing Y in acute non-lymphocytic leukemia (ANLL). Cancer. 1980 Jan 1;45(1):84-90 who have no evidence of disease. Subjects with no evidence of disease rarely exhibit more than 75% of cells with 45,X,-Y. Thus, if fewer than 75% of metaphase cells are -Y, the disease association is uncertain. However, if 75-100% of metaphase cells are -Y, the karyotype probably is disease-associated, even in older men. Chromosome rearrangements involving the Y chromosome are rare in cancer and leukemia. Loss of the Y chromosome, in contrast, is a common secondary change in cancer cells and in a few leukemias (see below). Holmes RI, Keating MJ, Cork A, Trujillo JM, McCredie KB, Freireich EJ. Loss of the Y chromosome in acute myelogenous leukemia: a report of 13 patients. Cancer Genet Cytogenet. 1985 Jul;17(3):269-78 . Acute myelogenous leukemia with an 8;21 translocation. A report on 148 cases from the Groupe Français de Cytogénétique Hématologique. Cancer Genet Cytogenet. 1990 Feb;44(2):169-79 Nowinski GP, Van Dyke DL, Tilley BC, Jacobsen G, Babu VR, Worsham MJ, Wilson GN, Weiss L. The frequency of aneuploidy in cultured lymphocytes is correlated with age and gender but not with reproductive history. Am J Hum Genet. 1990 Jun;46(6):1101-11 Probes All available probe for the Y chromosome. Additional anomalies . Loss of the Y chromosome from normal and neoplastic bone marrows. United Kingdom Cancer Cytogenetics Group (UKCCG) Genes Chromosomes Cancer. 1992 Jul;5(1):83-8 In association with t(9;22) in CML and with t(8;21) in FAB-M2 ANLL, loss of the Y chromosome is generally considered a secondary event of no added clinical significance. Abeliovich D, Yehuda O, Ben-Neriah S, Or R. Loss of Y chromosome. An age-related event or a cytogenetic marker of a malignant clone? Cancer Genet Cytogenet. 1994 Aug;76(1):70-1 Kirk JA, VanDevanter DR, Biberman J, Bryant EM. Y chromosome loss in chronic myeloid leukemia detected in both normal and malignant cells by interphase fluorescence in situ hybridization. Genes Chromosomes Cancer. 1994 Nov;11(3):141-5 Genes involved and proteins Note Genes involved, if any, are unknown. Riske CB, Morgan R, Ondreyco S, Sandberg AA. X and Y chromosome loss as sole abnormality in acute nonlymphocytic leukemia (ANLL) Cancer Genet Cytogenet. 1994 Jan;72(1):44-7 To be noted Note It is not known whether the Y chromosome loss is the critical mutational event. Likewise, it is not known whether the Y chromosome loss is a secondary genetic change, or if the critical (submicroscopic) genetic change simply occurs by chance in a -Y cell. Speculatively, loss of the Y could provide a proliferative advantage simply because it tends to replicate late in S-phase. Its loss might therefore shorten the cell cycle slightly. Weber-Matthiesen K, Deerberg J, Poetsch M, Grote W, Schlegelberger B. Clarification of dubious karyotypes in Hodgkin's disease by simultaneous fluorescence immunophenotyping and interphase cytogenetics (FICTION). Cytogenet Cell Genet. 1995;70(3-4):243-5 Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS, Mohamed A, Paietta E, Willman CL, Head DR, Rowe JM, Forman SJ, Appelbaum FR. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood. 2000 Dec 15;96(13):4075-83 References Wiktor A, Rybicki BA, Piao ZS, Shurafa M, Barthel B, Maeda K, Van Dyke DL. Clinical significance of Y chromosome loss in hematologic disease. Genes Chromosomes Cancer. 2000 Jan;27(1):11-6 Pierre RV, Hoagland HC. Age-associated aneuploidy: loss of Y chromosome from human bone marrow cells with aging. Cancer. 1972 Oct;30(4):889-94 Berger R, Bernheim A. Y chromosome loss in leukemias. Cancer Genet Cytogenet. 1979;1:1-8. This article should be referenced as such: Van Dyke DL. -Y, Y loss in leukemia. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):56-57. Abe S, Golomb HM, Rowley JD, Mitelman F, Sandberg AA. Chromosomes and causation of human cancer and leukemia. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 57 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Solid Tumour Section Mini Review Nervous system: Astrocytic tumors Anne-Marie Capodano Laboratoire de Cytogénétique Oncologique, Hôpital de la Timone, 264 rue Saint Pierre, 13005 Marseille, France (AMC) Published in Atlas Database: November 2000 Online updated version : http://AtlasGeneticsOncology.org/Tumors/AstrocytID5007.html DOI: 10.4267/2042/37713 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Clinics Classification 1- Pilocytic Astrocytomas/Grade I: pilocytic astrocytomas arise throughout the neuraxis and are common in children and in young adults; pilocytic tumors of the optic nerve cause loss of vision; pilocytic astrocytoma of the hypothalamus and third ventricular region primarily affect children; but tumors of the cerebral hemispheres generally occur in patients older than those with visual system or hypothalamic involvement. 2- Fibrillary Astrocytomas/Grade II: fibrillary astrocytomas arise in the cerebral hemisphere of young to middle-aged adults and the brain stem of children; occasional examples occur in the cerebellum or spinal cord; at any site these astrocytomas must be distinguished from pilocytic astrocytomas; all such tumors are pilocytic astrocytomas in the optic nerve whereas most are of the fibrillary type in the brain stem. 3- Anaplastic Astrocytomas / Grade III: anaplastic astrocytomas occur in the same locations as astrocytomas (I-II) and glioblastoma, but the majority affect the cerebral hemispheres; anaplastic astrocytomas generally occur in patients a decade older than those with better differenciated astrocytomas and a decade younger than those with glioblastomas. 4- Glioblastoma Multiforme / Grade IV: glioblastoma is by far the most common glioma; it affects principally the cerebral hemispheries in adults and the brain stem in children; but they are most frequent after the fifth decade; most glioblastomas are solitary but occasional examples are geographically separate in the same patient and warrant the designation " multicentric "; usually, it appears as a central area of hypodensity surrounded by a ring of contrast enhanced and penumbra of cerebral oedema. Note Astrocytic tumors comprise a wide range of neoplasms that differ in their location within the central nervous system (CNS), age and gender distribution, growth potential, extent of invasiveness, morphological features, tendency for progression and clinical course; there is increasing evidence that these differences reflect the type and sequence of genetic alterations acquired during the process of transformation. Classification The following clinicopathological entities can be distinguished: Pilocytic Astrocytomas (Grade I). Fibrillary Astrocytomas (Grade II). Anaplastic Astrocytomas (Grade III). Glioblastoma Multiforme (Grade IV). Clinics and pathology Etiology Gliomas have been observed following therapeutic irradiation. Familial clustering of gliomas is not uncommon: the association with defined inherited tumor syndrome incuding the Li-Fraumeni syndrome, Turcot syndrome, and the NF1 syndrome. Epidemiology Diffuse astrocytomas are the most frequent intracranial neoplasm and account for more than 60% of all primary brain tumors; the incidence differs between regions, but there are 5 to 7 new cases per 100.000 population per year. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 58 Nervous system: Astrocytic tumors Capodano AM Glioblastoma multiforme may develop de novo (primary glioblastoma) or though progression from low-grade or anaplastic astrocytoma (secondary glioblastoma); patients with a primary glioblastoma are usually older, present a rapid tumor progression and a poor prognosis; patient with secondary glioblastomas are younger and tumor progress more slowly, with a better prognosis; these two groups are histologically indistinguishable. time after surgery is 6-8 years in low-grade astrocytomas; after surgery, the prognosis depends on whether the neoplasm undergoes progression to a more malignant phenotype; in pilocytic astrocytomas, total cure is possible after total resection; in fibrillary astrocytomas reccurrence is frequent. In anaplastic astrocytomas and in glioblastomas, evaluation of the extent of resection can be a prognostic factor; prognosis is generally poor (about one year); patients below 45 yrs have a considerably better prognosis than elderly patients; primary glioblastomas have a short clinical history with a poor prognosis; survival is better in secondary glioblastomas. Pathology 1- Pilocytic Astrocytomas / Grade I: this predominantly peadiatric brain tumor is a circumscribed astrocytoma composed in varying proportions of compacted and loose textured astrocytes associated with rosenthal fibers, eosinophilic granular bodies, or both; the lesion described is sometimes referred to as the " juvenile pilocytic astrocytoma ". 2- Fibrillary Astrocytomas / Grade II: this tumor is a well differanciated diffusely infiltrating neoplasm of fibrillary astrocytes. 3- Anaplastic Astrocytomas / Grade III: this tumor is an astrocytic tumor of fibrillary type which is intermediate in differenciation between the better differenciated astrocytoma and glioblastoma; it is an astrocytic neoplasm that typically exceeds well differenciated astrocytoma in terms of cellularity, nuclear pleomorphism and hyperchromasia necrosis of glioblastoma. 4- Glioblastoma Multiforme / Grade IV: this tumor is a highly malignant glioma most closely related to fibrillary or diffuse astrocytic neoplasms; glioblastomas are cellular masses with varied tissue patterns; it appears either infiltrating or discrete, with typical or atypical mitoses, endothelial vascular proliferation and necrosis another subgroup of glioblastoma can be distinguished: the giant cell glioblastomas; histologically it is a glioblastoma with giant cells (500 mm in diameter): it develops clinically "de novo"; it is associated with a favorable prognosis Cytogenetics Cytogenetics Morphological In astrocytomas grade I, normal karyotype is observed most frequently; among the cases with abnormal karyotypes, the most frequent chromosomal abnormalityis loss of the X and Y sex- chromosomes; loss of 22q is found in 20-30% of astrocytomas; other abnormalities observed in low grade tumors include gains on chromosome 8q, 10p, and 12p, and losses on chromosomes 1p, 4q, 9p, 11p 16p, 18 and 19. In anaplastic astrocytomas, chromosome gains or losses are frequent: trisomy 7 (the most frequent), loss of chromosome 10, loss of chromosome 22, loss of 9p, 13q; other abnormalities, less frequently described are: gains of chromosomes 1q, 11q, 19, 20, and Xq. Glioblastomas show several chromosomal changes: by frequency order, gain of chromosome 7 (50-80% of glioblastomas), double minute chromosomes, total or partial monosomy for chromosome 10 (70% of tumors) associated with the later step in the progression of glioblastomas partial deletion of 9p is frequent (64% of tumors): 9pter-23; partial loss of 22q in 22q13 is frequently reported. Loss or deletion of chromosome 13, 13q14-q31 is found in some glioblastomas. Trisomy 19 was reported in glioblastomas by cytogenetic and comparative genomic hybridization (CGH) analysis; the loss of 19q in 19q13.2-qter was detected by loss of heterozigocity (LOH) studies in glioblastomas. Deletion of chromosome 4q, complete or partial gains of chromosome 20 has been described; gain or amplification of 12q14-q21 has been reported. The loss of chromosome Y might be considered, when it occurs in addition to other clonal abnormalities. Treatment Treatment differs according to grade and location of tumor. Pilocytic astrocytomas can be cured by complete resection of tumor; if exeresis is not possible due to the location of the tumor, chemotherapy is indicated in young children and radiotherapy in adults in fibrillary astrocytomas, the treatment consists of total and extent resection of tumor in anaplastic tumors and glioblastoma multiforme, the treatment consists of total resection and radiotherapy and chemotherapy after surgery. Genes involved and proteins Note Alteration of genes involved in cell-cycle control: it is known that the progression of the-cell cycle is controled by positive and negative regulators; some autors report alteration in cell-cycle gene expression in human brain tumors. The p16 gene and the p15 gene Prognosis In low grade astrocytomas, a correlation of proliferation was reported (Ki67 index) with clinical outcome; the proliferative potential correlates inversely with survival and time to recurrence; the mean survival Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 59 Nervous system: Astrocytic tumors Capodano AM The LG11 novel gene located in 10p24 region is a suppressor gene rearranged in several glioblastomas tumors. Allelic loss of chromosome 22q wich contains the neurofibromatosis type 2, tumor suppressor gene NF2 is observed in 20-30% of astrocytomas. But another possibility is the involvement of another gene located on chromosome 22 in the tumorogenesis of astrocytomas. Most of these genes participate in the progression of astrocytomas (fig 1). Expression of growth factors and growth factor receptors: The epidermal growth factor receptor (EGFR) coded by the EGFR cellular oncogene is located on human chromosome 7 at locus 7p12-p14; EGRF is amplified in 40-60% of glioblastomas; it constitues a hallmark: primary glioblastomas rarely contain EGFR overexpression; patients with anaplastic astrocytomas or glioblastomas have a poorer prognosis when EGFR gene amplification is present; amplification could be a significant prognostic factor in these tumors Over expression of PDGFR-a (platelet derived growth factor) is asociated with loss of heterozygosity of chromosome 17p and p53 mutations in secondary glioblastomas Others growth factors expressed in gliomas include fibroblast growth factors (FGFs), insulin-like growth factors (IGFs), and vascular endothelial growth factor (VEGF). are located in 9p21, a chromosome region commonly deleted in astrocytomas; expression of p16 gene is frequently altered in these tumors: in 33-68% of primary glioblastomas and 25% of anaplastic astrocytomas. The Rb gene located on13q chromosome plays an important role in the malignant progression of gliomas. The p53 gene is a tumor suppressor gene located on chromosome 17p13.1; loss or mutation of p53 gene has been detected in many types of gliomas and represents an early genetic event in these tumors. Overexpression of MDM2 is also seen in primary glioblastomas. Others oncogenes have been found to be amplified in a few cases of astrocytomas: oncogenes Gli, MYC, MYCN, MET and N-Ras. Loss or inactivation of tumor suppressor genes: In addition to p53 gene, others tumor suppression genes play a role in astrocytomas Loss of chromosome 10 is the most frequent abnormality associated with the progression of malignant astrocytic tumors; more than 70% of glioblastomas show LOH on chromosome 10; amplification of EGFR is always associated with loss of chromosome 10 The PTEN gene located at the 10q23 locus is implicated more frequently in glioblastomas than in anaplastic astrocytomas. Another suppressor gene the MXII gene has also been located on the distal portion of chromosome 10 at the 10q24 at the 10q24-p25 locus. Homozygous deletion in the DMTB gene located on the region 10q25.3-26.1 have been reported in glioblastomas. Molecular pathways in the progression of astrocytomas (from Ho-Keung and Paula Y.P. Lam). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 60 Nervous system: Astrocytic tumors Capodano AM genomic alterations associated with glioma progression by comparative genomic hybridization. Oncogene. 1996 Sep 5;13(5):983-94 References Bigner SH, Burger PC, Wong AJ, Werner MH, Hamilton SR, Muhlbaier LH, Vogelstein B, Bigner DD. Gene amplification in malignant human gliomas: clinical and histopathologic aspects. J Neuropathol Exp Neurol. 1988 May;47(3):191-205 Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittmann M, Tycko B, Hibshoosh H, Wigler MH, Parsons R. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997 Mar 28;275(5308):1943-7 Bigner SH, Mark J, Bigner DD. Cytogenetics of human brain tumors. Cancer Genet Cytogenet. 1990 Jul 15;47(2):141-54 Griffin CA, Long PP, Carson BS, Brem H. Chromosome abnormalities in low-grade central nervous system tumors. Cancer Genet Cytogenet. 1992 May;60(1):67-73 Rasheed BK, Stenzel TT, McLendon RE, Parsons R, Friedman AH, Friedman HS, Bigner DD, Bigner SH. PTEN gene mutations are seen in high-grade but not in low-grade gliomas. Cancer Res. 1997 Oct 1;57(19):4187-90 Rasheed BK, Fuller GN, Friedman AH, Bigner DD, Bigner SH. Loss of heterozygosity for 10q loci in human gliomas. Genes Chromosomes Cancer. 1992 Jul;5(1):75-82 Shapiro JR, Scheck AC. Brain tumors. In: Cytogenetic Cancer Markers. Wolman SR and Sell S (eds) 1997; Humana Press, Totowa, New Jersey, pp319-368. Thiel G, Losanowa T, Kintzel D, Nisch G, Martin H, Vorpahl K, Witkowski R. Karyotypes in 90 human gliomas. Cancer Genet Cytogenet. 1992 Feb;58(2):109-20 Steck PA, Pershouse MA, Jasser SA, Yung WK, Lin H, Ligon AH, Langford LA, Baumgard ML, Hattier T, Davis T, Frye C, Hu R, Swedlund B, Teng DH, Tavtigian SV. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet. 1997 Apr;15(4):356-62 Bello MJ, de Campos JM, Kusak ME, Vaquero J, Sarasa JL, Pestaña A, Rey JA. Molecular analysis of genomic abnormalities in human gliomas. Cancer Genet Cytogenet. 1994 Apr;73(2):122-9 Wechsler DS, Shelly CA, Petroff CA, Dang CV. MXI1, a putative tumor suppressor gene, suppresses growth of human glioblastoma cells. Cancer Res. 1997 Nov 1;57(21):4905-12 Rasheed BK, McLendon RE, Herndon JE, Friedman HS, Friedman AH, Bigner DD, Bigner SH. Alterations of the TP53 gene in human gliomas. Cancer Res. 1994 Mar 1;54(5):132430 Chernova OB, Somerville RP, Cowell JK. A novel gene, LGI1, from 10q24 is rearranged and downregulated in malignant brain tumors. Oncogene. 1998 Dec 3;17(22):2873-81 Schlegel J, Merdes A, Stumm G, Albert FK, Forsting M, Hynes N, Kiessling M. Amplification of the epidermal-growth-factorreceptor gene correlates with different growth behaviour in human glioblastoma. Int J Cancer. 1994 Jan 2;56(1):72-7 Ng HK, Lam PY. The molecular genetics of central nervous system tumors. Pathology. 1998 May;30(2):196-202 Collins VP. Gene amplification in human gliomas. Glia. 1995 Nov;15(3):289-96 Nishizaki T, Ozaki S, Harada K, Ito H, Arai H, Beppu T, Sasaki K. Investigation of genetic alterations associated with the grade of astrocytic tumor by comparative genomic hybridization. Genes Chromosomes Cancer. 1998 Apr;21(4):340-6 Moulton T, Samara G, Chung WY, Yuan L, Desai R, Sisti M, Bruce J, Tycko B. MTS1/p16/CDKN2 lesions in primary glioblastoma multiforme. Am J Pathol. 1995 Mar;146(3):613-9 Sehgal A. Molecular changes during the genesis of human gliomas. Semin Surg Oncol. 1998 Jan-Feb;14(1):3-12 Schwechheimer K, Huang S, Cavenee WK. EGFR gene amplification--rearrangement in human glioblastomas. Int J Cancer. 1995 Jul 17;62(2):145-8 Burger PC, Scheithauer BW, Paulus W, Giannini C, Kleihues P. Pilocytc astrocytoma.In Pathology and Genetics of Tumors of the Nervous System-Kleihues P, Cavenee WK (eds) 2000; IARC Press, pp 29-33. Rosenberg JE, Lisle DK, Burwick JA, Ueki K, von Deimling A, Mohrenweiser HW, Louis DN. Refined deletion mapping of the chromosome 19q glioma tumor suppressor gene to the D19S412-STD interval. Oncogene. 1996 Dec 5;13(11):2483-5 Goussia AC, Agnantis NJ, Rao JS, Kyritsis AP. Cytogenetic and molecular abnormalities in astrocytic gliomas (Review). Oncol Rep. 2000 Mar-Apr;7(2):401-12 Schlegel J, Scherthan H, Arens N, Stumm G, Kiessling M. Detection of complex genetic alterations in human glioblastoma multiforme using comparative genomic hybridization. J Neuropathol Exp Neurol. 1996 Jan;55(1):81-7 This article should be referenced as such: Capodano AM. Nervous system: Astrocytic tumors. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):58-61. Weber RG, Sabel M, Reifenberger J, Sommer C, Oberstrass J, Reifenberger G, Kiessling M, Cremer T. Characterization of Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 61 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Cancer Prone Disease Section Mini Review Beckwith-Wiedemann syndrome Marcel Mannens DNA-diagnostics laboratory, University of Amsterdam, Academic Medical Center Department of Clinical Genetics PO Box 22700 1100 DE Amsterdam, the Netherlands (MM) Published in Atlas Database: November 2000 Online updated version : http://AtlasGeneticsOncology.org/Kprones/BeckwithWiedemannID10037.html DOI: 10.4267/2042/37714 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Clinics Alias Exomphalos-macroglossia-gigantism triad Inheritance Incidence of 7/105; given the variable expression of the symptoms, the actual frequency is likely to be higher; generally there is sporadic occurrence of the syndrome (85%); inheritance is mostly maternal (imprinting) with a more severe phenotype after maternal transmission. Note Clinically and genetically heterogeneous; three distinct regions on 11p15 have been associated with BWS (BWSCR1/2/3); BWSCR2 seems to be particularly associated with hemihypertrophy. Phenotype and clinics Multiple features that occur variably; most prominent is the EMG triad (exomphalos-macroglossia-gigantism): apart from the abdominal wall defects and pre- and postnatal growth abnormalities, earlobe pits or creases, facial nevus flammeus, hypoglycemia, renal abnormalities and hemihypertrophy (unilateral overgrowth) are frequently seen. Patient with Beckwith-Wiedemann syndrome. The face shows the enlarged tongue (macroglossia), the ear the typical earlobe creases Marcel Mannens. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 62 Beckwith-Wiedemann syndrome Mannens M Neoplastic risk Protein Description: Untranslated. Expression: Highly expressed in endodermal and mesodermal embryonic tissues; in adult brain, only in the pons and globus pallidus; in adult tissues, expression is primarily confined to skeletal and cardiac muscle; other tissues are down-regulated postnatal but re-expressed in tumours that express the gene during embryogenesis. Function: Putative tumour suppressor; proposed regulatory function for IGF2 expression (under debate). Mutations Germinal: Hypermethylated in 10-20% of sporadic BWS cases; familial transmission unclear yet; loss of imprinting (LOI) can be induced in deletion mouse models. Somatic: Hypermethylated in 10-20% of sporadic BWS cases mostly somatic events due to UPD in mosaic form; LOI in tumours. The increased risk for childhood solid tumours is 7.5% (thousand fold increase); tumours most frequently seen are nephroblastoma (Wilms tumour), adrenocortical carcinoma, rhabdomyosarcoma and hepatoblastoma; clinical risk factors are hemihypertrophy and nephromegaly; genetic risk factors are uniparental disomy (UPD) and H19/IGF2 imprinting defects. Treatment In general surgical correction of the abdominal wall defects and macroglossia; monitoring the glycemia during the first 3 days and early treatment of hypoglycemia (deleterious for central nervous system) is of importance to avoid further complications; frequent screening for tumour development. Prognosis Clinical features tend to become less with ageing; tumour risk decreases strongly after the 4-7th year of birth. IGF2 (insulin-like growth factor 2 (somatomedin A)) Cytogenetics Alias: IGF-II, somatomedin A, Hs.75963 Location: 11p15.5 DNA/RNA Transcription: 1356 bp mRNA, paternally expressed, maternal imprint. Protein Description: 180 amino acids, 20,14 kDa (unprocessed). Expression: IGF2 has the highest levels of expression in tissues that are affected by prenatal overgrowth in BWS; the main source of expression is liver; expression depends on promoter usage; P1 is exclusively active in adult liver, whereas P3 and P4 exert their action in liver prenatal; P2 is only active in certain tumour cell lines. Localisation: Secreted. Function: Embryonal growth factor, mitogen. Homology: Belongs to the insulin/IGF/relaxin family. Mutations Germinal: Hypomethylated; LOI in sporadic BWS cases; familial transmission unclear yet; BWS phenotype can be induced in igf2 overexpressing mouse models. Somatic: Hypomethylated, LOI in sporadic BWS cases; mostly somatic events due to UPD in mosaic form; LOI in tumours. Inborn conditions Paternal duplications of chromosome region 11p15, maternal translocations involving chromosome region 11p15.3-p15.5. Cytogenetics of cancer Apart from chromosome 11 aberrations, multiple chromosomes are involved in tumour development; promising prognostic indicators in Wilms tumour might be chromosome 1p and 16q aberrations; tther molecular abnormalities associated with an adverse outcome in Wilms tumour are 22q allele loss or P53 aberrations. Other findings Note In 10-20% of BWS cases, uniparental disomy of chromosome region 11p15 is seen, mostly in a mosaic form. Genes involved and proteins H19 Alias: D11S813E, D11S878E, ASM, ASM1 Location: 11p15.5 Note Imprinted, maternally expressed, untranslated mRNA. DNA/RNA Description: The human H19 gene is 2.7 kb long and includes 4 small introns; maternally expressed, paternal imprint. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) CDKN1C (cyclin-dependent kinase inhibitor 1C) Alias: KIP2, P57KIP2, P57, CDKN5 Location: 11p15.5 63 Beckwith-Wiedemann syndrome Mannens M ZNF215 DNA/RNA Description: 1511 bp messenger, preferentially maternally expressed (paternal imprint). Protein Description: 316 amino acids; 32,177 kDa, CDK inhibitory domain, PAPA repeat, conserved C-terminal domain. Expression: It is expressed in the heart, brain, lung, skeletal muscle, kidney, pancreas and testis; high levels are seen in the placenta, low levels in liver. Localisation: Nuclear. Function Summary: Cyclin-dependent kinase inhibitor 1C is a tight-binding inhibitor of several G1 cyclin/Cdk complexes and a negative regulator of cell proliferation; mutations of CDKN1C are implicated in sporadic cancers and Beckwith-Wiedemann syndrome suggesting that it is a tumour suppressor candidate; in BWS however, no evidence for tumour association was found. Homology: p21CIP1 CdK inhibitor gene family. Mutations Germinal: Mostly maternal, nucleotide substitutions, small deletions. Somatic: CDKN1C mutations are described in tumour formation; mouse mutation-models reveal part of the BWS phenotype in particular the abdominal-wall defects. Alias: zinc finger protein 215, BAZ2 Location: 11p15.4 DNA/RNA Description: mRNA of 3480 bp, 9 exons, at least 5 splice variants; exon 9 runs antisense of a second gene: ZNF214. Transcription: Imprinted in a tissue specific manner, the maternal allele being preferentially expressed. Protein Description: 517 amino acids, 60,048 kDa; KRABA domain; similarities to a KRABB domain; SCAN box; nuclear localisation signal KKKR; 2 x 2 zinc-fingers. Expression: Widely expressed at low levels; expression is highest in testis; splice variants of ZNF215 show tissue specific expression. Localisation: Nuclear. Function: Putative transcription factor; ZNF215 was cloned from a region associated with hemihypertrophy, cardiac abnormalities, Wilms tumour and minor BWS features; as such the gene might be responsible for a distinct phenotype in BWS. Homology: Belongs to the Krueppel family of C2H2type zinc finger proteins. Mutations Germinal: Various amino acids substitutions found in BWS / hemihypertrophy patients; causal relationship with phenotype unclear. Somatic: In tumours no mutations found so far. KCNQ1OT1 (KCNQ1 overlapping transcript) References Alias: KCNQ1 overlapping transcript 1, LIT1, KvDMR1, KvLQT1-AS, Long QT intronic transcript 1 Location: 11p15.5 DNA/RNA Description: Maternally imprinted gene, > 80 kb RNA. Transcription: Intronic transcript 1, embedded in intron 9 (and 10) of KCNQ1, in opposite orientation; expressed in most human tissues and from the paternal allele, the maternal allele being imprinted through a specific methylation of a CpG island; abnormally expressed in patients with Beckwith-Wiedemann syndrome, independently of IGF2 imprinting; no abnormal imprinting in Wilms tumour. Protein Expression: Untranslated. Function: Unknown; it is postulated that KCNQ1OT1 might influence the expression of nearby imprinted genes such as CDKN1C or IGF2/H19. Mutations Germinal: Aberrant methylation in 50-80% of BWS patients not always 100% (might be due to UPD in some cases); inheritance unclear. Somatic: Unclear; there is no association between aberrant methylation and tumour development. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Beckwith J. Extreme cytomegaly of the adrenal fetal cortex, omphalocele, hyperplasia of kidneys and pancreas, and Leydig-cell hyperplasia: Another syndrome? Western Society for Pediatric Research (abst) Los Angeles 1963 (Nov 11). WIEDEMANN HR. [FAMILIAL MALFORMATION COMPLEX WITH UMBILICAL HERNIA AND MACROGLOSSIA--A "NEW SYNDROME"?]. J Genet Hum. 1964 Sep;13:223-32 Beckwith J. Macroglossia, omphalocele, adrenal cytomegaly, gigantism, and hyperplastic visceromegaly. Birth Defects 1969;5:188-96. Elliott M, Bayly R, Cole T, Temple IK, Maher ER. Clinical features and natural history of Beckwith-Wiedemann syndrome: presentation of 74 new cases. Clin Genet. 1994 Aug;46(2):168-74 Hoovers JM, Kalikin LM, Johnson LA, Alders M, Redeker B, Law DJ, Bliek J, Steenman M, Benedict M, Wiegant J, Lengauer C, Taillon-Miller P, Schlessinger D, Edwards MC, Elledge SJ, Ivens A, Westerveld A, Little P, Mannens M, Feinberg AP. Multiple genetic loci within 11p15 defined by Beckwith-Wiedemann syndrome rearrangement breakpoints and subchromosomal transferable fragments. Proc Natl Acad Sci U S A. 1995 Dec 19;92(26):12456-60 Schneid H, Vazquez MP, Vacher C, Gourmelen M, Cabrol S, Le Bouc Y. The Beckwith-Wiedemann syndrome phenotype and the risk of cancer. Med Pediatr Oncol. 1997 Jun;28(6):4115 64 Beckwith-Wiedemann syndrome Mannens M DeBaun MR, Tucker MA. Risk of cancer during the first four years of life in children from The Beckwith-Wiedemann Syndrome Registry. J Pediatr. 1998 Mar;132(3 Pt 1):398-400 Alders M, Ryan A, Hodges M, Bliek J, Feinberg AP, Privitera O, Westerveld A, Little PF, Mannens M. Disruption of a novel imprinted zinc-finger gene, ZNF215, in Beckwith-Wiedemann syndrome. Am J Hum Genet. 2000 May;66(5):1473-84 Li M, Squire JA, Weksberg R. Molecular genetics of Wiedemann-Beckwith syndrome. Am J Med Genet. 1998 Oct 2;79(4):253-9 Steenman M, Westerveld A, Mannens M. Genetics of Beckwith-Wiedemann syndrome-associated tumors: common genetic pathways. Genes Chromosomes Cancer. 2000 May;28(1):1-13 Lee MP, DeBaun MR, Mitsuya K, Galonek HL, Brandenburg S, Oshimura M, Feinberg AP. Loss of imprinting of a paternally expressed transcript, with antisense orientation to KVLQT1, occurs frequently in Beckwith-Wiedemann syndrome and is independent of insulin-like growth factor II imprinting. Proc Natl Acad Sci U S A. 1999 Apr 27;96(9):5203-8 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Mannens M. Beckwith-Wiedemann syndrome. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):62-65. 65 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Cancer Prone Disease Section Mini Review Hereditary breast cancer Kaija Holli Department of Palliative Medicine, Department of Oncology, Tampere University Hospital, Tampere, Finland (KH) Published in Atlas Database: November 2000 Online updated version : http://AtlasGeneticsOncology.org/Kprones/HeredBreastCanID10062.html DOI: 10.4267/2042/37715 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology male breast cancer (6 %) and ovarian breast cancer (10% -20%); increased risk of prostate, laryngeal cancer and pancreatic cancer. Other genetic conditions associated with increased breast cancer risk are: Li-Fraumeni syndroma (p53 mutation on chromosome 17p13) is characterized by very early onset of neoplasms, including soft tissue sarcoma, osteosarcoma, brain tumours, leukemia, lung cancer, laryngeal cancer and adrenocorteal cancer.; lifetime risk for cancer: about 90 % for women and 70 % for men. Cowden syndroma (PTEN, 10q23) is a rare type of autosomal dominant inherited condition of multiple hamartomas with increased risk of bilateral breast cancers and thyroid tumours. Muir-Torre Syndroma (MSH2, MLH1), with also cancers of the gastro intestinal (GI)-tract, skin, genito urinary (Gu)-system. Peutz-Jeghers Syndroma, with also abnormal melarin deposits, GI-polyposis, cancers of the GI-tract, uterus, ovary and testis. Ataxia-teleangiectasia (linked to chromosome 11q21) autosomal recessive disorder with many clinical signs including increased risk for breast cancer; homozygous AT carriers have around 100-fold risk of cancer. One third of familiar breast cancers with hereditary background is still unknown; recent findings indicated that genotyping "BRCA3" locus at 13q and "BRCA2" locus at 2q may lead identifing the next mutations. Breast cancers of BRCA1 and, to lesser extent BRCA2 carriers differ from those of sporadic breast cancers: more high-grade tumours, pleomorphism, a higher mitotic count, less tubule formation, more often steroid receptor negative, DNA-aneuploid and more often higher s-phase fractions. Identity Alias: Site-specific breast cancer; Familiar breastovarian cancer Note: Hereditary or familiar form of breast cancerwith a familiar background. Inheritance Follows an autosomal dominant pattern. 5-10 % of all breast cancers have hereditary background. Hereditary susceptibility for breast cancer has been counted to be 30-40 % of BRCA1 (see below, gene section), 10-30 % of BRCA2, less than 1 % of Tp 53, less than 1 % of PTEN and one third of unknown mutations; frequency of BRCA1 mutation is around 0.2 % in general population, 200 carriers among 100 000 individuals. Clinics Note Hereditary breast cancer is a heterogenous entity including several clinical variants. Phenotype and clinics "Site specific breast cancer" is characterized by the predominance of breast cancer, while "hereditary breast-ovarian cancer" has neoplasms in both organs. BRCA1 mutation carrier (chromosome 17q12-21) has early age at onset, and lifetime risk for breast cancer 50 % - 85 % and ovarian cancer 15 %-45 %; about 500 different mutations have been reported; possible increased risk of prostate cancer and colon cancer. BRCA2 (chromosome 13q12-13) mutation carrier has risk for breast cancer 30 %-85 %; about 300 different mutations have been reported; it is also associated for Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 66 Hereditary breast cancer Holli K Treatment PTEN Prophylactic bilateral mastectomy (reduces the risk of about 90 %) and/or ovarian ablation. Chemoprevention (antiestrogens, aromataze inhibitors, retinoids) mainly in clinical trials (tamoxifen may reduce the risk about 45 %). Early detection of cancers by screening mammograms (ultrasound) yearly, palpation, transvaginal ultrasound. Location: 10q23 DNA/RNA Description: 9 exons. Protein Description: the PTEN protein (also called MMA1) is an evolutionary conserved dual-extensive similarity with the cyto-skeletal protein tensin Function: tumour suppression since bi allealic inactivations, inactivating germline mutations are responsible for a cancer prone síndrome. Mutations Germinal: heterozygeous germline mutations are responsible for the Cowden disease. Prognosis Prognosis is more dependent on extent of the disease at diagnosis than on the hereditary susceptibility. Genes involved and proteins NOTE: see also breast cancer. LKB1 BRCA1 Location: 19p13 DNA/RNA Description: 10 exons spanning 23 kb. Protein Description: 433 amino acids. Expression: wide. Function: serine/threonine kinase; tumor suppressor gene. Homology: Heterozygous mutations are responsible for the Peutz-Jeghers síndrome. Location: 17q21 DNA/RNA Description: 22 coding exons spanning over 70 kb of genomic DNA the BRCA1 mRNA has a size of 7.8 kb. Protein Description: the corresponding protein has 1863 amino acids, and 190-220 kDa. Expression: wide. Function: involved in DNA replication, repair transcriptional activation, cell cycle progression. Mutations Germinal: more than 500 sequence variations of the germline level have been reported. ATM Location: 11q22-23 DNA/RNA Description: 66 exons spanning 184 kb. Protein Description: 3056 amino acids, 350 kDa. Function: at the cell cycle checkpoint; induces G1 phase arrest. BRCA2 Location: 13q12-13 DNA/RNA Description: gene spanning more than 17 kb of genomic DNA; the coding sequence comprisons 26 exons (10 254 nucleotides). Protein Description: the corresponding protein has 3 418 amino acid residives (384 kDa). Mutations Germinal: more than 300 unique germ-line mutations have been reported. References Eng C, Murday V, Seal S, Mohammed S, Hodgson SV, Chaudary MA, Fentiman IS, Ponder BA, Eeles RA. Cowden syndrome and Lhermitte-Duclos disease in a family: a single genetic syndrome with pleiotropy? J Med Genet. 1994 Jun;31(6):458-61 Easton DF, Ford D, Bishop DT. Breast and ovarian cancer incidence in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Am J Hum Genet. 1995 Jan;56(1):265-71 P53 Location: 17p13 DNA/RNA Description: 11 exons. Protein Function: gene p53 encodes an ubiquitous nuclear protein involved in the control of genome integrity by preventing cells dividing before DNA damage is repaired. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Eeles R, Cole T, Taylor R, Lunt P, Baum M. Prophylactic mastectomy for genetic predisposition to breast cancer: the proband's story. Clin Oncol (R Coll Radiol). 1996;8(4):222-5 Knudson AG. Hereditary cancer: two hits revisited. J Cancer Res Clin Oncol. 1996;122(3):135-40 Struewing JP, Hartge P, Wacholder S, Baker SM, Berlin M, McAdams M, Timmerman MM, Brody LC, Tucker MA. The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med. 1997 May 15;336(20):1401-8 67 Hereditary breast cancer Holli K Fisher B, Costantino JP, Wickerham DL, Redmond CK, Kavanah M, Cronin WM, Vogel V, Robidoux A, Dimitrov N, Atkins J, Daly M, Wieand S, Tan-Chiu E, Ford L, Wolmark N. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst. 1998 Sep 16;90(18):1371-88 BRCA1 and BRCA2 mutations. J Natl Cancer Inst. 1998 Aug 5;90(15):1138-45 Thorlacius S, Struewing JP, Hartge P, Olafsdottir GH, Sigvaldason H, Tryggvadottir L, Wacholder S, Tulinius H, Eyfjörd JE. Population-based study of risk of breast cancer in carriers of BRCA2 mutation. Lancet. 1998 Oct 24;352(9137):1337-9 Fodor FH, Weston A, Bleiweiss IJ, McCurdy LD, Walsh MM, Tartter PI, Brower ST, Eng CM. Frequency and carrier risk associated with common BRCA1 and BRCA2 mutations in Ashkenazi Jewish breast cancer patients. Am J Hum Genet. 1998 Jul;63(1):45-51 Peto J, Collins N, Barfoot R, Seal S, Warren W, Rahman N, Easton DF, Evans C, Deacon J, Stratton MR. Prevalence of BRCA1 and BRCA2 gene mutations in patients with earlyonset breast cancer. J Natl Cancer Inst. 1999 Jun 2;91(11):943-9 Ford D, Easton DF, Stratton M, Narod S, Goldgar D, Devilee P, Bishop DT, Weber B, Lenoir G, Chang-Claude J, Sobol H, Teare MD, Struewing J, Arason A, Scherneck S, Peto J, Rebbeck TR, Tonin P, Neuhausen S, Barkardottir R, Eyfjord J, Lynch H, Ponder BA, Gayther SA, Zelada-Hedman M. Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Consortium. Am J Hum Genet. 1998 Mar;62(3):676-89 Kainu T, Juo SH, Desper R, Schaffer AA, Gillanders E, Rozenblum E, Freas-Lutz D, Weaver D, Stephan D, BaileyWilson J, Kallioniemi OP, Tirkkonen M, Syrjäkoski K, Kuukasjärvi T, Koivisto P, Karhu R, Holli K, Arason A, Johannesdottir G, Bergthorsson JT, Johannsdottir H, Egilsson V, Barkardottir RB, Johannsson O, Haraldsson K, Sandberg T, Holmberg E, Grönberg H, Olsson H, Borg A, Vehmanen P, Eerola H, Heikkila P, Pyrhönen S, Nevanlinna H. Somatic deletions in hereditary breast cancers implicate 13q21 as a putative novel breast cancer susceptibility locus. Proc Natl Acad Sci U S A. 2000 Aug 15;97(17):9603-8 Lakhani SR, Jacquemier J, Sloane JP, Gusterson BA, Anderson TJ, van de Vijver MJ, Farid LM, Venter D, Antoniou A, Storfer-Isser A, Smyth E, Steel CM, Haites N, Scott RJ, Goldgar D, Neuhausen S, Daly PA, Ormiston W, McManus R, Scherneck S, Ponder BA, Ford D, Peto J, Stoppa-Lyonnet D, Bignon YJ, Struewing JP, Spurr NK, Bishop DT, Klijn JG, Devilee P, Cornelisse CJ, Lasset C, Lenoir G, Barkardottir RB, Egilsson V, Hamann U, Chang-Claude J, Sobol H, Weber B, Stratton MR, Easton DF. Multifactorial analysis of differences between sporadic breast cancers and cancers involving Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Holli K. Hereditary breast cancer. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):66-68. 68 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Cancer Prone Disease Section Mini Review Variegated aneuploidy related centromere division (PCD) to premature Alberto Plaja Unitat de Genètica, Hospital Materno- Infantil Vall d'Hebron Pg. Vall d'Hebron 119- 129, 08035- Barcelona, Spain (AP) Published in Atlas Database: November 2000 Online updated version : http://AtlasGeneticsOncology.org/Kprones/VariegAneuplPCDID10069.html DOI: 10.4267/2042/37716 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology found in other patients with an expected increased cellular mortality (variegated aneuploidy without PCD and the "ring syndrome"). Identity Alias: (Variegated aneuploidy or mosaic aneuploidy) related to (PCD, C-anaphases,premature anaphase, premature chromatid separation or asynchrony of mitotic stages) Note The term premature centromere division has also been utilized to describe an unrelated cytogenetic phenomenon, the age related loss of centromeric function in chromosome X. Variegated aneuploidy has also been described in patients without PCD and patients with Roberts syndrome. Inheritance Only 11 patients known. Premature centromere division (PCD) without variegated aneuploidy has been shown to have an autosomal dominant inheritance, with an estimated frequency of 0.1% of the population. It has been proposed that patients with variegated aneuploidy related to PCD are homozygotes for this trait, but in several cases one of the parents do not show elevated frequency of PCD. A recessive inheritance with hormonal factors modifying the expression of PCD in a carrier, isodisomy of one chromosome or loss of heterozygosity has been suggested. Phenotype and clinics the clinical phenotype of the 11 patients described in the literature includes microcephaly (11/11), central nervous system (CNS) anomalies (5/6) with cerebellar defects and migration defects, mental retardation (8/9), prenatal (always noted over 23 weeks of gestation) and postnatal growth retardation (10/10), flat and broad nasal bridge (4/7), apparently low-set ears (5/8), eye abnormalities (8/10), skin abnormalities (3/9) and ambiguous genitalia in male patients (4/6); seizures have been reported in 5 patients; cancer is a major concern in the clinical management of these patients (5/11); birth weight corrected for gestational age ranges from -1.3 to -4.1 SD, birth length from -0.8 to -5.4 SD and OFC from -2.6 to -5.8. Neoplastic risk The occurrence of Wilms tumor in three patients, rhabdomyosarcoma in two others and acute leukemia in a fifth characterizes this condition as a chromosome instability disorder with a high risk of malignancy; interestingly enough, preferential loss of maternal 11p15.5 chromosome region has been repeatedly reported in Wilms tumor as well as in rhabdomyosarcoma. Prognosis Clinics Although published data is incomplete, at least 4 patients have died before 2 years of age, a fifth deceased at 42 years and one patient aged 18 month has an advanced, relapsed rhabdomyosarcoma; patient's death had been related to pneumonia (one patient), leukemia (one patient), and Wilms tumor (three cases). Note Patients show a remarkably constant clinical phenotype probably due to high cellular mortality induced by the aneuploidies; similar clinical findings have also been Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 69 Variegated aneuploidy related to premature centromere division (PCD) Plaja A mosaic aneuploidies, microcephaly, mental retardation and a variety of malformations; in these patients, trisomy is by far more frequent than monosomy; trisomies of chromosome 8, 18 and X predominate in lymphocyte cultures and trisomy 2, 7, 12 and 20 predominate in fibroblasts; usually, at least one of the parents shows an elevated PCD frequency (range 12.642.5) but not variegated aneuploidy. High levels of PCD have been reported in skin fibroblasts (althought in at least one case no aneupoidies where found), hair-root, bone marrow and trophoblastic cells of chorionic villi; there is no data of PCD expression in amniocytes but pseudomosaicism of chromosome 7 and 21 has been reported in amniocytes; cord blood chromosome analysis in one case showed PCD and variegated aneuploidies. Cytogenetics Inborn conditions The terms premature centromere division (PCD), Canaphases, premature anaphase, premature chromatid separation and asynchrony of mitotic stages describe cells in division which have overcome a colchicineinduced metaphase block; the resulting mitotic configuration shows split centromeres and splayed chromatids in all or most of the chromosomes. Control individuals generally show low frequencies of PCD (up to 3% of the mitoses), which seems to have no pathological relevance, but in 0.1% of the population an elevated PCD frequency (>5%) is found in colchicine exposed lymphocyte cultures; this type of PCD shows autosomal dominant inheritance and has traditionally considered to be harmless with the possible exception of some patients with subfertility or repeated abortion. In few patients high levels of PCD (25- 87%) are found in combination with an increased number of cells with Cytogenetics of cancer Cytogenetic analysis of one embryonal rhabdosarcoma showed normal karyotype in cultured cells and extensive aneuploidy with some estructural aberrations in the only two cells obtained from direct harvest. cell showing premature centromere division (PCD) phenomenon, with split centromeres and splayed chromatids in all the chromosomes Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 70 Variegated aneuploidy related to premature centromere division (PCD) Plaja A References premature chromatid separation trait. Am J Med Genet. 1998 Jul 7;78(3):245-9 Gabarrón J, Jimenez A, Glover G. Premature centromere division dominantly inherited in a subfertile family. Cytogenet Cell Genet. 1986;43(1-2):69-71 Kawame H, Sugio Y, Fuyama Y, Hayashi Y, Suzuki H, Kurosawa K, Maekawa K. Syndrome of microcephaly, DandyWalker malformation, and Wilms tumor caused by mosaic variegated aneuploidy with premature centromere division (PCD): report of a new case and review of the literature. J Hum Genet. 1999;44(4):219-24 Scheres JM JC, Hustinx TWJ, Madam K, Beltman J D, Lindhout D. A mitotic mutant causing non- disjunction in man. In: 7h International Congress of Human Genetics. Berlin. 1986;Abst p163. Limwongse C, Schwartz S, Bocian M, Robin NH. Child with mosaic variegated aneuploidy and embryonal rhabdomyosarcoma. Am J Med Genet. 1999 Jan 1;82(1):20-4 Kosztolányi G. Does "ring syndrome" exist? An analysis of 207 case reports on patients with a ring autosome. Hum Genet. 1987 Feb;75(2):174-9 D'Agostino A, Calzone R, Poggi V, Zatterale A, Nitsch L. Mosaic variegated aneuploidies in a newborn with growth retardation and dismorphic phenotype. In: European Human Genetics Conference, Amsterdam, 2000; Abst p80. Madan K, Lindhout D, Palan A. Premature centromere division (PCD): a dominantly inherited cytogenetic anomaly. Hum Genet. 1987 Oct;77(2):193-6 Matsuura S, Ito E, Tauchi H, Komatsu K, Ikeuchi T, Kajii T. Chromosomal instability syndrome of total premature chromatid separation with mosaic variegated aneuploidy is defective in mitotic-spindle checkpoint. Am J Hum Genet. 2000 Aug;67(2):483-6 Chamla Y. C-anaphases in lymphocyte cultures versus premature centromere division syndromes. Hum Genet. 1988 Feb;78(2):111-4 Miller K, Müller W, Winkler L, Hadam MR, Ehrich JH, Flatz SD. Mitotic disturbance associated with mosaic aneuploidies. Hum Genet. 1990 Mar;84(4):361-4 Plaja A, Vendrell T, Smeets D, Sarret E, Gili T, Català V, Mediano C, Scheres JM. Variegated aneuploidy related to premature centromere division (PCD) is expressed in vivo and is a cancer-prone disease. Am J Med Genet. 2001 Jan 22;98(3):216-23 Warburton D, Anyane-Yeboa K, Taterka P, Yu CY, Olsen D. Mosaic variegated aneuploidy with microcephaly: a new human mitotic mutant? Ann Genet. 1991;34(3-4):287-92 Domínguez MG, Rivera H. C-anaphases: a mitotic variant. Ann Genet. 1992;35(3):183-5 This article should be referenced as such: Plaja A. Variegated aneuploidy related to premature centromere division (PCD). Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):69-71. Kajii T, Kawai T, Takumi T, Misu H, Mabuchi O, Takahashi Y, Tachino M, Nihei F, Ikeuchi T. Mosaic variegated aneuploidy with multiple congenital abnormalities: homozygosity for total Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 71 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Cancer Prone Disease Section Review Hereditary pancreatic cancer Ralph H Hruban, Scott E Kern The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA (RHH, SEK) Published in Atlas Database: December 2000 Online updated version : http://AtlasGeneticsOncology.org/Kprones/HeredPancrCanID10068.html DOI: 10.4267/2042/37717 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology have a 56-fold increased risk of developing pancreatic cancer. Each of the five clinically recognized syndromes associated with the familial aggregation of pancreatic cancer has its own unique clinical findings. Second breast cancer syndrome: the BRCA2 tumor suppressor gene is located on chromosome 13q and carriers of germline BRCA2 mutations have a significant lifetime risk of developing breast cancer (30-85%) at a young age; they are also at risk for bilateral breast cancer; BRCA2 is also associated with an increased risk of male breast cancer, ovarian cancer, prostate cancer and pancreatic cancer; the lifetime risk of pancreatic cancer in carriers of germline BRCA2 mutations is approximately 10%; germline BRCA2 mutations are particularly common amongst individuals of Ashkenazi Jewish heritage because of a founder effect. Familial atypical multiple mole melanoma (FAMMM) syndrome has an autosomal dominant mode of transmission; most cases are caused by germline mutations in the p16 tumor suppressor gene on chromosome 9p; individuals affected with FAMMM develop multiple melanocytic nevi, some of which can be atypical; they also are at increased risk of developing melanoma and pancreatic cancer; the lifetime risk of pancreatic cancer in individuals with germline p16 mutations is about 20%. The Peutz-Jeghers Syndrome is inhertied in an autosomal dominant mode; it has recently been shown to be caused by germline mutaitons in the STK11/LKB1 gene on chromosome 19p; individuals with this syndrome typically develop multiple mucocutaneus melanin macules, harmartomatous gastrointestinal polyps and they have an increased risk of developing cancers of the gastrointestinal tract; it has been estimated that the lifetime risk of pancreatic cancer in patient with the Peutz-Jeghers Syndrome is approximately 30%. Identity Alias: Familial pancreatic cancer Inheritance It has been estimated that as many as 10% of pancreatic cancers have a hereditary basis; five genetic syndromes have been identified that are associated with the familial aggregation of pancreatic cancer; these include: The second breast cancer syndrome (BRCA2), the familial atypical multiple mole melanoma (FAMMM), the Peutz-Jeghers Syndrome, the hereditary pancreatitis and the hereditary non-polyposis colorectal cancer (HNPCC) syndrome. Most kindreds with familial pancreatic cancer, however, do not fall into one of these well-defined syndromes and these are referred to simply as "family pancreatic cancer." Clinics Note a generally accepted definition of familial pancreatic cancer is a kindred in which at least a pair of firstdegree relatives (sibling-sibling or parent-child) have been diagnosed with pancreatic cancer; several large registries have been established to define the patterns of inheritance and genetic basis for the familial aggregation of pancreatic cancer in these kindreds, the National Pancreas Tumor Registry (NFPTR) is the largest such registry; over 260 familial pancreatic cancer kindreds have enrolled in this registry and studies of these kindreds has revealed that when followed prospectively, apparently healthy, first-degree relatives of patients with familial pancreatic cancer have an 18-fold increased risk of developing pancreatic cancer; when there are three or more family members with pancreatic cancer in a kindred, the first-degree relatives of the index patient with pancreatic cancer Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 72 Hereditary pancreatic cancer Hruban RH, Kern SE Hereditary pancreatitis has an autosomal dominant mode of transmission; it is caused by germline mutations in the cationic trypsinogen gene (called PRSS1) on chromosome 7q35; affected individuals develop recurrent episodes of pancreatitis at a young age and they have an elevated lifetime risk of developing pancreatic cancers that approaches 40%. The hereditary nonpolyposis colorectal cancer (HNPCC) syndrome is caused by germiline mutations in one of the DNA mismatch repair genes (such as hMLH1 on chromosome 3 p and hMSH2 on chromosome 2p); in addition to colorectal neoplasia, affected family members have an increased risk of developing pancreatic cancer; the pancreatic cancers that arise in patients with HNPCC often have a distinct histologic appearance referred to as "medullary" histology. The ataxia-telangectasia and familial adenomatous polyposis syndromes have also been associated with an increased risk of developing pancreatic cancer, however, these associations are not well-established. DNA/RNA Description: the coding sequence comprises 3 exons: this locus gives rise to 2 distinct transcripts from different promoters (p16 and p16(ARF)). Protein Description: the corresponding protein, called cyclindependent kinase inhibitor-2A, has 156 amino acid residues. Function: cyclin-dependent kinase inhibitor 2A binds to CDK4 and inhibits the ability of CDK4 to interact with cyclinA thereby inducing a G1 cell cycle arrest. Mutations Germinal: germline mutations are associated with the FAMMM Syndrome. Somatic: virtually all invasive pancreatic carcinomas show inactivation of the p16 gene; forty percent by homozygous deletion, 40% by an intragenic mutation coupled with loss of heterozygocity (LOH) and 15% by hypermethylation of the p16 promoter. STK11 Treatment Location: 19p13.3 DNA/RNA Description: gene Spanning 23kb of genomic DNA, the coding sequence comprises 9 exons (1446bp). Protein Description: the corresponding protein has 433 amino acid residues. Function: serine throeonine protein kinase 11. Mutations Germinal: almost all germline mutations are predicted to disrupt the function of the kinase domain. Somatic: approximately 4% of sporadic pancreatic cancers have somatic inactivation of STK11. Currently, there are no effective methods to screen individuals at-risk for early pancreatic cancer; several studies are underway to examine the effectiveness of endoscopic ultrasound (EUS) in the early detection of pancreatic cancer. Prognosis Prognosis will depend on the stage of the disease at diagnosis more than it does on hereditary sysceptibility. Genes involved and proteins BRCA2 Location: 13q12 3 DNA/RNA Description: gene spanning more than 70kb of genomic DNA; the coding sequence comprises 27 exons (11 395 nucleotides). Protein Description: the corresponding protein has 3 418 amino acid residues (384 kDa). Function: the Brca2 protein binds to Rad51 and serves as an important co-factor in the Rad51 -dependent DNA repair of double strand breaks; the Brca2 protein may also have transcription activation potential. Mutations Germinal: more than 300 unique germ-line mutations have been reported; the 6174 delT mutation is particularly common in Jewish subjects Somatic: acquired mutations in BRCA2 rare in pancreatic cancer. PRSS1 Location: 7q35 DNA/RNA Description: the coding sequence comprise 5 exons (800bp). Protein Description: trypsin, which is active in the pacreas, in inactivated by cleavage; mutations which abrogate this cleavage site can result in autodigestion and pancreatitis. Mutations Germinal: the arg117-to-his mutation (R117H) is the most common mutation identified to date. hMLH1 Location: 3p21.3 DNA/RNA Description: the coding sequence comprises 2484b. p16 Location: 9p21 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 73 Hereditary pancreatic cancer Hruban RH, Kern SE ST, Toskes PP, Liddle R, McGrath K, Uomo G, Post JC, Ehrlich GD. Hereditary pancreatitis is caused by a mutation in the cationic trypsinogen gene. Nat Genet. 1996 Oct;14(2):1415 Protein Description: MLH1 forms a complex with other DNA mismatch repair gene; functions in DNA mismatch repairs2. Mutations Germinal: one of at least 5 known human mismatch repair genes associated with the hereditary nonpolyposis colorectal cancer syndrome: the neoplasms that develop in these patients typically show microsatellite instability. Whitcomb DC, Preston RA, Aston CE, Sossenheimer MJ, Barua PS, Zhang Y, Wong-Chong A, White GJ, Wood PG, Gates LK Jr, Ulrich C, Martin SP, Post JC, Ehrlich GD. A gene for hereditary pancreatitis maps to chromosome 7q35. Gastroenterology. 1996 Jun;110(6):1975-80 Lowenfels AB, Maisonneuve P, DiMagno EP, Elitsur Y, Gates LK Jr, Perrault J, Whitcomb DC. Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. J Natl Cancer Inst. 1997 Mar 19;89(6):442-6 hMSH2 Location: 2p22-p21 DNA/RNA Description: the MSH2 locus covers approximately 73kb and contains 16 exons. Protein Description: MSH2 functions in DNA mismatch repair. Mutations Germinal: one of at least 5 known human mismatch repair genes associated with the hereditary nonpolyposis colorectal cancer syndrome; the neoplasms that develop in these patients typically show microsatellite instability. Ozçelik H, Schmocker B, Di Nicola N, Shi XH, Langer B, Moore M, Taylor BR, Narod SA, Darlington G, Andrulis IL, Gallinger S, Redston M. Germline BRCA2 6174delT mutations in Ashkenazi Jewish pancreatic cancer patients. Nat Genet. 1997 May;16(1):17-8 Abbott DW, Freeman ML, Holt JT. Double-strand break repair deficiency and radiation sensitivity in BRCA2 mutant cancer cells. J Natl Cancer Inst. 1998 Jul 1;90(13):978-85 Goggins M, Offerhaus GJ, Hilgers W, Griffin CA, Shekher M, Tang D, Sohn TA, Yeo CJ, Kern SE, Hruban RH. Pancreatic adenocarcinomas with DNA replication errors (RER+) are associated with wild-type K-ras and characteristic histopathology. Poor differentiation, a syncytial growth pattern, and pushing borders suggest RER+. Am J Pathol. 1998 Jun;152(6):1501-7 References Hemminki A, Markie D, Tomlinson I, Avizienyte E, Roth S, Loukola A, Bignell G, Warren W, Aminoff M, Höglund P, Järvinen H, Kristo P, Pelin K, Ridanpää M, Salovaara R, Toro T, Bodmer W, Olschwang S, Olsen AS, Stratton MR, de la Chapelle A, Aaltonen LA. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature. 1998 Jan 8;391(6663):184-7 JEGHERS H, McKUSICK VA, KATZ KH. Generalized intestinal polyposis and melanin spots of the oral mucosa, lips and digits; a syndrome of diagnostic significance. N Engl J Med. 1949 Dec 22;241(25):993, illust; passim Lynch HT, Voorhees GJ, Lanspa SJ, McGreevy PS, Lynch JF. Pancreatic carcinoma and hereditary nonpolyposis colorectal cancer: a family study. Br J Cancer. 1985 Aug;52(2):271-3 Hruban RH, Petersen GM, Ha PK, Kern SE. Genetics of pancreatic cancer. From genes to families. Surg Oncol Clin N Am. 1998 Jan;7(1):1-23 Giardiello FM, Welsh SB, Hamilton SR, Offerhaus GJ, Gittelsohn AM, Booker SV, Krush AJ, Yardley JH, Luk GD. Increased risk of cancer in the Peutz-Jeghers syndrome. N Engl J Med. 1987 Jun 11;316(24):1511-4 Jenne DE, Reimann H, Nezu J, Friedel W, Loff S, Jeschke R, Müller O, Back W, Zimmer M. Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet. 1998 Jan;18(1):38-43 Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, Weinstein CL, Hruban RH, Yeo CJ, Kern SE. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet. 1994 Sep;8(1):27-32 Mattson K. Docetaxel (Taxotere) in the neo-adjuvant setting in non-small-cell lung cancer. Ann Oncol. 1999;10 Suppl 5:S6972 Su GH, Hruban RH, Bansal RK, Bova GS, Tang DJ, Shekher MC, Westerman AM, Entius MM, Goggins M, Yeo CJ, Kern SE. Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. Am J Pathol. 1999 Jun;154(6):1835-40 Goldstein AM, Fraser MC, Struewing JP, Hussussian CJ, Ranade K, Zametkin DP, Fontaine LS, Organic SM, Dracopoli NC, Clark WH Jr. Increased risk of pancreatic cancer in melanoma-prone kindreds with p16INK4 mutations. N Engl J Med. 1995 Oct 12;333(15):970-4 Tascilar M, Tersmette AC, Offerhaus GJ, Hruban RH. Pancreatic cancer--more familial than you thought. Anal Cell Pathol. 1999;19(3-4):105-10 Berman DB, Costalas J, Schultz DC, Grana G, Daly M, Godwin AK. A common mutation in BRCA2 that predisposes to a variety of cancers is found in both Jewish Ashkenazi and nonJewish individuals. Cancer Res. 1996 Aug 1;56(15):3409-14 Giardiello FM, Brensinger JD, Tersmette AC, Goodman SN, Petersen GM, Booker SV, Cruz-Correa M, Offerhaus JA. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology. 2000 Dec;119(6):1447-53 Goggins M, Schutte M, Lu J, Moskaluk CA, Weinstein CL, Petersen GM, Yeo CJ, Jackson CE, Lynch HT, Hruban RH, Kern SE. Germline BRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas. Cancer Res. 1996 Dec 1;56(23):5360-4 Lal G, Liu G, Schmocker B, Kaurah P, Ozcelik H, Narod SA, Redston M, Gallinger S. Inherited predisposition to pancreatic adenocarcinoma: role of family history and germ-line p16, BRCA1, and BRCA2 mutations. Cancer Res. 2000 Jan 15;60(2):409-16 Whitcomb DC, Gorry MC, Preston RA, Furey W, Sossenheimer MJ, Ulrich CD, Martin SP, Gates LK Jr, Amann Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 74 Hereditary pancreatic cancer Hruban RH, Kern SE Lynch HT, Brand RE, Lynch JF, Fusaro RM, Smyrk TC, Goggins M, Kern SE. Genetic counseling and testing for germline p16 mutations in two pancreatic cancer-prone families. Gastroenterology. 2000 Dec;119(6):1756-60 Tersmette AC, Petersen GM, Offerhaus GJ, Falatko FC, Brune KA, Goggins M, Rozenblum E, Wilentz RE, Yeo CJ, Cameron JL, Kern SE, Hruban RH. Increased risk of incident pancreatic cancer among first-degree relatives of patients with familial pancreatic cancer. Clin Cancer Res. 2001 Mar;7(3):738-44 Wilentz RE, Goggins M, Redston M, Marcus VA, Adsay NV, Sohn TA, Kadkol SS, Yeo CJ, Choti M, Zahurak M, Johnson K, Tascilar M, Offerhaus GJ, Hruban RH, Kern SE. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: A newly described and characterized entity. Am J Pathol. 2000 May;156(5):1641-51 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) This article should be referenced as such: Hruban RH, Kern SE. Hereditary pancreatic cancer. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):72-75. 75 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Cancer Prone Disease Section Mini Review Li-Fraumeni syndrome Jenny M Varley Cancer Genetics Group, Paterson Institute for Cancer Research, Wilmslow Road, Manchester M20 9BX, UK (JMV) Published in Atlas Database: December 2000 Online updated version : http://AtlasGeneticsOncology.org/Kprones/LiFraumeniID10011.html DOI: 10.4267/2042/37718 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology DNA/RNA Description: 11 exons, the first of which is non-coding. Protein Description: p53, a 393 amino acid protein. Function: p53 is the most commonly mutated gene in human cancers possessing multiple properties; p53 has two major roles. Firstly in cell cycle arrest, predominantly in the G1 phase of the cell cycle, but also with a role in G2 and mitotic checkpoints. Secondly the induction of apoptosis (programmed cell death). Both these are induced upon DNA damage, and the response depends on many things including the type of damage and the cell type. p53 is a transcription factor with a central sequencespecific DNA binding domain and a N-terminal transactivation domain; upon DNA damage, the level of p53 increases markedly, and the DNA-binding properties are activated; the levels of p53 are regulated primarily post-transcriptionally (including phosphorylation and acetylation). Mutations Germinal: there are over 200 published reports of germline mutations. Over 75% of families with classic LFS have a germline TP53 mutation. Lower proportions of families with some features of LFS have such mutations. Children with adrenocortical carcinoma have an extremely high incidence of germline mutations (over 80%). The spectrum of mutations in the germline is superficially the same as somatic mutations, but there are some significant differences. Identity Note Families with Li-Fraumeni syndrome (LFS) are defined by: a proband with a sarcoma aged under 45 years, with a first degree relative with cancer under 45 years and another first or second degree relative with any cancer under 45 years or a sarcoma at any age. Inheritance Autosomal dominant, high penetrance (100% lifetime risk in females, 75% in males). Clinics Phenotype and clinics No associated dysmorphologies or abnormalities. Neoplastic risk Very high. The main neoplastic risks are bone, cartilage and soft tissue sarcomas, early-onset female breast cancer, brain and spinal cord tumours, childhood adrenocortical tumours, Wilms' tumour and malignant phyllodes tumours. There is no increased incidence of a number of cancers which occur frequently within the population, such as colorectal, lung, bladder and gynaecological malignancies. Some other tumour types occur rarely, but more frequently than expected; these include pancreas, peripheral nervous system, leukaemia and stomach. Genes involved and proteins TP53 Location: 17p13 Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) 76 Li-Fraumeni syndrome Varley JM 21 Li-Fraumeni families. Cancer Res. 1994 Mar 1;54(5):1298304 hCHK2 Location: 22q12.1 DNA/RNA Description: 14 exons. Protein Description: a 543 amino acid protein with homology. To Saccharomyces cerevisiae RAD53 andSchizosaccharomyces pombe cds1. Function: a protein kinase which is required for DNA damage and replication checkpoints; CHK2 is phosphorylated by ATM, and in turn can phosphorylate p53 at serine-20; it appears that germline hCHK2 mutations are uncommon in LFS. Varley JM, Evans DG, Birch JM. Li-Fraumeni syndrome--a molecular and clinical review. Br J Cancer. 1997;76(1):1-14 Varley JM, McGown G, Thorncroft M, Santibanez-Koref MF, Kelsey AM, Tricker KJ, Evans DG, Birch JM. Germ-line mutations of TP53 in Li-Fraumeni families: an extended study of 39 families. Cancer Res. 1997 Aug 1;57(15):3245-52 Birch JM, Blair V, Kelsey AM, Evans DG, Harris M, Tricker KJ, Varley JM. Cancer phenotype correlates with constitutional TP53 genotype in families with the Li-Fraumeni syndrome. Oncogene. 1998 Sep 3;17(9):1061-8 Bell DW, Varley JM, Szydlo TE, Kang DH, Wahrer DC, Shannon KE, Lubratovich M, Verselis SJ, Isselbacher KJ, Fraumeni JF, Birch JM, Li FP, Garber JE, Haber DA. Heterozygous germ line hCHK2 mutations in Li-Fraumeni syndrome. Science. 1999 Dec 24;286(5449):2528-31 References Varley JM, McGown G, Thorncroft M, James LA, Margison GP, Forster G, Evans DG, Harris M, Kelsey AM, Birch JM. Are there low-penetrance TP53 Alleles? evidence from childhood adrenocortical tumors. Am J Hum Genet. 1999 Oct;65(4):9951006 Li FP, Fraumeni JF Jr. Soft-tissue sarcomas, breast cancer, and other neoplasms. A familial syndrome? Ann Intern Med. 1969 Oct;71(4):747-52 Li FP, Fraumeni JF Jr, Mulvihill JJ, Blattner WA, Dreyfus MG, Tucker MA, Miller RW. A cancer family syndrome in twentyfour kindreds. Cancer Res. 1988 Sep 15;48(18):5358-62 Chompret A, Brugières L, Ronsin M, Gardes M, DessarpsFreichey F, Abel A, Hua D, Ligot L, Dondon MG, Bressac-de Paillerets B, Frébourg T, Lemerle J, Bonaïti-Pellié C, Feunteun J. P53 germline mutations in childhood cancers and cancer risk for carrier individuals. Br J Cancer. 2000 Jun;82(12):19327 Malkin D, Li FP, Strong LC, Fraumeni JF Jr, Nelson CE, Kim DH, Kassel J, Gryka MA, Bischoff FZ, Tainsky MA. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science. 1990 Nov 30;250(4985):1233-8 This article should be referenced as such: Birch JM, Hartley AL, Tricker KJ, Prosser J, Condie A, Kelsey AM, Harris M, Jones PH, Binchy A, Crowther D. Prevalence and diversity of constitutional mutations in the p53 gene among Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1) Varley JM. Li-Fraumeni syndrome. Atlas Genet Cytogenet Oncol Haematol. 2001; 5(1):76-77. 77 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 Editorial@AtlasGeneticsOncology.org. Rules, Copyright Notice and Disclaimer Conflicts of Interest: Authors must state explicitly whether potential conflicts do or do not exist. Reviewers must disclose to editors any conflicts of interest that could bias their opinions of the manuscript. The editor and the editorial board members must disclose any potential conflict. Privacy and Confidentiality – Iconography: Patients have a right to privacy. Identifying details should be omitted. If complete anonymity is difficult to achieve, informed consent should be obtained. Property: As "cards" are to evolve with further improvements and updates from various contributors, the property of the cards belongs to the editor, and modifications will be made without authorization from the previous contributor (who may, nonetheless, be asked for refereeing); contributors are listed in an edit history manner. Authors keep the rights to use further the content of their papers published in the Atlas, provided that the source is cited. Copyright: The information in the Atlas of Genetics and Cytogenetics in Oncology and Haematology is issued for general distribution. All rights are reserved. The information presented is protected under international conventions and under national laws on copyright and neighbouring rights. Commercial use is totally forbidden. Information extracted from the Atlas may be reviewed, reproduced or translated for research or private study but not for sale or for use in conjunction with commercial purposes. Any use of information from the Atlas should be accompanied by an acknowledgment of the Atlas as the source, citing the uniform resource locator (URL) of the article and/or the article reference, according to the Vancouver convention. Reference to any specific commercial products, process, or service by trade name, trademark, manufacturer, or otherwise, does not necessarily constitute or imply its endorsement, recommendation, or favouring. The views and opinions of contributors and authors expressed herein do not necessarily state or reflect those of the Atlas editorial staff or of the web site holder, and shall not be used for advertising or product endorsement purposes. The Atlas does not make any warranty, express or implied, including the warranties of merchantability and fitness for a particular purpose, or assumes any legal liability or responsibility for the accuracy, completeness, or usefulness of any information, and shall not be liable whatsoever for any damages incurred as a result of its use. In particular, information presented in the Atlas is only for research purpose, and shall not be used for diagnosis or treatment purposes. No responsibility is assumed for any injury and/or damage to persons or property for any use or operation of any methods products, instructions or ideas contained in the material herein. See also: "Uniform Requirements for Manuscripts Submitted to Biomedical Journals: Writing and Editing for Biomedical Publication - Updated October 2004": http://www.icmje.org. http://AtlasGeneticsOncology.org © ATLAS - ISSN 1768-3262 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS