vol 21 12 2017 -...

56
Volume 1 - Number 1 May - September 1997 Volume 21 - Number 12 December 2017

Upload: others

Post on 12-Feb-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Volume 1 - Number 1 May - September 1997

Volume 21 - Number 12 December 2017

Page 2: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

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

INIST-CNRS

OPEN ACCESS JOURNAL

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 is made for and by: clinicians and researchers in cytogenetics, molecular biology, oncology, haematology, and pathology.

One main scope of the Atlas is to conjugate the scientific information provided by cytogenetics/molecular genetics to the

clinical setting (diagnostics, prognostics and therapeutic design), another is to provide an encyclopedic knowledge in cancer

genetics. The Atlas deals with cancer research and genomics. It is at the crossroads of research, virtual medical university

(university and post-university e-learning), and telemedicine. It contributes to "meta-medicine", this mediation, using

information technology, between the increasing amount of knowledge and the individual, having to use the information.

Towards a personalized medicine of cancer.

It presents structured review articles ("cards") on:

1- Genes,

2- Leukemias,

3- Solid tumors,

4- Cancer-prone diseases, and also

5- "Deep insights": more traditional review articles on the above subjects and on surrounding topics.

It also present

6- Case reports in hematology and

7- Educational items in the various related topics for students in Medicine and in Sciences.

The Atlas of Genetics and Cytogenetics in Oncology and Haematology does not publish research articles.

See also: http://documents.irevues.inist.fr/bitstream/handle/2042/56067/Scope.pdf

Editorial correspondance

Jean-Loup Huret, MD, PhD,

Genetics, Department of Medical Information,

University Hospital

F-86021 Poitiers, France

phone +33 5 49 44 45 46

[email protected]

Editor, Editorial Board and Publisher See:http://documents.irevues.inist.fr/bitstream/handle/2042/48485/Editor-editorial-board-and-publisher.pdf

The Atlas of Genetics and Cytogenetics in Oncology and Haematology is published 12 times a year by ARMGHM, a non

profit organisation, and by the INstitute for Scientific and Technical Information of

the French National Center for Scientific Research (INIST-CNRS) since 2008.

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

Staff: Vanessa Le Berre

Philippe Dessen is the Database Directorof the on-line version (Gustave Roussy Institute – Villejuif – France).

Publisher Contact: INIST-CNRS

Mailing Address: Catherine Morel, 2,Allée du Parc de Brabois, CS 10130, 54519 Vandoeuvre-lès-Nancy France.

Email Address:[email protected]

Articles of the ATLAS are free in PDF format, and metadata are available on the web in Dublin Core XML format and freely

harvestable.A Digital object identifier (DOI®), recorded at the International Agency CrossRefhttp://www.crossref.org/ is

assigned to each article.

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

Page 3: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12)

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS

OPEN ACCESS JOURNAL

Editor-in-Chief Jean-Loup Huret (Poitiers, France) Lymphomas Section Editor Antonino Carbone (Aviano, Italy)

Myeloid Malignancies Section Editor Robert S. Ohgami (Stanford, California)

Bone Tumors Section Editor Judith Bovee (Leiden, Netherlands)

Head and Neck Tumors Section Editor Cécile Badoual (Paris, France)

Urinary Tumors Section Editor Paola Dal Cin (Boston, Massachusetts)

Pediatric Tumors Section Editor Frederic G. Barr (Bethesda, Maryland)

Cancer Prone Diseases Section Editor Gaia Roversi (Milano, Italy)

Cell Cycle Section Editor João Agostinho Machado-Neto (São Paulo, Brazil)

DNA Repair Section Editor Godefridus Peters (Amsterdam, Netherlands)

Hormones and Growth factors Section Editor Gajanan V. Sherbet (Newcastle upon Tyne, UK)

Mitosis Section Editor Patrizia Lavia (Rome, Italy)

WNT pathway Section Editor Alessandro Beghini (Milano, Italy)

B-cell activation Section Editors Anette Gjörloff Wingren and Barnabas Nyesiga (Malmö,

Sweden)

Oxidative stress Section Editor Thierry Soussi (Stockholm, Sweden/Paris, France)

Board Members

Sreeparna Banerjee Department of Biological Sciences, Middle East Technical University, Ankara, Turkey; [email protected]

Alessandro

Beghini Department of Health Sciences, University of Milan, Italy; [email protected]

Judith Bovée 2300 RC Leiden, The Netherlands; [email protected]

Antonio Cuneo Dipartimento di ScienzeMediche, Sezione di Ematologia e Reumatologia Via Aldo Moro 8, 44124 - Ferrara, Italy;

[email protected]

Paola Dal Cin Department of Pathology, Brigham, Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; [email protected]

François Desangles IRBA, Departement Effets Biologiques des Rayonnements, Laboratoire de Dosimetrie Biologique des Irradiations, Dewoitine C212,

91223 Bretigny-sur-Orge, France; [email protected]

Enric Domingo Molecular and Population Genetics Laboratory, Wellcome Trust Centre for Human Genetics, Roosevelt Dr. Oxford, OX37BN, UK

[email protected]

Ayse Elif Erson-

Bensan Department of Biological Sciences, Middle East Technical University, Ankara, Turkey; [email protected]

Ad Geurts van

Kessel

Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, 6500 HB

Nijmegen, The Netherlands; [email protected]

Oskar A. Haas Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University Vienna, Children's Cancer

Research Institute Vienna, Vienna, Austria. [email protected]

Anne Hagemeijer Center for Human Genetics, University Hospital Leuven and KU Leuven, Leuven, Belgium; [email protected]

Nyla Heerema Department of Pathology, The Ohio State University, 129 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, USA;

[email protected]

Sakari Knuutila Hartmann Institute and HUSLab, University of Helsinki, Department of Pathology, Helsinki, Finland; [email protected]

Lidia Larizza Lab Centro di Ricerche e TecnologieBiomedicheIRCCS-IstitutoAuxologico Italiano Milano, Italy; l.larizza@auxologico

Roderick Mc Leod Department of Human, Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms, Cell Cultures, Braunschweig,

Germany; [email protected]

Cristina Mecucci Hematology University of Perugia, University Hospital S.Mariadella Misericordia, Perugia, Italy; [email protected]

Fredrik Mertens Department of Clinical Genetics, University and Regional Laboratories, Lund University, SE-221 85 Lund, Sweden;

[email protected]

Konstantin Miller Institute of Human Genetics, Hannover Medical School, 30623 Hannover, Germany; [email protected]

Felix Mitelman Department of Clinical Genetics, University and Regional Laboratories, Lund University, SE-221 85 Lund, Sweden;

[email protected]

Hossain Mossafa Laboratoire CERBA, 95066 Cergy-Pontoise cedex 9, France; [email protected]

Stefan Nagel Department of Human, Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms, Cell Cultures, Braunschweig,

Germany; [email protected]

Florence Pedeutour Laboratory of Solid Tumors Genetics, Nice University Hospital, CNRSUMR 7284/INSERMU1081, France; [email protected]

Susana Raimondi Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 250, Memphis, Tennessee 38105-

3678, USA; [email protected]

Clelia Tiziana

Storlazzi Department of Biology, University of Bari, Bari, Italy; [email protected]

Sabine Strehl CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschunge.V., Vienna, Austria; [email protected]

Nancy Uhrhammer Laboratoire Diagnostic Génétique et Moléculaire, Centre Jean Perrin, Clermont-Ferrand, France; [email protected]

Dan L. Van Dyke Mayo Clinic Cytogenetics Laboratory, 200 First St SW, Rochester MN 55905, USA; [email protected]

Roberta Vanni Universita di Cagliari, Dipartimento di ScienzeBiomediche(DiSB), CittadellaUniversitaria, 09042 Monserrato (CA) - Italy;

[email protected]

Franck Viguié Service d'Histologie-Embryologie-Cytogénétique, Unité de Cytogénétique Onco-Hématologique, Hôpital Universitaire Necker-Enfants

Malades, 75015 Paris, France; [email protected]

Page 4: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS

OPEN ACCESS JOURNAL

Volume 21, Number 12, December 2017

Table of contents

Gene Section

CEBPE (CCAAT/enhancer binding protein epsilon) 438 Thomas Burmeister

MAPK4 (mitogen-activated protein kinase 4) 441 Simon Mathien, Sylvain Meloche

Leukaemia Section

del(18)(p11) 443 Lubomir Mitev, Lilya Grachlyova, Aselina Asenova

Early T-cell precursor acute lymphoblastic leukemia 447 Steven Richebourg

Multiple Myeloma 451 Matthew Ho Zhi Guang, Kenneth C. Anderson, Giada Bianchi

t(5;17)(p11;q11) and t(5;17)(q11-12;q11-12) 464 Adriana Zamecnikova, Soad al Bahar

t(5;17)(q35;q21) NPM1/RARA 466 Adriana Zamecnikova, Soad al Bahar

Solid Tumour Section

Nervous system: Astrocytoma with t(1;17)(p36;q21) SPOP/PRDM16 470 Jean-Loup Huret

Chromophobe renal cell carcinoma 472 Paola Dal Cin, Michelle S. Hirsch

Cancer Prone Disease Section

Ataxia telangiectasia (A-T) 475 Yossi Shiloh

Page 5: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Gene Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 438

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

CEBPE (CCAAT/enhancer binding protein epsilon) Thomas Burmeister

Charite, Med. Klinik fur Hamatologie, Onkologie und Tumorimmunologie, Hindenburgdamm 30,

12200 Berlin, Germany; [email protected]

Published in Atlas Database: March 2017

Online updated version : http://AtlasGeneticsOncology.org/Genes/CEBPEID42984ch14q11.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69005/03-2017-CEBPEID42984ch14q11.pdf DOI: 10.4267/2042/69005

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Review on CEBPE, with data on DNA, on the

protein encoded, and where the gene is implicated.

Keywords

CEBPE; Transcription factor; Neutrophil specific

granule deficiency; Acute lymphoblastic leukemia;

Translocation.

Identity

Other names: CRP1

HGNC (Hugo): CEBPE

Location: 14q11.2

Location (base pair)

Starts at 23117306 and ends at 23119611 bp from

pter (according to GRCh38.p7 Annotation Release

108, May 5 2016)

DNA/RNA

Description

CEBPE is located on chromosome 14q11.2 in a

telomer-centromer orientation. Conflicting data

have been published on the gene structure of

CEBPE. According to the GRCh38.p7 assembly

annotation (2016/03/21) the gene consists of two

exons (1030 bp and 517 bp), which are partially

coding and separated by a 759 bp intron (Figure

1a).

This is in conflict with some previously published

papers. Yamanaka et al. (1997) described an

alternative 3-exon-organization of the human

CEBPE gene (Figure 1b). However, exon 1, as

described by Yamanaka et al. contains a frameshift

according to the GRCh38.p7 NCBI assembly.

Transcription

Various transcripts have been reported, resulting in

four protein isoforms (Lekstrom-Himes 2001,

Yamanaka 1997; Figure 1c). All transcripts share a

common 3' end.

Protein Description

CEBPE is a member of the CCAAT/enhancer-

binding protein (C/EBP) family, which also

includes CEBPA, CEBPB, CEBPG, CEBPD and

CEBPZ (Ramji & Foka; 2002). A common

structural feature of the C/EBP proteins is the

presence of a highly conserved 55-65 amino acid

sequence at the C-terminus which encodes a basic

leucine zipper motif (bZIP domain) that functions

as a dimerization domain. In the aminoterminal part

all C/EBP proteins possess a DNA-binding domain

with relative specificity for the CCAAT DNA

motif. C/EBP proteins exert their physiological

functions as either homo- or heterodimers. They

can also interact with other bZIP- and non-bZIP

transcription factors. Different protein domains

have been characterized. The full-length CEBPE

protein basically consists of an activation domain at

the aminoterminal end, a repression domain in the

center and the leucine zipper at the carboxyterminus

(Williamson et al. 1998).

Page 6: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

CEBPE (CCAAT/enhancer binding protein epsilon) Burmeister T

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 439

Figure 1: Human CEBPE gene and mRNA transcript: a. according to the GRCh38.p7 assembly annotation (2016/03/21); b. according to Yamanaka et al. (1997); c. transcripts based on the Yamanaka exon organization

At least four different CEBPE protein isoforms of

32, 30, 27, and 14 kDa have been described, but

their functional significance is unclear (Lekstrom-

Hines et al. 2001, Figure 1c).

The CEBPE translation product can undergo a

number of post-translational modifications.

Phosphorylation of CEBPE on threonine 75,

located in the transactivation domain, is associated

with increased DNA binding capacity and

transcriptional activation (Williamson et al., 2005).

Sumoylation of lysine residues within the

repression domain has been found to modulate

CEBPE function (Kim et al., 2005). Acetylation of

lysine-121 and lysine-198 was found to be critical

for terminal neutrophil differentiation (Bartels et.

al., 2015).

Expression

CEBPE is predominantly expressed in cells of the

hematopoietic system and to a much lesser extent in

ovarian tissue (Yamanaka, et al., 1997). In normal

hematopoietic cells CEBPE is preferentially

expressed in myeloid-committed cells and the

protein is virtually only detectable in

metamyelocytes and myelocytes. The gene is also

expressed in more immature myeloid cells but

protein translation is repressed by miRNA-130a

(Larsen et al., 2014).

The expression of CEBPE protein induces growth

arrest, morphological differentiation, secondary

granule proteins and has proapoptotic effects

(Nakajima et al. 2006).

Localisation

Nuclear.

Function

CEBPE is a transcription factor, important for

monocyte and granulocyte development. The

transcription factor binds as a homodimer or

heterodimer (with CEBPD) to specific DNA

regulatory regions. Shorter CEBPE protein

isoforms are hypothetical attenuators of the

transcriptional activity of the long isoform.

Homozygous CEBPE knock-out (-/-) mice appear

healthy at birth but survive only 2-5 months after

birth, while heterozygous CEBPE knock-out mice

appear normal. CEBPE (-/-) mice showed a marked

increase in immature myeloid progenitors,

increased numbers of morphologically abnormal

neutrophils, that were functionally defect and

lacked an oxidative burst, and decreased numbers

of eosinophils. Thus it was concluded that CEBPE

is essential for a normal terminal differentiation of

committed granulocyte progenitor cells (Yamanaka,

et al., 1997).

Page 7: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

CEBPE (CCAAT/enhancer binding protein epsilon) Burmeister T

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 440

Implicated in

Neutrophil specific granule deficiency (SGD)

Some, but not all of the very few known patients

with SGD harboured CEBPE mutations which led

to loss of the dimerization domain; phenotypically,

SGD patients show bilobed nuclei, impaired

chemotaxis and bactericidal activity with

susceptibility to severe bacterial infections

(Gombart & Koeffler 2002).

Acute lymphoblastic leukemia (ALL)

The CEBPE single nucleotide polymorphism

rs2239633 has been implicated as a susceptibility

factor for the development of B lineage ALL in

children and adults. The relative risk (odds ratio)

conferred is 1.1-1.6 (Papaemmanuil et al., 2009;

Burmeister et al. 2014).

t(14;14)(q11;q32) CEBPE/IGH and inv(14)(q11q32) CEBPE/IGH

CEBPE is found recurrently translocated to the

immunoglobulin heavy chain locus ( IGH) on

14q32 in acute lymphoblastic leukemia patients

with inv(14)(q11q32)/t(14;14)(q11;q32). The

translocation leads to an overexpression of CEBPE

under the control of the immunoglobulin heavy

chain gene promoters. At least five cases have been

described (Akasaka et al. 2007).

References Akasaka T, Balasas T, Russell LJ, Sugimoto KJ, Majid A, Walewska R, Karran EL, Brown DG, Cain K, Harder L, Gesk S, Martin-Subero JI, Atherton MG, Brüggemann M, Calasanz MJ, Davies T, Haas OA, Hagemeijer A, Kempski H, Lessard M, Lillington DM, Moore S, Nguyen-Khac F, Radford-Weiss I, Schoch C, Struski S, Talley P, Welham MJ, Worley H, Strefford JC, Harrison CJ, Siebert R, Dyer MJ. Five members of the CEBP transcription factor family are targeted by recurrent IGH translocations in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Blood. 2007 Apr 15;109(8):3451-61

Bartels M, Govers AM, Fleskens V, Lourenço AR, Pals CE, Vervoort SJ, van Gent R, Brenkman AB, Bierings MB, Ackerman SJ, van Loosdregt J, Coffer PJ. Acetylation of C/EBPε is a prerequisite for terminal neutrophil differentiation. Blood. 2015 Mar 12;125(11):1782-92

Burmeister T, Bartels G, Gröger D, Trautmann H, Schwartz S, Lenz K, Tietze-Bürger C, Viardot A, Wäsch R, Horst HA, Reinhardt R, Gökbuget N, Hoelzer D, Kneba M, Brüggemann M. Germline variants in IKZF1, ARID5B, and CEBPE as risk factors for adult-onset acute lymphoblastic leukemia: an analysis from the GMALL study group.

Haematologica. 2014 Feb;99(2):e23-5

Gombart AF, Koeffler HP. Neutrophil specific granule deficiency and mutations in the gene encoding transcription factor C/EBP(epsilon). Curr Opin Hematol. 2002 Jan;9(1):36-42

Han Y, Xue Y, Zhang J, Wu Y, Pan J, Wang Y, Shen J, Dai H, Bai S. Translocation (14;14)(q11;q32) with simultaneous involvement of the IGH and CEBPE genes in B-lineage acute lymphoblastic leukemia. Cancer Genet Cytogenet. 2008 Dec;187(2):125-9

Kim J, Sharma S, Li Y, Cobos E, Palvimo JJ, Williams SC. Repression and coactivation of CCAAT/enhancer-binding protein epsilon by sumoylation and protein inhibitor of activated STATx proteins. J Biol Chem. 2005 Apr 1;280(13):12246-54

Larsen MT, Häger M, Glenthøj A, Asmar F, Clemmensen SN, Mora-Jensen H, Borregaard N, Cowland JB. miRNA-130a regulates C/EBP-ε expression during granulopoiesis. Blood. 2014 Feb 13;123(7):1079-89

Lekstrom-Himes JA. The role of C/EBP(epsilon) in the terminal stages of granulocyte differentiation Stem Cells 2001;19(2):125-33

Nakajima H, Watanabe N, Shibata F, Kitamura T, Ikeda Y, Handa M. N-terminal region of CCAAT/enhancer-binding protein epsilon is critical for cell cycle arrest, apoptosis, and functional maturation during myeloid differentiation J Biol Chem 2006 May 19;281(20):14494-502

Papaemmanuil E, Hosking FJ, Vijayakrishnan J, Price A, Olver B, Sheridan E, Kinsey SE, Lightfoot T, Roman E, Irving JA, Allan JM, Tomlinson IP, Taylor M, Greaves M, Houlston RS. Loci on 7p12 2, 10q21 2 and 14q11

Prasad RB, Hosking FJ, Vijayakrishnan J, Papaemmanuil E, Koehler R, Greaves M, Sheridan E, Gast A, Kinsey SE, Lightfoot T, Roman E, Taylor M, Pritchard-Jones K, Stanulla M, Schrappe M, Bartram CR, Houlston RS, Kumar R, Hemminki K. Verification of the susceptibility loci on 7p12 2, 10q21 2, and 14q11

Ramji DP, Foka P. CCAAT/enhancer-binding proteins: structure, function and regulation Biochem J 2002 Aug 1;365(Pt 3):561-75

Williamson EA, Williamson IK, Chumakov AM, Friedman AD, Koeffler HP. CCAAT/enhancer binding protein epsilon: changes in function upon phosphorylation by p38 MAP kinase Blood 2005 May 15;105(10):3841-7

Yamanaka R, Kim GD, Radomska HS, Lekstrom-Himes J, Smith LT, Antonson P, Tenen DG, Xanthopoulos KG. CCAAT/enhancer binding protein epsilon is preferentially up-regulated during granulocytic differentiation and its functional versatility is determined by alternative use of promoters and differential splicing Proc Natl Acad Sci U S A 1997 Jun 10;94(12):6462-7

This article should be referenced as such:

Burmeister T. CEBPE (CCAAT/enhancer binding protein epsilon). Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):438-440.

Page 8: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Gene Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 441

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

MAPK4 (mitogen-activated protein kinase 4) Simon Mathien, Sylvain Meloche

Institute of Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec H3C 3J7,

Canada; [email protected]; [email protected]

Published in Atlas Database: March 2017

Online updated version : http://AtlasGeneticsOncology.org/Genes/MAPK4ID41293ch18q21.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69006/03-2017-MAPK4ID41293ch18q21.pdf DOI: 10.4267/2042/69006

This article is an update of : MAPK4 (mitogen-activated protein kinase 4). Atlas Genet Cytogenet Oncol Haematol 2017;21(12) Meloche S. MAPK4 (mitogen-activated protein kinase 4). Atlas Genet Cytogenet Oncol Haematol 2009;13(1)

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Review on MAPK4, with data on DNA, on the

protein encoded, and where the gene is implicated.

Keywords

MAPK4; ERK4; Kinase; signaling pathway; RAS-

RAF-MAPK pathway

Identity

HGNC (Hugo): MAPK4

Location: 18q21.1

Other names: ERK4, PRKM4, pP63mapk

Local order: The MAPK4 gene is located between

the genes SKA1 (C18orf24) and MRO on

chromosome 18

DNA/RNA

Description

The MAPK4 gene spans 171.7 kb on the long arm

of chromosome 18 and is transcribed in the

centromere-to-telomere orientation.

The gene is composed of 6 exons with the

translation initiation codon located in exon 2. The

first two exons are separated by a long intron of

102.8 kb.

Transcription The MAPK4 transcribed mRNA has 4,736 bp. No

splice variants have been reported.

Pseudogene None.

Protein Description

Extracellular signal-regulated kinase 4 (ERK4) is

an atypical member of the mitogen-activated

protein (MAP) kinase family of serine/threonine

kinases. The human ERK4 protein is made of 587

amino acids and contains a typical kinase domain

located at the N-terminal extremity. Another region

with homology to the MAP kinase ERK3 (C34

domain) has been identified after the kinase

domain. The function of the C34 domain is

unknown.

Figure 1. Genomic organization of the MAPK4 gene on chromosome 18.

Page 9: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

MAPK4 (mitogen-activated protein kinase 4) Mathien S, Meloche S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 442

Figure 2. Schematic representation of the ERK4 protein structure. Kinase, catalytic kinase domain; C34 conserved region in ERK3 and ERK4; SEG, activation loop motif containing the regulatory phosphorylation residue Ser186.

Expression

MAPK4 mRNA is expressed to the highest level in

the brain. Other sites of expression include the

heart, lung, kidney, intestine, pancreas, parathyroid

gland, prostate, thymus, ovary, eye and ear.

Localisation

ERK4 localizes to the cytoplasm and nucleus of a

variety of cultured cells.

Function

Little is known about the regulation and functions

of ERK4. The only known substrate of ERK4 is the

protein kinase MAPKAPK5 (MK5).

Homology

ERK4 display 73% amino acid identity with ERK3

in the kinase domain. ERK4 and ERK3 define a

distinct subfamily of MAP kinases.

Mutations The R114C/H mutation has been reported in several

types of cancer (colorectal adenocarcinoma, diffuse

glioma, non-small cell lung cancer, cutaneous

melanoma, stomach adenocarcinoma, uterine

carcinosarcoma). The functional impact of this

mutation on the expression or activity of ERK4 is

not known.

Implicated in

Cancer

Analysis of copy number alterations (CNAs) from

TCGA datasets show that MAPK4 gene is deleted

in several adenocarcinomas, includingprostate,

esophageal, stomach and lung adenocarcinomas.

Also, a chromosomal subregion containing the

MAPK4 gene is deleted with high frequency (23%)

in pancreatic adenocarcinoma. On the other hand,

MAPK4 is amplified with low frequency in

neuroendocrine prostate cancer, diffuse large B-cell

lymphoma and sarcomas.

Interrogation of the Oncomine database reveals that

expression of MAPK4 mRNA is downregulated in

breast and prostate cancer.

Consistent with CNA analysis, MAPK4 mRNA is

upregulated in diffuse large B-cell lymphoma.

References Aberg E, Perander M, Johansen B, Julien C, Meloche S, Keyse SM, Seternes OM. Regulation of MAPK-activated protein kinase 5 activity and subcellular localization by the atypical MAPK ERK4/MAPK4. J Biol Chem. 2006 Nov 17;281(46):35499-510

Coulombe P, Meloche S. Atypical mitogen-activated protein kinases: structure, regulation and functions. Biochim Biophys Acta. 2007 Aug;1773(8):1376-87

Gonzalez FA, Raden DL, Rigby MR, Davis RJ. Heterogeneous expression of four MAP kinase isoforms in human tissues. FEBS Lett. 1992 Jun 15;304(2-3):170-8

Kant S, Schumacher S, Singh MK, Kispert A, Kotlyarov A, Gaestel M. Characterization of the atypical MAPK ERK4 and its activation of the MAPK-activated protein kinase MK5. J Biol Chem. 2006 Nov 17;281(46):35511-9

Nowak NJ, Gaile D, Conroy JM, McQuaid D, Cowell J, Carter R, Goggins MG, Hruban RH, Maitra A. Genome-wide aberrations in pancreatic adenocarcinoma. Cancer Genet Cytogenet. 2005 Aug;161(1):36-50

Turgeon B, Lang BF, Meloche S. The protein kinase ERK3 is encoded by a single functional gene: genomic analysis of the ERK3 gene family. Genomics. 2002 Dec;80(6):673-80

This article should be referenced as such:

Mathien S, Meloche S. MAPK4 (mitogen-activated protein kinase 4). Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):441-442.

Page 10: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 443

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

del(18)(p11) Lubomir Mitev, Lilya Grachlyova, Aselina Asenova

Military Medical Academy, Department of Cytogenetics and Molecular Biology, Sofia, Bulgaria,

[email protected]

Published in Atlas Database: February 2017

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/del18p11ID1314.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69007/02-2017-del18p11ID1314.pdf DOI: 10.4267/2042/69007

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract Review on del(18)(p11) in haematological

malignancies, with data on the genes possibly

involved.

Keywords

Chromosome 18; Chronic myeloid leukemia;

Myelodysplastic syndromes; Acute myeloid

leukemia; Acute lymphoblastic leukemia; Follicular

lymphoma

Clinics and pathology del(18)(p11) is a very rare structural anomaly found

only in 24 cases with onco-hematological disorders.

This deletion has been observed in both myeloid

(14 cases) and lymphoid (10 cases) malignancies

and has been predominantly associated with a

complex karyotype. In all cases 18p- is presented as

a single copy in the karyotype. The anomaly was

determined by conventional cytogenetic as terminal

deletion with sub-band location of the breakpoint in

18p11.2. Microarray comparative genome

hybridization (aCGH) studies in patients with acute

myeloid leukemia have demonstrated that the

deleted segment in 18p could be also interstitial and

variable in size as has been proven in other tumor

associated deletions. Three submicroscopic deleted

regions have been identified - 18p11.32-p11.31,

18p11.23 and 18p11.22-p11.21 (Itzhar et al, 2011).

Disease

Acute Myeloid Leukemia (AML)

Epidemiology

del(18)(p11) is found in 9 cases ( 0.1% of all AML

cases with an abnormal karyotype) (Alimena et al.,

1981; Brodeur et al., 1983; GFCH, 1990; Lawler et

al., 1990; Mohamed et al., 1993; Davey et al., 1995;

Krauter et al., 1998; Babicka et al., 2007; Kasyan et

al., 2010) - 1 case with M1 French-American-

British (FAB) phenotype, 2 cases with M2, 1 case

with M3, 1 case with M4, 1 case with M6 and 3

with AML, NOS (one diagnosed as therapy related

AML).

The sex ratio is significantly unbalanced, near

M:F=3.5:1.

The age is documented in 5 cases: 6 and 35 (M2),

59 (M3), 42 (M4) and 61 (M6).

Cytogenetics

In 5 cases del(18)(p11) is found in complex

karyotypes and in one as a sole anomaly. Three

cases are with -5/del(5q), two with +8 and one case

of each of the following well known

rearrangements: -7; del(17p); t(8;21)(q22;q22);

t(17;20)(q21;q11) in a M3 ( RARA not checked). In

one case both arms of chromosome 18 are affected-

der(18)del(18)(p11)del(18)(q21). In 3 cases 18p- is

associated with sex chromosome abnormalities - 2

with a loss of Y chromosome and one with

del(X)(q26).

Disease

Other Myeloproliferative Disorders

Epidemiology

del(18)(p11) is described in 2 cases (one male and

one female) with chronic myeloid leukemia (CML)

(Ohyashiki et al., 1997; Sun et al,. 2011), 2 cases

(males) with a myelodysplastic syndrome (Cerretini

et al., 2002; Volkert et al., 2014) and one case

(female) with primary myelofibrosis (PMF)

involving a lymph node (Hu et al., 2009).

Page 11: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

del(18)(p11) Mitev L, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 444

Cytogenetics

In 2 cases with MDS and one case with CML

del(18)(p11) is associated with a complex

karyotype, in one case with CML it is a second

event and in one case (with PMF) it is a sole

anomaly.

In the two cases with CML 18p- anomaly is present

in additional deviating sub-clones (sidelines) and in

one case with MDS in two clones with

dic(17;20)(p11-12;q11) and dic(18;18)(p11;p11).

Disease

Acute Lymphoblastic Leukemia (ALL)

Epidemiology

del(18)(p11) is found in 8 cases with ALL (0.08%

of all ALL cases with an abnormal karyotype)

(Oshimura et al., 1977; Kanerva et al., 2002; van

der Burg et al., 2002; Soulier et al., 2003; Karst et

al., 2006; Shin et al., 2011; Schmiegelow et al.,

2012; Lundin et al., 2014).

The sex ratio is significantly unbalanced, near

M:F=1.7:1.

The anomaly has been observed in young, as well

as older patients (average age 22.6 years; range 5-

65).

Cytogenetics

In 5 cases del(18)(p11) is found in complex

karyotypes and in 3 cases it is accompanied with

second anomalies - der(21) in two cases and

dup(18q)(q11q11) in one (unrelated clone). In 4 of

the 5 cases with complex karyotypes 18p- is

associated with one or more deletions including 6p-

, 9p-, 11q- and 12p- and in two cases with

t(7;14)(p13;q32) and t(14;18)(q32;q21) (Shin et al.,

2011), respectively.

Disease

Follicular lymphoma (FL)

Cytogenetics

del(18)(p11) is reported in 2 cases with FL (Bosga-

Bouwer et al., 2003; Horsman et al., 2003). Both

have hyperdiplpoid, highly complex karyotypes.

Genetics A tumor suppressor gene(s) (TSG) important to the

described hematological malignances may be is

involved in the molecular pathogenesis of

del(18)(p11).

A number of TSG candidates are mapped in the

deleted segment 18p11.2->18pter including PTPN2

(18p11.21), LDLRAD4 (C18orf1) (18p11.21),

RALBP1 (18p11.22), PPP4R1 (18p11.22), PTPRM

(18p11.23), EPB41L3 (18p11.31), L3MBTL4

(18p11.31), TGIF1 (18p11.31), ZEP161 (18p11.31)

and SMCHD1 (18p11.32).

Almost all are implicated in the pathogenesis of a

variety of solid tumors, but there are several lines of

data suggesting that four of them (PTPN2, PTPRM,

TGIF1 and SMCHD1) could also have a role in

leukogenesis and respectively may act as TSG in

the hematopoietic system.

Genes involved and proteins

PTPN2 (protein tyrosine phosphatase, non-receptor type 2)

Location

18p11.21

Protein

PTPN2 (Tyrosine-protein phosphatase non-receptor

type2) is a negative regulator of multiple signaling

pathways, including IL2 (interleukin 2) mediated

JAK2/STAT cascade and is inactivated by nonsense

mutation in 5% and deleted in 6% of the cases with

adult acute T-cell lymphoblastic leukemia (Kleppe

et al., 2010).

PTPRM (protein tyrosine phosphatase, receptor type M)

Location

18p11.23

Protein

PTPRM (Tyrosine-protein phosphatase, receptor

type M) is targeted by aCGH within the deleted

region 18p11.23 found in 17 cases with AML.

The protein encoded by this gene is involved in

cell-cell adhesion through hemophilic interactions.

Hypermethylation of PTPRM in cases with ALL

(Stevenson et al, 2013) and submicroscopic

deletion of PTPRM in cases of adult T cell

leukemia/lymphoma have been observed (Kataoka

et al 2015).

TGIF1 (TGFB induced factor homeobox 1)

Location

18p11.31

Protein

TGiF1 (TG-interacting factor 1) is targeted by

aCGH within the deleted region 18p11.32p11.31

found in 14 cases with AML.

The protein encoded by this gene plays an essential

role in the regulation of hematopoiesis inhibiting

the signaling pathways of RA (retinoic acid) and

TGFB1 (transforming growth factor beta) by

transcriptional repression of SMAD2.

It has been demonstrated that TGIF1 is a negative

regulator of KMT2A (MLL)-rearranged AML

(Willer et al, 2014).

Page 12: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

del(18)(p11) Mitev L, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 445

SMCHD1 (structural maintenance of chromosomes flexible hinge domain containing 1)

Location

18p11.32

Protein

SMCHD1 (Structural maintenance of chromosome

flexible hinge domain containing 1) is also targeted

by aCGH within the region 18p11.32p11.31.

This gene encodes a protein required for the

maintenance of an X chromosome inactivation in

females. It has been reported that SMCHD1 is

associated with increased tumorigenesis in a mouse

model.

Global gene expressing profiling revealed that

SMCHD1 normally repress genes activated by

MLL- chimeric fusion proteins in leukemia (Leong

et al, 2012).

These data have suggested that the loss of

SMCHD1 may cause hematological malignances of

both cell lineage - myeloid and lymphoid.

References Minami Y, Emori Y, Kawasaki H, Suzuki K. E-F hand structure-domain of calcium-activated neutral protease (CANP) can bind Ca2+ ions. J Biochem. 1987 Apr;101(4):889-95

Carmichael IH, Koster S. Bovine parafilariosis in Southern Africa: a preliminary report. Onderstepoort J Vet Res. 1978 Sep;45(3):213-4

Volkert S, Kohlmann A, Schnittger S, Kern W, Haferlach T, Haferlach C. Association of the type of 5q loss with complex karyotype, clonal evolution, TP53 mutation status, and prognosis in acute myeloid leukemia and myelodysplastic syndrome. Genes Chromosomes Cancer. 2014 May;53(5):402-10

Davey FR, Abraham N Jr, Brunetto VL, MacCallum JM, Nelson DA, Ball ED, Griffin JD, Baer MR, Wurster-Hill D, Mayer RJ. Morphologic characteristics of erythroleukemia (acute myeloid leukemia; FAB-M6): a CALGB study. Am J Hematol. 1995 May;49(1):29-38

Akakpo AJ, Some MJ, Bornarel P, Jouan A, Gonzalez JP. [Epidemiology of Rift Valley fever in western Africa. I. Serologic survey in domestic ruminants of Burkina Faso]. Bull Soc Pathol Exot Filiales. 1989;82(3):321-31

Leong HS, Chen K, Hu Y, Lee S, Corbin J, Pakusch M, Murphy JM, Majewski IJ, Smyth GK, Alexander WS, Hilton DJ, Blewitt ME. Epigenetic regulator Smchd1 functions as a tumor suppressor. Cancer Res. 2013 Mar 1;73(5):1591-9

Willer A, Jakobsen JS, Ohlsson E, Rapin N, Waage J, Billing M, Bullinger L, Karlsson S, Porse BT. TGIF1 is a negative regulator of MLL-rearranged acute myeloid leukemia. Leukemia. 2015 May;29(5):1018-31

Alimena G, Dallapiccola B, De Cuia MR, Mandelli F, Mitelman F. Acute lymphocytic and myelomonocytic leukemia associated with low platelet counts and a 21q- marker chromosome. Hum Genet. 1981;57(3):329-31

Babicka L, Ransdorfova S, Brezinova J, Zemanova Z, Sindelarova L, Siskova M, Maaloufova J, Cermak J, Michalova K.. Analysis of complex chromosomal

rearrangements in adult patients with MDS and AML by multicolor FISH. Leuk Res. 2007 Jan;31(1):39-47. Epub 2006 May 9.

Bosga-Bouwer AG, van Imhoff GW, Boonstra R, van der Veen A, Haralambieva E, van den Berg A, de Jong B, Krause V, Palmer MC, Coupland R, Kluin PM, van den Berg E, Poppema S. Follicular lymphoma grade 3B includes 3 cytogenetically defined subgroups with primary t(14;18), 3q27, or other translocations: t(14;18) and 3q27 are mutually exclusive. Blood. 2003 Feb 1;101(3):1149-54.

Brodeur GM, Williams DL, Kalwinsky DK, Williams KJ, Dahl GV.. Cytogenetic features of acute nonlymphoblastic leukemia in 73 children and adolescents. Cancer Genet Cytogenet. 1983 Feb;8(2):93-105.

Cerretini R, Acevedo S, Chena C, Belli C, Larripa I, Slavutsky I.. Evaluation of constitutional chromosome aberrations in hematologic disorders. Cancer Genet Cytogenet. 2002 Apr 15;134(2):133-7

GFCH (Groupe Francais de Cytogenetique Hematologique). 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.

Horsman DE, Okamoto I, Ludkovski O, Le N, Harder L, Gesk S, Siebert R, Chhanabhai M, Sehn L, Connors JM, Gascoyne RD.. Follicular lymphoma lacking the t(14;18)(q32;q21): identification of two disease subtypes. Br J Haematol. 2003 Feb;120(3):424-33.

Kanerva J, Vettenranta K, Autio K, Knuutila S, Saarinen-Pihkala UM.. Minimal residual disease by metaphase FISH in children with ALL: clonal cells during or after chemotherapy may not predict relapse. Leuk Res. 2002 Jun;26(6):545-50

Karst C, Gross M, Haase D, Wedding U, Höffken K, Liehr T, Mkrtchyan H. Novel cryptic chromosomal rearrangements detected in acute lymphoblastic leukemia detected by application of new multicolor fluorescent in situ hybridization approaches. Int J Oncol. 2006 Apr;28(4):891-7.

Kasyan A, Medeiros LJ, Zuo Z, Santos FP, Ravandi-Kashani F, Miranda R, Vadhan-Raj S, Koeppen H, Bueso-Ramos CE.. Acute erythroid leukemia as defined in the World Health Organization classification is a rare and pathogenetically heterogeneous disease. Mod Pathol. 2010 Aug;23(8):1113-26. doi: 10.1038/modpathol.2010.96. Epub 2010 May 14.

Kataoka K, Nagata Y, Kitanaka A, Shiraishi Y, Shimamura T, Yasunaga J, Totoki Y, Chiba K, Sato-Otsubo A, Nagae G, Ishii R Muto S, Kotani S, Watatani Y, Takeda J, Sanada M, Tanaka H, Suzuki H, Sato Y, Shiozawa Y, Yoshizato T, Yoshida K, Makishima H, Iwanaga M, Ma G,Nosaka K, Hishizawa M, Itonaga H, Imaizumi Y, Munakata W, Ogasawara H, Sato T, Sasai K, Muramoto K, Penova M, Kawaguchi T,Nakamura H, Hama N, Shide K, Kubuki Y, Hidaka T, Kameda T, Nakamaki T, Ishiyama K, Miyawaki S, Yoon SS, Tobinai K, Miyazaki Y,Takaori-Kondo A, Matsuda F, Takeuchi K, Nureki O, Aburatani H, Watanabe T, Shibata T, Matsuoka M, Miyano S, Shimoda K, Ogawa S. Integrated molecular analysis of adult T cell leukemia/lymphoma. Nat Genet. 2015 Nov;47(11):1304-15. doi: 10.1038/ng.3415. Epub 2015 Oct 5.

Kleppe M, Lahortiga I, El Chaar T, De Keersmaecker K, Mentens N, Graux C, Van Roosbroeck K, Ferrando AA, Langerak AW, Meijerink JP, Sigaux F,Haferlach T, Wlodarska I, Vandenberghe P, Soulier J, Cools J.. Deletion of the protein tyrosine phosphatase gene PTPN2

Page 13: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

del(18)(p11) Mitev L, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 446

in T-cell acute lymphoblastic leukemia. Nature Genetics 2010 Jun;42(6):530-5. doi: 10.1038/ng.587. Epub 2010 May 16.

Krauter J, Peter W, Pascheberg U, Heinze B, Bergmann L, Hoelzer D, Lübbert M, Schlimok G, Arnold R, Kirchner H, Port M, Ganser A. Detection of karyotypic aberrations in acute myeloblastic leukaemia: a prospective comparison between PCR/FISH and standard cytogenetics in 140 patients with de novo AML. Br J Haematol. 1998 Oct;103(1):72-8

Lawler SD, Khokhar MT, Davies H, Williams GJ, Powles R.. Cytogenetic studies of leukemic recurrence in recipients of bone marrow allografts. Cancer Genet Cytogenet. 1990 Jul 15;47(2):249-63.

Lundin C, Forestier E, Klarskov Andersen M, Autio K, Barbany G, Cavelier L, Golovleva I, Heim S, Heinonen K, Hovland R, Johannsson JH, Kjeldsen E,Nordgren A, Palmqvist L, Johansson B; Nordic Society of Pediatric Hematology Oncology (NOPHO); Swedish Cytogenetic Leukemia Study Group (SCLSG);NOPHO Leukemia Cytogenetic Study Group (NLCSG). Clinical and genetic features of pediatric acute lymphoblastic leukemia in Down syndrome in the Nordic countries. J Hematol Oncol. 2014 Apr 11;7:32. doi: 10.1186/1756-8722-7-32.

Mitelman F, Johansson B, Mertens F. Mitelman Database of Chromosome Aberration in Cancer https://cgap.nci.nih.gov/

Ohyashiki K, Ohyashiki JH, Iwama H, Hayashi S, Shay JW, Toyama K.. Telomerase activity and cytogenetic changes in chronic myeloid leukemia with disease progression. Leukemia. 1997 Feb;11(2):190-4

Oshimura M, Freeman AI, Sandberg AA.. Chromosomes and causation of human cancer and leukemia. XXVI. Banding studies in acute lymphoblastic leukemia (ALL). Cancer. 1977 Sep;40(3):1161-72

Schmiegelow K, Lausten Thomsen U, Baruchel A, Pacheco CE, Pieters R, Pombo-de-Oliveira

MS, Andersen EW, Rostgaard K, Hjalgrim H, Pui CH. High concordance of subtypes of childhood acute lymphoblastic leukemia within families: lessons from sibships with multiple cases of leukemia. Leukemia. 2012 Apr;26(4):675-81. doi: 10.1038/leu.2011.274. Epub 2011 Oct 18.

Shin DY, Kim I, Kim KH, Choi Y, Beom SH, Yang Y, Lim Y, Lee E, Lee JK, Kim JY, Kim HK, Yoon SS, Lee DS, Park S, Kim BK.. Acute lymphoblastic leukemia in elderly patients: a single institution's experience. Korean J Intern Med. 2011 Sep;26(3):328-39. doi: 10.3904/kjim.2011.26.3.328. Epub 2011 Sep 13

Soulier J, Trakhtenbrot L, Najfeld V, Lipton JM, Mathew S, Avet-Loiseau H, De Braekeleer M, Salem S, Baruchel A, Raimondi SC, Raynaud SD.. Amplification of band q22 of chromosome 21, including AML1, in older children with acute lymphoblastic leukemia: an emerging molecular cytogenetic subgroup. Leukemia. 2003 Aug;17(8):1679-82.

Stevenson WS, Best OG, Przybylla A, Chen Q, Singh N, Koleth M, Pierce S, Kennedy T, Tong W, Kuang SQ, Garcia-Manero G.. DNA methylation of membrane-bound tyrosine phosphatase genes in acute lymphoblastic leukaemia. Leukemia. 2014 Apr;28(4):787-93. doi: 10.1038/leu.2013.270. Epub 2013 Sep 18.

Sun J, Yin CC, Cui W, Chen SS, Medeiros LJ, Lu G.. Chromosome 20q deletion: a recurrent cytogenetic abnormality in patients with chronic myelogenous leukemia in remission. Am J Clin Pathol. 2011 Mar;135(3):391-7. doi: 10.1309/AJCPQFSC9ZJNMAZ6

van der Burg M, Smit B, Brinkhof B, Barendregt BH, Verschuren MC, Dib M, Beverloo HB, van Dongen JJ, Langerak AW.. A single split-signal FISH probe set allows detection of TAL1 translocations as well as SIL-TAL1 fusion genes in a single test. Leukemia. 2002 Apr;16(4):755-61.

This article should be referenced as such:

Mitev L, Grachlyova L, Asenova A. del(18)(p11). Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):443-446.

Page 14: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 447

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

Early T-cell precursor acute lymphoblastic leukemia Steven Richebourg

Laboratoire de cytogénétique onco-hématologique, Département de pathologie, Hôpital du Saint

Sacrement, CHU de Québec Université Laval, 1050, Chemin Sainte Foy, Département de Médecine

Moléculaire, Faculté de médecine, Université Laval, Québec, G1S4L8 Québec, Qc, Canada;

[email protected]

Published in Atlas Database: February 2017

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/EarlyTcellALLID1667.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69008/02-2017-EarlyTcellALLID1667.pdf DOI: 10.4267/2042/69008

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract Review on early T-cell precursor acute

lymphoblastic leukemia, with data on clinics and

the genes possibly involved.

Keywords

T-cell; acute lymphoblastic leukemia

Identity

Other names

Early T-cell precursor lymphoblastic

leukemia/ETP-ALL/ETP T-ALL

Clinics and pathology

Note

Identified in 2009 from gene expression profiling

data, Early T cell Precursor Acute Lymphoblastic

Leukemia (ETP-ALL) represents a subset of T-

ALL sharing transcriptional and immunophenotypic

similarities with early T-cell precursors (Coustan-

Smith et al., 2009).

ETP-ALL is currently defined by a distinctive

phenotype characterized by a lack of expression of

the T-lineage cell surface markers CD1a and CD8,

weak or absent expression of CD5 and aberrant

expression of one or more myeloid or stem cell

markers (Coustan-Smith et al., 2009).

Since the description, genetic alterations and

prognostic data have been reported in literature

improving our understanding of this subgroup.

Therefore, ETP-ALL has been included as a

provisional entity in the 2016 revision of the WHO

classification of Acute Leukemias (Arber et al.,

2016).

Disease

Phenotype/cell stem origin

Early T-cell precursors (ETPs) are immature

progenitors that have recently immigrated from the

bone marrow to the thymus and which retain a

multilineage differentiation potential (T-lymphoid,

natural killer, dendritic and myeloid cell

differentiation potential). Animal model based

studies of ETPs demonstrate similarities with

immature myeloid progenitors and hematopoietic

stem cells (Bell and Bandhoola, 2008; Wada et al.,

2008). Because gene expression profiling is not part

of routine laboratory investigations, ETP-ALL

cases are currently identified through the phenotype

of blast cells : CD1a-, CD8-, CD5- /weak, and

positivity for one or more stem cell and/or myeloid

antigens (CD117, CD34, HLA-DR, CD13, CD33,

CD11b, and/or CD65) (Coustan-Smith et al., 2009;

Chopra et al., 2014). ETP-ALL typically also

express CD2, CD7 and cytoplasmic CD3 and may

express CD4.

Epidemiology

Initially described from children ALL cohorts,

ETP-ALL have also been identified in adults.

Frequency of ETP-ALL varies among studies

around 10-15% of T ALL.

Page 15: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Early T-cell precursor acute lymphoblastic leukemia Richebourg S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 448

Courtesy Linda Boulay and Frédéric Barabé, Flow Cytometry Laboratory, Hôpital du Saint Sacrement, CHU de Québec Université Laval

In children, ETP-ALL has been reported in 11% to

16% of T-ALL (Coustan-Smith et al., 2009; Patrick

et al., 2014).

In adults, ETP-ALL frequency ranges from 7,4%

(Neumann et al., 2012) to 17% of T-ALL (Jain et

al., 2016).

Based on gene expression profiling, some authors

suggest that there is an immature signature related

to ETPs (called "near ETP' or "close to ETP")

which may be more prevalent (Van Vlierberghe et

al., 2013; Haydu and Ferrando, 2013).

Clinics

Most studies report no significant association

between ETP-ALL signature and clinical features

including sex, age, white blood cell count, and

central nervous system involvement (Coustan-

Smith et al., 2009; Inukai et al., 2012; Neumann et

al., 2012).

Though, two no recurrent features were identified :

an older age in paediatric population (Coustan-

Smith et al., 2009) and a lower frequency of

mediastinal mass at diagnosis in adult population

(Neumann et al., 2012).

Cytology

No specific morphologic features have been

reported to date.

Treatment

Since there is no consensus on the prognosis (see

below), no specific protocol is recommended.

Because of the trend to negative impact on

prognosis, some authors suggest that new

therapeutic strategies are needed to improve the

outcomes of ETP-ALL (Jain et al., 2016; Neumann

et al., 2013), including the use myeloid-targeted

therapies such as tyrosine kinase inhibitors

(Neumann et al., 2012; Neumann et al., 2013;

Zhang et al., 2012)

Prognosis

There is currently no consensus on the prognosis of

ETP-ALL.

Initial prognostic studies between 2009 and 2012

reported a negative prognostic impact on response

rate and survival (Coustan-Smith et al., 2009,

Inukai et al., 2012; Ma et al., 2012) and a higher

risk of relapse (Allen and al., 2013).

These data were not confirmed in more recent

studies with larger cohorts (Brent et al., 2014;

Patrick et al., 2014; Zuurbier et al., 2014).

However, in 2016, a MD Anderson study reported

again a negative prognostic impact on the response

rate and the overall survival of patients with ETP-

ALL (Jain et al., 2016).

Page 16: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Early T-cell precursor acute lymphoblastic leukemia Richebourg S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 449

Cytogenetics No specific cytogenetic abnormality is associated

with ETP-ALL subtype.

In most studies, ETP-ALL patients present highly

variable karyotypes with remarkably a lower

frequency of classic recurrent rearrangements

associated with T-ALL (Patrick et al., 2014).

Coustan Smith and al reported a higher frequency

of deletion 13q (Coustan-Smith et al., 2009) and

Patrick et al a higher frequency of KMT2A

rearrangements (Patrick et al., 2014).

SNP analysis demonstrated a higher frequency of

copy number alterations in ETP-ALL vs non ETP-

ALL (Coustan-Smith et al., 2009).

Genes involved and proteins

ETP-ALL subgroup is characterized by a high

genetic heterogeneity and present a distinct

genomic profile defined by a lower incidence of

typical mutations associated with T ALL such as

NOTCH1 or FBXW7 and a high frequency of

myeloid associated gene mutations ( FLT3, RAS

mutations, DNMT3A, IDH1 / IDH2 mutations)

(Neumann et al., 2013; Van Vlierberghe et al.,

2011; Zhang et al., 2012).

Whole exome and whole genome sequencing

approaches identified mutations in multiple

pathways including mutations in genes activating

cytokine receptor and RAS signalling ( NRAS ,

KRAS, FLT3, IL7R, JAK3, JAK1, SH2B3 and

BRAF), inactivating haematopoietic transcription

factors ( GATA3, ETV6, RUNX1, IKZF1 and

EP300) and in epigenetic regulators (EZH2, EED,

SUZ12, SETD2 and EP300) (Zhang et al., 2012)

References Allen A, Sireci A, Colovai A, Pinkney K, Sulis M, Bhagat G, Alobeid B. Early T-cell precursor leukemia/lymphoma in adults and children. Leuk Res. 2013 Sep;37(9):1027-34

Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016 May 19;127(20):2391-405

Bell JJ, Bhandoola A. The earliest thymic progenitors for T cells possess myeloid lineage potential. Nature. 2008 Apr 10;452(7188):764-7

Brent L. Wood, Stuart S. Winter, Kimberly P. Dunsmore, Meenakshi Devidas, Si Chen, Barbara Asselin, Natia Esiashvili, Mignon L. Loh, Naomi J. Winick, William L. Carroll, Elizabeth A. Raetz and Stephen P. Hunger. T-Lymphoblastic Leukemia (T-ALL) Shows Excellent Outcome, Lack of Significance of the Early Thymic Precursor (ETP) Immunophenotype, and Validation of the Prognostic Value of End-Induction Minimal Residual Disease (MRD) in Children's Oncology Group (COG) Study AALL0434 Blood 2014 124:1

Chopra A, Bakhshi S, Pramanik SK, Pandey RM, Singh S, Gajendra S, Gogia A, Chandramohan J, Sharma A, Kumar L, Seth R, Rai S, Kumar R. Immunophenotypic analysis of T-acute lymphoblastic leukemia A CD5-based ETP-ALL perspective of non-ETP T-ALL Eur J Haematol

Coustan-Smith E, Mullighan CG, Onciu M, Behm FG, Raimondi SC, Pei D, Cheng C, Su X, Rubnitz JE, Basso G, Biondi A, Pui CH, Downing JR, Campana D. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia Lancet Oncol 2009 Feb;10(2):147-56

Haydu JE, Ferrando AA. Early T-cell precursor acute lymphoblastic leukaemia Curr Opin Hematol 2013 Jul;20(4):369-73

Inukai T, Kiyokawa N, Campana D, Coustan-Smith E, Kikuchi A, Kobayashi M, Takahashi H, Koh K, Manabe A, Kumagai M, Ikuta K, Hayashi Y, Tsuchida M, Sugita K, Ohara A. Clinical significance of early T-cell precursor acute lymphoblastic leukaemia: results of the Tokyo Children's Cancer Study Group Study L99-15 Br J Haematol 2012 Feb;156(3):358-65

Jain N, Lamb AV, O'Brien S, Ravandi F, Konopleva M, Jabbour E, Zuo Z, Jorgensen J, Lin P, Pierce S, Thomas D, Rytting M, Borthakur G, Kadia T, Cortes J, Kantarjian HM, Khoury JD. Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype Blood 2016 Apr 14;127(15):1863-9

Ma M, Wang X, Tang J, Xue H, Chen J, Pan C, Jiang H, Shen S. Early T-cell precursor leukemia: a subtype of high risk childhood acute lymphoblastic leukemia Front Med 2012 Dec;6(4):416-20

Neumann M, Coskun E, Fransecky L, Mochmann LH, Bartram I, Sartangi NF, Heesch S, Gökbuget N, Schwartz S, Brandts C, Schlee C, Haas R, Dührsen U, Griesshammer M, Döhner H, Ehninger G, Burmeister T, Blau O, Thiel E, Hoelzer D, Hofmann WK, Baldus CD. FLT3 mutations in early T-cell precursor ALL characterize a stem cell like leukemia and imply the clinical use of tyrosine kinase inhibitors PLoS One 2013;8(1):e53190

Neumann M, Heesch S, Schlee C, Schwartz S, Gökbuget N, Hoelzer D, Konstandin NP, Ksienzyk B, Vosberg S, Graf A, Krebs S, Blum H, Raff T, Brüggemann M, Hofmann WK, Hecht J, Bohlander SK, Greif PA, Baldus CD. Whole-exome sequencing in adult ETP-ALL reveals a high rate of DNMT3A mutations Blood 2013 Jun 6;121(23):4749-52

Patrick K, Wade R, Goulden N, Mitchell C, Moorman AV, Rowntree C, Jenkinson S, Hough R, Vora A. Outcome for children and young people with Early T-cell precursor acute lymphoblastic leukaemia treated on a contemporary protocol, UKALL 2003 Br J Haematol 2014 Aug;166(3):421-4

Van Vlierberghe P, Ambesi-Impiombato A, Perez-Garcia A, Haydu JE, Rigo I, Hadler M, Tosello V, Della Gatta G, Paietta E, Racevskis J, Wiernik PH, Luger SM, Rowe JM, Rue M, Ferrando AA. ETV6 mutations in early immature human T cell leukemias J Exp Med 2011 Dec 19;208(13):2571-9

Wada H, Masuda K, Satoh R, Kakugawa K, Ikawa T, Katsura Y, Kawamoto H. Adult T-cell progenitors retain myeloid potential Nature 2008 Apr 10;452(7188):768-72

Zhang J, Ding L, Holmfeldt L, Wu G, Heatley SL, Payne-Turner D, Easton J, Chen X, Wang J, Rusch M, Lu C, Chen SC, Wei L, Collins-Underwood JR, Ma J, Roberts KG, Pounds SB, Ulyanov A, Becksfort J, Gupta P, Huether R, Kriwacki RW, Parker M, McGoldrick DJ, Zhao D, Alford

Page 17: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Early T-cell precursor acute lymphoblastic leukemia Richebourg S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 450

D, Espy S, Bobba KC, Song G, Pei D, Cheng C, Roberts S, Barbato MI, Campana D, Coustan-Smith E, Shurtleff SA, Raimondi SC, Kleppe M, Cools J, Shimano KA, Hermiston ML, Doulatov S, Eppert K, Laurenti E, Notta F, Dick JE, Basso G, Hunger SP, Loh ML, Devidas M, Wood B, Winter S, Dunsmore KP, Fulton RS, Fulton LL, Hong X, Harris CC, Dooling DJ, Ochoa K, Johnson KJ, Obenauer JC, Evans WE, Pui CH, Naeve CW, Ley TJ, Mardis ER, Wilson RK, Downing JR, Mullighan CG. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia Nature 2012 Jan 11;481(7380):157-63

Zuurbier L, Gutierrez A, Mullighan CG, Canté-Barrett K,

Gevaert AO, de Rooi J, Li Y, Smits WK, Buijs-Gladdines JG, Sonneveld E, Look AT, Horstmann M, Pieters R, Meijerink JP. Immature MEF2C-dysregulated T-cell leukemia patients have an early T-cell precursor acute lymphoblastic leukemia gene signature and typically have non-rearranged T-cell receptors Haematologica 2014 Jan;99(1):94-102

This article should be referenced as such:

Richebourg S. Early T-cell precursor acute lymphoblastic leukemia. Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):447-450.

Page 18: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Leukaemia Section Review

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 451

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

Multiple Myeloma Matthew Ho Zhi Guang, Kenneth C. Anderson, Giada Bianchi

LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Department

of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115.

[email protected]; [email protected];

[email protected]

Published in Atlas Database: January 2017

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/MultipleMyelomaID1776.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69009/01-2017-MultipleMyelomaID1776.pdf DOI: 10.4267/2042/69009

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Multiple Myeloma (MM) is a cancer of plasma

cells resulting from the abnormal proliferation of

malignant plasma cells within the bone marrow

(BM) microenvironment. MM accounts for 1.3% of

all malignancies and 12% of hematologic cancers,

and is the second most commonly diagnosed blood

cancer after non-Hodgkin lymphoma. The hallmark

characteristics of MM include: high levels of intact

monoclonal immunoglobulin or its fragment (free

light chain) in serum or urine, and excess

monotypic plasma cells in the bone marrow in

conjunction with evidence of end organ damage

related to MM: (1) hypercalcemia, (2) renal failure,

(3) anemia, and (4) osteolytic bone lesions or severe

osteopenia, known as CRAB criteria. Even though

novel agents targeting MM cells in the context of

the BM microenvironment such as proteasome

inhibitors, immunomodulatory drugs (IMiDs), and

monoclonal antibodies have significantly prolonged

survival in MM patients, the disease remains

incurable. A deeper understanding of the molecular

mechanisms of MM growth, survival, and

resistance to therapy, such as genomic instability,

clonal heterogeneity and evolution, as well as MM-

BM microenvironmental host immune and other

factors, will provide the framework for

development of novel therapies to further improve

patient outcome.

Keywords

multiple myeloma, bone marrow

microenvironment, monoclonal gammopathy of

undetermined significance

Identity

Other names

Plasma cell myeloma, Myelomatosis, Kahler's

disease

Note

This paper is an update of Multiple myeloma in

2004.

Clinics and pathology

Disease

MM is a plasma cell cancer which is preceded by

an asymptomatic, premalignant condition called

monoclonal gammopathy of undetermined

significance (MGUS) which then progresses to MM

or related malignancies with a rate of about 1% per

year (Zingone and Kuehl 2011).

Phenotype/cell stem origin

Antigen-selected, post-germinal center, terminally

differentiated plasma cell (Anderson and Carrasco

2011)

Page 19: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 452

Top: Normal Bone Marrow; Bottom: Multiple Myeloma Bone Marrow (note: ≥ 10% clonal bone marrow plasma cells). Image taken from: http://www.thrombocyte.com/causes-of-multiple-myeloma-cancer/

Etiology

Etiology not known. No confirmed predisposing

factors.

Possible (unconfirmed and controversial) risk

factors include (Sundar Jagannath et al 2016):

Environmental factors such as radiation exposure,

occupational exposure (agricultural, chemical,

metallurgical, rubber plant, pulp, wood, paper), and

chemical exposure (formaldehyde, epichlorohydrin,

Agent orange, hair dyes, paint sprays, asbestos)

Viral infection: Herpesvirus 8 infection noted in

some patients with MM

Genetic predisposition

The transformation of normal plasma cells into

myeloma cells is thought to result from one of two

primary genetic events: either (1) hyperdiploidy or

(2) aberrant class switch recombination (CSR),

likely occurring in the germinal center, leading to

MGUS. Secondary cytogenetic abnormalities result

in the progression of MGUS to SMM, MM, and

plasma cell leukemia (PCL) (see below:

Cytogenetics). MM cells are dependent upon the

BM microenvironment for growth, survival, and

drug resistance, due both to tumor cell adhesion to

BM accessory cells and release of growth factors

and cytokines including (1) interleukin-6 ( IL6), (2)

vascular endothelial growth factor ( VEGFA), (3)

insulin-like growth factor 1 ( IGF1), (4) members

of the superfamily of tumor necrosis factor, (5)

transforming growth factor beta1 (TGFB1), and (6)

interleukin-10 ( IL10) (Palumbo and Anderson

2011). Coupled with various genetic changes, these

abnormal microenvironmental interactions between

MM cells and BM cells contribute to aberrant

angiogenesis and MM disease progression

(Palumbo and Anderson 2011).

Epidemiology

See table below.

Clinics

The most common presenting symptoms of MM are

fatigue and bone or back pain. Multiple myeloma

cells typically grow within the BM of the spine,

skull, ribs, sternum, pelvis, humeri, and femora,

causing pain, osteopenia, and frequently

pathological fractures (Palumbo and Anderson

2011).

Myeloma cells typically secrete an excess of a

monoclonal immunoglobulin or its fragments (free

light chain), which can then be detected in the

patient's serum and/or urine via protein

electrophoresis and serum free light chain (sFLC)

testing, respectively. Immunofixation shows the

myeloma (M) protein to be monoclonal in nature

and identifies heavy (IgG/IgA/IgM/IgD, in order of

frequency) and light chain ( κ/λ) specific isotype.

Page 20: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 453

Rarely, MM may be non-secretory and neither a

monoclonal Ig nor an excess sFLC can be

identified. The diagnosis of MM is made based on

the percentage of bone marrow involvement by

clonal MM cells, size of M protein spike, and

presence/absence of end-organ damage (CRAB) or

myeloma-defining biomarkers (Rajkumar,

Dimopoulos et al. 2014).

CRAB criteria:

Hypercalemia -> (C) Up to 20% of newly diagnosed patients have

hypercalcemia due to bone destruction.

Hypercalcemia is associated with high tumor

burden and requires prompt treatment with

aggressive hydration and loop diuretic therapy,

bisphosphonates, calcitonin, and anti-myeloma

therapy for disease control.

Renal Failure -> (R) Renal dysfunction (anuria or oliguria) resulting

from direct tubular damage by free light chain

tubular or glomerular deposition, hypercalcemia,

dehydration, and nephrotoxic medications (NSAIDs

for pain control, IV radiographic contrast,

bisphosphonates) is present in 20 to 40% of newly

diagnosed patients.

Light-chain cast nephropathy is the most common

cause of renal failure in MM. Other causes include

amyloidosis and light-chain deposition disease.

Anemia-> (A) At diagnosis, symptomatic normocytic,

normochromic anemia (typically secondary to

myelophthisis and hyporegenerative BM) is present

in approximately 73% of patients. Mean

corpuscular volume (MCV) may be macrocytic; an

artifact related to rouleaux formation.

Bone Disease -> (B) Up to 58% of patients report bone pain (especially

from compression fractures of vertebrae or ribs),

and up to 80% of newly diagnosed patients have

bony lesions.

The characteristic "punched-out" osteolytic lesions

result from lytic bone destruction that is uncoupled

from reactive bone formation. MM cells increase

the activity of osteoclasts by upregulating osteoclast

inducers (i.e. TNFSF11 (RANKL, TRANCE),

CCL3 and CCL4 (MIP-1 alpha /beta), CXCL12

(SDF-1 alpha), IL1B (IL-1 beta), TNF (TNF-

alpha), IL6,) and downregulating TNFRSF11B

(OPG, decoy receptor for RANKL).

Simultaneously, MM cells suppress osteoblast

differentiation and function (by producing DKK1)

resulting in an imbalance favouring bone resorption

(osteoclast activation) over bone formation

(osteoblast suppression) (Sezer 2009).

Revised International Staging System (R-ISS) (Palumbo, Avet-Loiseau et al. 2015)*Standard-risk: No high-risk chromosomal abnormality. High-risk: Presence of del(17p) and/or translocation t(4;14) and/or translocation t(14;16)

Page 21: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 454

Page 22: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 455

International Myeloma Working Group (IMWG) diagnostic criteria (Rajkumar, Dimopoulos et al. 2014)

Page 23: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 456

M-protein Left: Serum protein electrophoresis showing characteristic "M-protein" spike. Image taken from:

http://bestpractice.bmj.com/best-practice/images/bp/en-gb/179-5-iline_default.gif and http://www.aafp.org/afp/1999/0401/p1885.html; Right: Urine protein electrophoresis showing gamma-globulin peak

corresponding to Bence-Jones proteinuria. Image taken from: https://ahdc.vet.cornell.edu/sects/clinpath/test/immun/electro.cfm

Pathology

MM is characterized by the presence of ≥10%

malignant plasma cells in the bone marrow. MM

can be divided into (1/>= secretory MM, (2)

oligosecretory MM (aka light chain MM), and (3)

non-secretory MM based on whether M-protein is

secreted and detectable (Lonial and Kaufman

2013). Non-secretory MM accounts for <5% of

cases and can be further divided into producer (i.e.

patients who have detectable M-protein within MM

cells but do not secrete M-protein) and non-

producer MM (patients who do not have detectable

M-protein even within MM cells) (Lonial and

Kaufman 2013). The presence of Bence-Jones

protein (BJP) in the urine indicates the excessive

production of monoclonal light-chain proteins that

exceeds the re-absorptive ability of the proximal

tubules. These filtered light-chains are, in their

various forms (free, tubular casts, amyloid),

nephrotoxic and are responsible for the most

common cause of renal failure in patients with MM.

Another hallmark feature of MM is the presence of

osteolytic bone lesions that results from an

imbalance favoring bone resorption over bone

formation due to increased osteoclast activity and

reduced osteoblast differentiation and function,

secondary to secreted factors from MM cells (Sezer

2009). Associated with bone destruction are

complications such as bone pain, pathological

fractures, and hypercalcemia. Anemia is another

frequent finding in patients with MM that results

from multiple mechanisms including anemia of

chronic disease, EPO deficiency (secondary to renal

impairment), myelosuppression from

chemotherapy, and bone marrow infiltration by

plasma cells.

Osteolytic bone lesions (a-d) X-rays showing characteristic osteolytic bone lesions typical sites such as the (a) skull, (b) tibia, (c) femur, and (d) pelvis. Image taken from: http://orthoinfo.aaos.org/topic.cfm?topic=A00086 ; (e) Sagittal CT showing multiple

osteolytic bone lesions of the vertebral column. Image taken from: https://radiopaedia.org/cases/multiple-myeloma-skeletal-survey

Page 24: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 457

MM kidney disease Left: Normal kidney biopsy; Right: Monoclonal protein-containing casts surrounded by histiocytes and giant cells. Note the presence of acute tubular injury and interstitial nephritis which are commonly seen in MM kidney disease. Images

taken from: https://ajkdblog.org/2012/06/14/test-your-knowledge-myeloma-and-the-kidney/#prettyPhoto (courtesy of Dr. Tibor Nadasdy)

Natural History of MM Monoclonal Gammopathy of Undetermined Significance (MGUS; premalignant; asymptomatic) -> Smoldering Multiple Myeloma (SMM; pre-malignant; asymptomatic) -> Multiple Myeloma (MM; malignant; symptomatic) -> Plasma cell Leukemia (PCL), extramedullary disease. MM remains incurable in the long-term as most patients inevitably, yet

unpredictably, develop refractory relapse disease (i.e. disease that fails to respond to induction or salvage therapy, or progresses within 60 days of last therapy). Images taken from: Kyle et al, NEJM, Volume 356:2582-2590 (Kyle, Remstein et al. 2007) and

Roman Hajek, Intech open, DOI: 10.5772/55366 (Hajek 2013)

Page 25: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 458

Page 26: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 459

Treatment

See tables

(NCCN guidelines version 3.2017) and IMWG

RESPONSE CRITERIA (Kumar, Paiva et al. 2016)

Prognosis

Varies greatly depending on:

-Stage of disease (see above: ISS)

-Cytogenetics (see below: cytogenetics)

-LDH levels (high levels associated with

extramedullary disease, plasma cell leukemia,

plasmablastic disease, plasma cell hypoploidy, drug

resistance, and poor outcomes)

-Plasma cell labeling index

-C-reactive protein (high levels associated with

poor outcomes)

-Plasmablastic histology

-Extramedullary disease

-Age

Type of treatment available

- Conventional therapy: OS ~3 years; EFS <2 years

- High-dose chemotherapy and stem-cell

transplantation: 5-year OS >50%

In general, poor prognosticators include:

-Large tumor burden

- Hypercalcemia

- High LDH

- Bence-Jones proteinuria

- Renal impairment

- IgA subtype

- Extramedullary disease at presentation

Genetics See figure below.

Cytogenetics

Cytogenetics morphological

See figures below.

Genes involved and proteins

FGFR3 (Fibroblast Growth Factor Receptor 3)

Location

4p16.3

Note

Involved in t(4;14)(p16;q32)

Both FGFR3 and WHSC1 (MMSET) are

implicated in the translocation with IGH

Incidence: 6-12%

NSD2 (MMSET)

Location

4p16.3

Note

Involved in t(4;14)(p16;q32)

CCND1 (B-cell leukemia/lymphoma 1)

Location

11q13.3

Note

Involved in t(11;14)(q13;q32)

Incidence: 15-20%

Page 27: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 460

Genetics

Page 28: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 461

Primary Cytogenetic Abnormalities (Normal plasma cell --> MGUS/SMM). Ref : Rajan and Rajkumar (2015)

Page 29: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 462

Secondary Cytogenetic Abnormalities (MGUS/SMM --> MM --> RR MM, PCL) Ref : Morgan, Walker et al. (2012).

CCND3 (cyclin D3)

Location

6p21.1

Note

Involved in t(6;14)(p21;q32)

Incidence: 5%

MAF (v-maf musculoaponeurotic fibrosarcoma oncogene homolog (avian))

Location

16q23.2

Note

Involved in t(14;16)(q32;q23)

Incidence: 4-10%

IRF4 (interferon regulatory factor 4)

Location

6p25.3

Note

Involved in t(6;14)(p25;q32)

Incidence: 5%

MYC v-myc myelocytomatosis viral oncogene homolog (avian)

Location

8q24.21

Note

Involved in t(8;14)(q24;q32)

Incidence: <10%

MAFB (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog B)

Location

20q12

Note

Involved in t(14;20)(q32;q11)

Incidence: 1 - 5%

BCL9 (B-cell CLL/lymphoma 9)

Location

1q21.2

Note

Incidence: Frequent

Both BCL9, IL6R, and MCL1 can be deleted

Page 30: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Multiple Myeloma Ho M, et al.

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 463

IL6R (interleukin 6 receptor)

Location

1q21.3

Note

Incidence: Frequent

MCL1 (MCL1, BCL2 family apoptosis regulator)

Location

1q21.2

Note

Incidence: Frequent

References Anderson KC, Carrasco RD. Pathogenesis of myeloma. Annu Rev Pathol. 2011;6:249-74

Becker N. Epidemiology of multiple myeloma. Recent Results Cancer Res. 2011;183:25-35

Hajek, R.. Multiple Myeloma - A Quick Reflection on the Fast Progress

Huang SY, Yao M, Tang JL, Lee WC, Tsay W, Cheng AL, Wang CH, Chen YC, Shen MC, Tien HF. Epidemiology of multiple myeloma in Taiwan: increasing incidence for the

past 25 years and higher prevalence of extramedullary myeloma in patients younger than 55 years Cancer 2007 Aug 15;110(4):896-905

Kyle RA, Remstein ED, Therneau TM, Dispenzieri A, Kurtin PJ, Hodnefield JM, Larson DR, Plevak MF, Jelinek DF, Fonseca R, Melton LJ 3rd, Rajkumar SV. Clinical course and prognosis of smoldering (asymptomatic) multiple myeloma N Engl J Med 2007 Jun 21;356(25):2582-90

Lonial S, Kaufman JL. Non-secretory myeloma: a clinician's guide Oncology (Williston Park) 2013

Sep;27(9):924-8, 930

Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma Nat Rev Cancer 2012 Apr 12;12(5):335-48

Palumbo A, Avet-Loiseau H, Oliva S, Lokhorst HM, Goldschmidt H, Rosinol L, Richardson P, Caltagirone S, Lahuerta JJ, Facon T, Bringhen S, Gay F, Attal M, Passera R, Spencer A, Offidani M, Kumar S, Musto P, Lonial S, Petrucci MT, Orlowski RZ, Zamagni E, Morgan G, Dimopoulos MA, Durie BG, Anderson KC, Sonneveld P, San Miguel J, Cavo M, Rajkumar SV, Moreau P. Revised International Staging System for Multiple Myeloma: A Report From International Myeloma Working Group J Clin

Oncol 2015 Sep 10;33(26):2863-9

Rajan AM, Rajkumar SV. Interpretation of cytogenetic results in multiple myeloma for clinical practice Blood Cancer J 2015 Oct 30;5:e365

Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos MV, Kumar S, Hillengass J, Kastritis E, Richardson P, Landgren O, Paiva B, Dispenzieri A, Weiss B, LeLeu X, Zweegman S, Lonial S, Rosinol L, Zamagni E, Jagannath S, Sezer O, Kristinsson SY, Caers J, Usmani SZ, Lahuerta JJ, Johnsen HE, Beksac M, Cavo M, Goldschmidt H, Terpos E, Kyle RA, Anderson KC, Durie BG, Miguel JF. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma Lancet Oncol 2014 Nov;15(12):e538-48

Sezer O. Myeloma bone disease: recent advances in biology, diagnosis, and treatment Oncologist 2009 Mar;14(3):276-83

Sundar Jagannath, Paul G. Richardson, Nikhil C. Munshi. Multiple Myeloma and Other Plasma Cell Dyscrasias Cancer Network

This article should be referenced as such:

Ho M, Anderson KC, Bianchi G. Multiple Myeloma. Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):451-463.

Page 31: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 464

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

t(5;17)(p11;q11) and t(5;17)(q11-12;q11-12) Adriana Zamecnikova, Soad al Bahar

Kuwait Cancer Control Center, Department of Hematology [email protected]

Published in Atlas Database: January 2017

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t0517p11q11ID1772.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69010/01-2017-t0517p11q11ID1772.pdf DOI: 10.4267/2042/69010

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Review on t(5;17)(p11;q11) and t(5;17)(q11-

12;q11-12), with data on clinics.

Keywords

Chromosome 5; chromosome 17; Acute myeloid

leukaemia

Clinics and pathology

Disease

Acute myeloid leukemia (AML)

Epidemiology

Described in 2 male and 3 female patients aged 28

to 70 years; Acute myeloblastic leukemia with

maturation (FAB type M2) in 3 (Huebner et al.,

2000; Paietta et al., 1988; Arnaud et al., 2005) and

AML-NOS in 2 patients (Suciu et al., 1993;

Kerndrup and Kjeldsen., 2001) (Table 1).

Prognosis

May represent an unfavorable cytogenetic

prognostic category in association with monosomy

7 and/or complex karyotypes.

Figure 1. (A) Partial karyotypes showing the t(5;17)(p11;q11). (B) Fluorescence in situ hybridization with LSI TP53/CEP17 probe (Vysis, Abott Moleculars, US) showing the chromosome 17 centromere (green) and the p53 gene (red) on der(17) chromosome containing the short arms of chromosomes 5 and 17. (C) Hybridization with LSI CSF1R/D5S23/D5S721 hybridizing on 5p13.2 (green) and 5q33 (red) showing the signal for 5p (green) on der(17) and the signal for 5q33 (red) on der(5) chromosome. (D)

Simultaneous hybridization with LSI TP53/CEP17 and LSI CSF1R/D5S23/D5S721 probes demonstrating the presence of TP53/CEP17 and 5p13 signals on der(17) and the 5q33 signal on der(5) chromosome containing the long arm of chromosome

17.

Page 32: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

t(5;17)(p11;q11) and t(5;17)(q11-12;q11-12) Zamecnikova A, al Bahar S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 465

Table 1. Reported patients with t(5;17)(p11-12-q11;q11-12). Abbreviations: M, male; F, female; AML-M2, Acute myeloblastic leukemia with maturation (FAB type M2), AML, Acute myeloid leukemia, NOS. 1. Huebner et al., 2000; 2. Paietta et al., 1988; 3.

Suciu et al., 1993; 4. Kerndrup & Kjeldsen., 2001; 5. Arnaud et al., 2005.

Cytogenetics

Additional anomalies

Complex chromosome rearrangements in all the 5

described patients, found in association with

monosomy 7 in 2 (Huebner et al., 2000; Arnaud et

al., 2005) and trisomy 8 in 3 patients (Paietta et al.,

1988; Suciu et al., 1993; Kerndrup and Kjeldsen.,

2001).

Result of the chromosomal anomaly

Fusion protein

Oncogenesis

The reciprocal, apparently balanced t(5;17)(p11-

q11;q11) is a rare but non-random anomaly in

acute myeloid leukemia, that may be particularly

associated with acute myeloblastic leukemia with

maturation (AML-M2). The key mechanism of

oncogenesis is unknown; however as it presents in

association with known anomalies such as

monosomy 7 or trisomy 8 in all the described cases,

it is likely that it represents a secondary anomaly

that developed during the multistep process of

leukemogenesis.

References Huebner G, Karthaus M, Pethig K, Freund M, Ganser A. Myelodysplastic syndrome and acute myelogenous leukemia secondary to heart transplantation. Transplantation. 2000 Aug 27;70(4):688-90

Kerndrup GB, Kjeldsen E. Acute leukemia cytogenetics: an evaluation of combining G-band karyotyping with multi-color spectral karyotyping. Cancer Genet Cytogenet. 2001 Jan 1;124(1):7-11

Paietta E, Papenhausen P, Gucalp R, Wiernik PH. Translocation t(12;19)(q13;q13.3). A new recurrent abnormality in acute nonlymphocytic leukemia with atypical erythropoiesis. Cancer Genet Cytogenet. 1988 Aug;34(1):19-23

Suciu S, Zeller W, Weh HJ, Hossfeld DK. Immunophenotype of mitotic cells with clonal chromosome abnormalities demonstrating multilineage involvement in acute myeloid leukemia. Cancer Genet Cytogenet. 1993 Oct 1;70(1):1-5

This article should be referenced as such:

Zamecnikova A, al Bahar S. t(5;17)(p11;q11) and t(5;17)(q11-12;q11-12). Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):464-465.

Page 33: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 466

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

t(5;17)(q35;q21) NPM1/RARA Adriana Zamecnikova, Soad al Bahar

Kuwait Cancer Control Center, Department of Hematology [email protected]

Published in Atlas Database: January 2017

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t517ID1081.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69011/01-2017-t517ID1081.pdf DOI:10.4267/2042/69011

This article is an update of : Viguié F. t(5;17)(q35;q21). Atlas Genet Cytogenet Oncol Haematol 2000;4(2)

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Acute promyelocytic leukemia (APL) is

characterized by distinct clinical and biological

features and by the reciprocal translocation

t(15;17)(q22;q21) in the majority of patients. The

translocation generates the fusion of the

promyelocytic leukemia (PML) gene to the gene for

retinoic acid receptor alpha (RARA) and these

patients are responsive to differentiation treatment

with all-trans retinoic acid (ATRA). Rare cases of

patients with a morphological diagnosis of APL

have variant chromosome translocations, which

fuse RARA gene with partner genes other than

PML, such as in the variant translocation

t(5;17)(q35;q21) that fuses the N-terminus of

nucleophosmin (NPM1) gene at 5q35 to the retinoic

acid receptor alpha at 17q21.

Keywords

Chromosome 5; chromosome 17; Acute myeloid

leukaemia; Acute promyelocytic leukemia; RARA;

NPM1

Clinics and pathology

Disease

Acute myeloid leukemia (AML)

Phenotype/cell stem origin

Acute promyelocytic leukemia (AML-M3

according to the FAB classification)

Etiology

Exceptional; only 7 cases with balanced

t(5;17)(q35;q12-21) translocation and the

underlying NPM1/RARA fusion have been

identified (5 males and 2 females, aged 2.5 to 52

years).

There were 2 adult males, aged 29 and 52 years and

4 patients were pediatric cases, among them 2 were

12-year-old males and 2 were 2.5 and 9-year-old

females (Table 1).

Clinics

Disseminated intravascular coagulation was present

at diagnosis in one case; remission obtained with

chemotherapy and/or ATRA; first relapse at 7 and 5

months in 2 cases (Corey et al., 1994 ; Hummel et

al., 1999). The 6-month-old boy described by

Otsubo et al., presented with cutaneous

mastocytosis and aleukemic leukemia cutis that

regressed without any therapy within 6 months.

Both adult patients also presented with myeloid

sarcoma, therefore it is likely that it may occur

frequently in NPM1/RARA associated APL (Nicci

et., 2005; Kikuma et al.,2015).

Cytology

Hypergranular and hypogranular bilobed

promyelocytes; absence of Auer rods; typical

microspeckeled pattern with anti-RARa antibodies;

terminal differentiation of blasts and promyelocytes

in vitro with ATRA.

Page 34: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

t(15;17)(q22;q21) NPM1/RARA Zamecnikova A, al Bahar S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 467

Table 1. Reported cases with t(5;17)(q35;q12-21) and confirmed NPM1-RARA fusion. Abbreviations: M, male; F, female; ATRA; all-trans-retinoic acid. 1. Corey et al., 1994 and Redner et al., 1996. 2. Hummel et al., 1999; 3. Grimwade et al., 2000; 4.

Xu et al., 2001; 5. Nicci et al., 2005; 6. Otsubo et al., 2012; Kikuma et al., 2015.

Prognosis

The 2.5 years-old child (Corey et al., 1994; Redner

et al., 1996) was treated with ATRA while in partial

remission and relapsed shortly after ATRA

cessation. One of the 12-years old males (Hummel

et al., 1999) received chemotherapy in induction

and consolidation and relapsed after 5 months;

remission was obtained with Ara-C and ATRA

therapy followed by allogenic BM transplantation,

but relapsed with therapy refractoriness. The second

12-years old male who presented with severe DIC

died of cerebral hemorrhage after 5 days of ATRA

treatment (Xu et al., 2001). The 9-year-old female

was treated with ATRA as a part of induction

therapy and was alive in first CR at 29 months

(Grimwade et al 2000). The last pediatric patient

presented with aleukemic leukemia cutis and

t(5;17)(q35;q12) NPM1/RARA fusion at the age of

6-month-old (Kanegane et al., 2009; Otsubo et al.,

2012). He showed no sign of leukemia without any

therapy after 12 months, except for the presence of

NPM1-RARA transcript in the bone marrow, but

developed APL at the age of 4 years with complete

remission to ATRA. Both adult patients received

ATRA as part of induction therapy, resulting in

cytogenetic but not a molecular remission in 1

patient who relapsed at 22 months after diagnosis

(Nicci et al., 2005), and in complete hematological

and molecular remission in the other case (Kikuma

et al., 2015).

From these data, the response to ATRA is difficult

to assess since it was not part of induction treatment

in some cases and due to the limited number of

patients. However, patients with NPM1/RARA

fusion appear to be sensitive to ATRA (Hummel et

al., 1999; Grimwade et al., 2000; Kikuma et al.,

2015) and cells bearing the t(5;17) terminally

differentiate in its response (Redner et al., 1996),

indicating that ATRA can be used to treat

NPM1/RARA-positive APL patients. It is also

possible that the presence of the additional/complex

karyotypic abnormalities may be related to the

prognosis in this group of patients.

Cytogenetics Additional anomalies

Sole anomaly in both adult patients (Nicci et al.,

2005; Kikuma et al., 2015) and associated with

additional anomalies in pediatric patients: del(12p)

in 1 (Xu et al., 2001), i(21)(q10) in 1 (Otsubo et al.,

2012) and complex anomalies in 2 cases (Hummel

et al., 1999; Grimwade et al., 2000).

Variants

Variant chromosome translocations, which fuse

RARA with 1 of the partner genes: PML

(promyelocytic leukemia protein) in

t(15;17)(q22;q21) that is found in the majority of

APL patients; ZBTB16 (zinc finger and BTB

domain containing 16, previously known as PLZF)

Page 35: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

t(15;17)(q22;q21) NPM1/RARA Zamecnikova A, al Bahar S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 468

in t(11;17)(q23;q21) (De Braekeleer et al., 2014);

NUMA1 (nuclear matrix-mitotic apparatus protein

1 gene) in t(11;17)(q13;q21) (Wells et al., 1997);

STAT5B (signal transducer and activator of

transcription 5 beta) in dup(17)(q21.3q23) (Chen et

al., 2012); PRKAR1A (protein kinase, cAMP-

dependent, regulatory, type I, alpha) in

t(17;17)(q21;q24)/del(17)(q21q24) (Catalano et al.,

2007); FIP1L1 (factor interacting with PAP 1-like

1) in t(4;17)(q12;q21) (Buijs et al., 2007); NABP1

(OBFC2A: oligonucleotide/oligosaccharide-binding

fold containing 2A) in der(2)t(2;17)(q32;q21) (Won

et al., 2013); TBL1XR1 (TBLR1, GenBank

KF589333) in a complex t(3;17)(q26;q21),

t(7;17)(q11.2;q21) (Chen et al., 2014); BCOR

(BCL6 corepressor gene) in t(X;17)(p11.4;q21)

(Ichikawa et l., 2015) and the recently described

new RARA partner IRF2BP2 (interferon regulatory

factor 2 binding protein 2) in t(1;17)(q42.3; q21)

(Yin et al., 2015).

Genes involved and proteins

NPM1 (nucleophosmin)

Location

5q35.1

Protein

Gene for the nucleolar phosphoprotein

nucleophosmin; would participate in ribosome

assembly.

RARA (Retinoic acid receptor, alpha)

Location

17q21.2

Protein

Gene for the retinoic acid receptor alpha. Ligand-

dependent transcription factor specifically involved

in hematopoietic cells differentiation and

maturation. Receptor for all-trans retinoic acid

(ATRA) and 9-cis RA. After linking with ATRA,

RARA binds with RXR (retinoid X receptor

protein) to the RARE domain (retinoic acid

response elements), a DNA sequence common to a

number of genes. The breakpoint lies within the

second intron of the gene, as in t(15;17) and

t(11;17) translocations.

Result of the chromosomal anomaly

Hybrid gene

Description

Two reciprocal fusion genes are generated: 5'-

NPM1 + 3'- RARA on der(5) and 5'-RARa + 3'-

NPM on der(17); both fusion genes are transcribed,

the crucial one is NPM1/RARA; two NPM1/RARA

chimeric cDNAs are generated, one short and one

long differing from 129 bp, with corresponding

transcripts of 2.3 and 2.4 kb (alternatively spliced

transcripts); in one case, only the short

NPM1/RARA isoform could be detected; the 5' end

of NPM1/RARA cDNAs contains the first 442 bp

of the NPM1 cDNA; the 3' end contains RARA

sequences of exon 3 through the 3' end of RARA; a

reciprocal RARA/NPM1 transcript is detected:

RARA exons 1 and 2 are fused to 3' NPM1

downstream bp 443.

Detection

Nested RT-PCR.

Fusion protein

Description

Two NPM1/RARA proteins, of 563 and 520 amino

acids, are encoded (MW 62 and 57 kDa);

NPM1/RARA fusion protein acts as a retinoic acid-

responsive transcriptional activator: increase of

activity in a concentration dependant manner.

References Brunel V, Lafage-Pochitaloff M, Alcalay M, Pelicci PG, Birg F. Variant and masked translocations in acute promyelocytic leukemia. Leuk Lymphoma. 1996 Jul;22(3-4):221-8

Buijs A, Bruin M. Fusion of FIP1L1 and RARA as a result of a novel t(4;17)(q12;q21) in a case of juvenile myelomonocytic leukemia. Leukemia. 2007 May;21(5):1104-8

Chen H, Pan J, Yao L, Wu L, Zhu J, Wang W, Liu C, Han Q, Du G, Cen J, Xue Y, Wu D, Sun M, Chen S. Acute promyelocytic leukemia with a STAT5b-RARα fusion transcript defined by array-CGH, FISH, and RT-PCR. Cancer Genet. 2012 Jun;205(6):327-31

Chen Y, Li S, Zhou C, Li C, Ru K, Rao Q, Xing H, Tian Z, Tang K, Mi Y, Wang B, Wang M, Wang J. TBLR1 fuses to retinoid acid receptor α in a variant t(3;17)(q26;q21) translocation of acute promyelocytic leukemia. Blood. 2014 Aug 7;124(6):936-45

Cheng GX, Zhu XH, Men XQ, Wang L, Huang QH, Jin XL, Xiong SM, Zhu J, Guo WM, Chen JQ, Xu SF, So E, Chan LC, Waxman S, Zelent A, Chen GQ, Dong S, Liu JX, Chen SJ. Distinct leukemia phenotypes in transgenic mice and different corepressor interactions generated by promyelocytic leukemia variant fusion genes PLZF-RARalpha and NPM-RARalpha. Proc Natl Acad Sci U S A. 1999 May 25;96(11):6318-23

Corey SJ, Locker J, Oliveri DR, Shekhter-Levin S, Redner RL, Penchansky L, Gollin SM. A non-classical translocation involving 17q12 (retinoic acid receptor alpha) in acute promyelocytic leukemia (APML) with atypical features. Leukemia. 1994 Aug;8(8):1350-3

De Braekeleer E, Douet-Guilbert N, De Braekeleer M. RARA fusion genes in acute promyelocytic leukemia: a review. Expert Rev Hematol. 2014 Jun;7(3):347-57

Grimwade D. The pathogenesis of acute promyelocytic leukaemia: evaluation of the role of molecular diagnosis and monitoring in the management of the disease. Br J

Page 36: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

t(15;17)(q22;q21) NPM1/RARA Zamecnikova A, al Bahar S

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 469

Haematol. 1999 Sep;106(3):591-613

Hummel JL, Wells RA, Dubé ID, Licht JD, Kamel-Reid S. Deregulation of NPM and PLZF in a variant t(5;17) case of acute promyelocytic leukemia. Oncogene. 1999 Jan 21;18(3):633-41

Ichikawa S, Ichikawa S, Ishikawa I, Takahashi T, Fujiwara T, Harigae H. Successful treatment of acute promyelocytic leukemia with a t(X;17)(p11.4;q21) and BCOR-RARA fusion gene. Cancer Genet. 2015 Apr;208(4):162-3

Kanegane H, Nomura K, Abe A, Makino T, Ishizawa S, Shimizu T, Naoe T, Miyawaki T. Spontaneous regression of aleukemic leukemia cutis harboring a NPM/RARA fusion gene in an infant with cutaneous mastocytosis. Int J Hematol. 2009 Jan;89(1):86-90

Kikuma T, Nakamachi Y, Noguchi Y, Okazaki Y, Shimomura D, Yakushijin K, Yamamoto K, Matsuoka H, Minami H, Itoh T, Kawano S. A new transcriptional variant and small azurophilic granules in an acute promyelocytic leukemia case with NPM1/RARA fusion gene. Int J Hematol. 2015 Dec;102(6):713-8

Otsubo K, Horie S, Nomura K, Miyawaki T, Abe A, Kanegane H. Acute promyelocytic leukemia following aleukemic leukemia cutis harboring NPM/RARA fusion gene. Pediatr Blood Cancer. 2012 Nov;59(5):959-60

Pandolfi PP. PML, PLZF and NPM genes in the molecular pathogenesis of acute promyelocytic leukemia. Haematologica. 1996 Sep-Oct;81(5):472-82

Redner RL, Corey SJ, Rush EA. Differentiation of t(5;17) variant acute promyelocytic leukemic blasts by all-trans retinoic acid. Leukemia. 1997 Jul;11(7):1014-6

Redner RL, Rush EA, Faas S, Rudert WA, Corey SJ. The t(5;17) variant of acute promyelocytic leukemia expresses a nucleophosmin-retinoic acid receptor fusion. Blood. 1996 Feb 1;87(3):882-6

Won D, Shin SY, Park CJ, Jang S, Chi HS, Lee KH, Lee JO, Seo EJ. OBFC2A/RARA: a novel fusion gene in variant acute promyelocytic leukemia. Blood. 2013 Feb 21;121(8):1432-5

Yin CC, Jain N, Mehrotra M, Zhagn J, Protopopov A, Zuo Z, Pemmaraju N, DiNardo C, Hirsch-Ginsberg C, Wang SA, Medeiros LJ, Chin L, Patel KP, Ravandi F, Futreal A, Bueso-Ramos CE. Identification of a novel fusion gene, IRF2BP2-RARA, in acute promyelocytic leukemia. J Natl Compr Canc Netw. 2015 Jan;13(1):19-22

This article should be referenced as such:

Zamecnikova A, al Bahar S. t(5;17)(q35;q21) NPM1/RARA. Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):466-469.

Page 37: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Solid Tumour Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 470

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

Nervous system: Astrocytoma with t(1;17)(p36;q21) SPOP/PRDM16 Jean-Loup Huret

Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers,

France. [email protected]

Published in Atlas Database: September 2016

Online updated version : http://AtlasGeneticsOncology.org/Tumors/t0117p36q21ID5447.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69012/09-2016-t0117p36q21ID5447.pdf DOI: 10.4267/2042/69012

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Review on t(1;17)(p36;q21) SPOP/PRDM16

translocation in astrocytic tumor, with data on the

genes involved.

Keywords

chromosome 1; chromosome 17; t(1;17)(p36;q21);

SPOP; PRDM16, Astrocytic tumor

Clinics and pathology

Disease

A t(1;17)(p36;q21) was found in a case of

astrocytoma grade I-II, but no further data is

available (Yoshihara et al 2015).

Clinics

Grade I astrocytomas are pilocytic astrocytoma and

subependymal giant cell astrocytoma, the latter

being the most common central nervous system

neoplasm in tuberous sclerosis; they are slow

growing tumors; they typically occur during the

first two decades of life. Grade II astrocytomas are

pilomyxoid astrocytoma occurring most often in

infants and very young children, diffuse

astrocytoma, seen at any age, but often between

ages 30-40, and pleomorphic xanthoastrocytoma,

typically developing in children and young adults.

Genes involved and proteins

PRDM16 (PR domain containing 16)

Location

1p36.32

DNA / RNA

11 splice variants

Protein

1276 amino acids and smaller proteins. Contains a

N-term PR domain; 7 Zinc fingers, a proline-rich

domain, and 3 Zinc fingers in the C-term. Binds

DNA.

Transcription activator; PRDM16 has an intrinsic

histone methyltransferase activity. PRDM16 forms

a transcriptional complex with CEBPB. PRDM16

plays a downstream regulatory role in mediating

TGFB signaling (Bjork et al., 2010).

PRDM16 induces brown fat determination and

differentiation. PRDM16 is expressed selectively in

the earliest stem and progenitor hematopoietic cells,

and is required for the maintenance of the

hematopoietic stem cell pool during development.

PRDM16 is also required for survival, cell-cycle

regulation and self-renewal in neural stem cells

(Chuikov et al., 2010; Kajimura et al., 2010; Aguilo

et al., 2011; Chi and Cohen, 2016).

SPOP (speckle type BTB/POZ protein)

Location

17q21.33

DNA / RNA

24 splice variants

Page 38: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Nervous system: Astrocytoma with t(1;17)(p36;q21) SPOP/PRDM16

Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 471

Protein

374 amino acids SPOP comprises a N-term MATH

domain (Meprin And TRAF Homology; substrate

recognition and binding), a BTB/POZ domain

(Bric-a-brac, Tramtrack and Broad complex/ Pox

virus and Zinc finger; connects to the CUL3 -

RBX1 (Cullin 3-RING box 1) scaffold protein), a

3-box domain (facilitating CUL3 binding and

resembling to F-/SOCS-boxes) and a C-terminal

nuclear localization sequence. SPOP is a E3

ubiquitin ligase adaptor protein that participates in

the degradation of various substrates. AR (androgen

receptor), DAXX, BMI1, BRMS1 and PDX1 are

targets of SPOP. SPOP is critically involved in

SETD2 (a tumor suppressor) stability. SPOP

enables homology-directed DNA repair of double

strand breaks, and mutant SPOP promotes genomic

rearrangements within chromosomes. SPOP

suppresses gastric tumorigenesis through inhibiting

hedgehog/ GLI2 signaling pathway. SPOP is

frequently mutated in prostate and endometrial

cancers. TMPRSS2 - ERG fusions, frequently seen

in prostate carcinoma, encode N-terminal-truncated

ERG proteins that are resistant to the SPOP-

mediated degradation. Decreased expression of

SPOP is associated with poor prognosis in glioma.

On the other hand, SPOP is highly expressed in

clear cell renal cell carcinoma (Zhuang et al., 2009;

Mani, 2014; Zeng et al., 2014; Karnes et al., 2015;

Ding et al., 2015; Rider and Cramer, 2015).

References Aguilo F, Avagyan S, Labar A, Sevilla A, Lee DF, Kumar P, Lemischka IR, Zhou BY, Snoeck HW. Prdm16 is a physiologic regulator of hematopoietic stem cells. Blood. 2011 May 12;117(19):5057-66

An J, Ren S, Murphy SJ, Dalangood S, Chang C, Pang X,

Cui Y, Wang L, Pan Y, Zhang X, Zhu Y, Wang C, Halling GC, Cheng L, Sukov WR, Karnes RJ, Vasmatzis G, Zhang Q, Zhang J, Cheville JC, Yan J, Sun Y, Huang H. Truncated ERG Oncoproteins from TMPRSS2-ERG

Fusions Are Resistant to SPOP-Mediated Proteasome Degradation. Mol Cell. 2015 Sep 17;59(6):904-16

Bjork BC, Turbe-Doan A, Prysak M, Herron BJ, Beier DR. Prdm16 is required for normal palatogenesis in mice. Hum Mol Genet. 2010 Mar 1;19(5):774-89

Chi J, Cohen P. The Multifaceted Roles of PRDM16: Adipose Biology and Beyond. Trends Endocrinol Metab. 2016 Jan;27(1):11-23

Chuikov S, Levi BP, Smith ML, Morrison SJ. Prdm16 promotes stem cell maintenance in multiple tissues, partly by regulating oxidative stress. Nat Cell Biol. 2010 Oct;12(10):999-1006

Ding D, Song T, Jun W, Tan Z, Fang J. Decreased expression of the SPOP gene is associated with poor prognosis in glioma. Int J Oncol. 2015 Jan;46(1):333-41

Kajimura S, Seale P, Kubota K, Lunsford E, Frangioni JV, Gygi SP, Spiegelman BM. Initiation of myoblast to brown fat switch by a PRDM16-C/EBP-beta transcriptional complex. Nature. 2009 Aug 27;460(7259):1154-8

Mani RS. The emerging role of speckle-type POZ protein (SPOP) in cancer development. Drug Discov Today. 2014 Sep;19(9):1498-502

Rider L, Cramer SD. SPOP the mutation. Elife. 2015 Oct 27;4

Yoshihara K, Wang Q, Torres-Garcia W, Zheng S, Vegesna R, Kim H, Verhaak RG. The landscape and therapeutic relevance of cancer-associated transcript fusions. Oncogene. 2015 Sep 10;34(37):4845-54

Zeng C, Wang Y, Lu Q, Chen J, Zhang J, Liu T, Lv N, Luo S. SPOP suppresses tumorigenesis by regulating Hedgehog/Gli2 signaling pathway in gastric cancer. J Exp Clin Cancer Res. 2014 Sep 11;33:75

Zhuang M, Calabrese MF, Liu J, Waddell MB, Nourse A, Hammel M, Miller DJ, Walden H, Duda DM, Seyedin SN, Hoggard T, Harper JW, White KP, Schulman BA. Structures of SPOP-substrate complexes: insights into molecular architectures of BTB-Cul3 ubiquitin ligases. Mol Cell. 2009 Oct 9;36(1):39-50

This article should be referenced as such:

Nervous system: Astrocytoma with t(1;17)(p36;q21) SPOP/PRDM16. Nervous system: Astrocytoma with t(1;17)(p36;q21) SPOP/PRDM16. Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):470-471.

Page 39: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Solid Tumour Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 472

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

Chromophobe renal cell carcinoma Paola Dal Cin, Michelle S. Hirsch

Department of Pathology, Brigham, Women's Hospital, 75 Francis Street, Boston, MA 02115, USA

Published in Atlas Database: November 2016

Online updated version : http://AtlasGeneticsOncology.org/Tumors/ChromophobeRenalID5124.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69013/11-2016-ChromophobeRenalID5124.pdf DOI: 10.4267/2042/69013

This article is an update of: Dal Cin P. Kidney: Chromophobe renal cell carcinoma. Atlas Genet Cytogenet Oncol Haematol 2002;6(1)

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Review on chromophobe renal cell carcinoma, with

data on clinics and cytogenetics.

Classification

Chromophobe renal cell carcinoma (ChRCC) is a

distinct subtype of renal cell carcinoma, possibly

originating from the the distal nephron.

Clinics and pathology

Epidemiology

ChRCC comprise ~5% of all renal cell carcinomas.

Most tumors are sporadic, with a slight male

predilection.

Pathology

ChRCC tumors can vary in size and have a tan to

brown cut surface. The growth pattern is often solid

with sheets of cells divided by vascular septae,

some of which may have perivascular hyalinized

stroma. ChRCC tumor cells have pale cytoplasm

and distinct cell membranes. Small eosinophilic

cells with granular appearance may be present.

Nuclei may appear atypical, but are usually small

with wrinkled nuclear membranes and

multinucleation (Fig1A). ChRCC grading is

complicated by the nuclear atypia, and Fuhrman

nuclear grading should not be used. Prognosis is

generally favorable with low grade, low stage

tumors; but increased cytologic atypia, increased

mitotic activity, necrosis, and vascular invasion are

poor prognostic indicators. Sarcomatoid

differentiation and high tumor stage are also

predictors of poor outcome.

Special stains and immunohistochemistry can be

used to distinguish ChRCC from other renal

epithelial neoplasms. The presence of Hale's

colloidal iron in the cytoplasm of tumor cells is

supportive of ChRCC (an apical staining pattern is

more supportive of oncocytoma) (Fig.1B).

Immunohistochemical expression of CK7 (patchy

to diffuse) combined with the absence of S100A1,

HNF1beta (nuclear), and CD10 is consistent with a

ChRCC.

Occasionally composite oncocytic tumors with

features of both oncocytoma and ChRCC tumors

have been described, and are most commonly seen

in patients with Birt-Hogg-Dube' syndrome and/or

oncocytosis.

Fig. 1A: ChRCC (H&E stain) is comprised of sheets of tumor cells with well-defined cell borders, round to

raisonoid nuclei, and perinuclear halos. Long linear, parallel vessels are common in ChRCC.

Page 40: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Chromophobe renal cell carcinoma Dal Cin P, Hirsch MS

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 473

Fig 1B: A Hale's colloidal iron stain is positive (blue) in the

cytoplasm of ChRCC tumor cells.

Cytogenetics

Cytogenetics Morphological

Chromophobe RCCs generally have a tendency to

grow very slowly in vitro in comparison to all other

type of renal tumors. This may be a reason why

cytogenetic reports are scarce and usually few

metaphases of poor quality were available for

investigation. A low chromosome number ranging

between 32-39, without discernible structural

changes was the most frequent cytogenetic finding.

Chromosomes 1, 2, 6, 10, 13, 17 and 21 were most

frequently lost (Fig.2). Endoreduplication of the

cells with hypodiploid karyotype has been

observed. It is of interest, the presence of an

hypodiploid clone can be disclosed by a DNA index

of 0.86. The low chromosome number has been

confirmed by other techniques such as flow

cytometry, comparative genomic hybridization

(CGH), restriction fragment length polymorphism

(RFLP) analysis, and polymorphic microsatellite

markers.

Genes involved and Proteins

Note

High resolution DNA-microarray analysis excluded

the occurrence of small specific alteration

confirming that this combination of monosomies

occurs exclusively in chromophobe subtype of

RCC. The most commonly mutated genes in

chRCC are TP53 and PTEN, combined with

chromosome 17 and 10 deletions (Haake et al

2016).

Whole genomes sequencing identified a number of

genomic rearrangements in the TERT promoter

region, these same tumors displayed elevated TERT

gene expression, suggesting a functional role for

these gene fusions.

TERT (telomerase reverse transcriptase)

Location 5p15.33

Note Structural rearrangements in the TERT promoter

region and TERT upregulation are found in a subset

of chromophobe RCCs.

Protein Telomerase encodes a catalytic subunit of the

telomerase enzyme, which functions to maintain

telomere ends. Telomerase is upregulated in a

variety of tumors and plays a role tumor cell

immortalization.

PTEN (Phosphatase and Tensin homolog deleted on chromosome Ten) Location 10q23.31

Note PTEN mutations are found in approximately 20%

of chromophobe RCCs1.

Protein PTEN mutations frequently co-occur with loss of

chromosome 10 resulting in complete loss of

function. PTEN is a tumor suppressor that functions

as a protein and lipid phosphatase and negatively

regulates the PI3K-AKT/PKB signaling pathway.

TP53 (Tumour protein p53 (Li-Fraumeni syndrome)) Location 17p13.1

Note Approximately 32% of chromophobe RCCs have a

TP53 mutation.

Protein TP53 encodes a tumor suppressor protein, which

plays a regulatory role in many cellular processes

including DNA repair, growth arrest, apoptosis,

senescence and metabolism. Mutations in TP53 are

found in a broad range of tumor types.

Page 41: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Chromophobe renal cell carcinoma Dal Cin P, Hirsch MS

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 474

Fig.2. GTG-banded karyotype showing combination of monosomies 1, 2, 6, 8, 10, 13, 15, 17 and 22, and loss of the Y-chromosome

References Eble J, Delahunt B, Moch H, Martignoni G, Amin MB, Srigley JR, Argani P, Tan PH, Cheville J, Tickoo SK. Papillary adenoma. In: World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of the Urinary System and Male Genital Organs. Moch, H., Humphrey, P.A., Ulbright, T.M., Reuter, V.E. eds; IARC Press, Lyon 2016, 42-43.

Moch, H., Humphrey, P.A., Ulbright, T.M., Reuter, V.E.World Health Organization Classification of Tumours of the Urinary Systems and Male Genital Organs eds (4th edition); IARC Press, Lyon 2016; 11-76.

ICGC Breast Cancer Consortium. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Børresen-Dale AL, Boyault S, Burkhardt B, Butler AP, Caldas C, Davies HR, Desmedt C, Eils R, Eyfjörd JE, Foekens JA, Greaves M, Hosoda F, Hutter B, Ilicic T, Imbeaud S, Imielinski M, Jäger N, Jones DT, Jones D, Knappskog S, Kool M, Lakhani SR, López-Otín C, Martin S, Munshi NC, Nakamura H, Northcott PA, Pajic M, Papaemmanuil E, Paradiso A, Pearson JV, Puente XS, Raine K, Ramakrishna M, Richardson AL, Richter J, Rosenstiel P, Schlesner M, Schumacher TN, Span PN, Teague JW, Totoki Y, Tutt AN, Valdés-Mas R, van Buuren MM, van 't Veer L, Vincent-Salomon A, Waddell N, Yates LR; Australian Pancreatic Cancer Genome Initiative; ICGC MMML-Seq Consortium ; ICGC PedBrain

Haake SM, Weyandt JD, Rathmell WK. Insights into the Genetic Basis of the Renal Cell Carcinomas from The Cancer Genome Atlas. Mol Cancer Res 2016 Jul;14(7):589-98

Hirsch MS, Signoretti S, Dal Cin P. Adult Renal Cell Carcinoma: A Review of Established Entities from Morphology to Molecular Genetics. Surg Pathol Clin 2015 Dec;8(4):587-621

Iqbal MA, Akhtar M, Ulmer C, Al-Dayel F, Paterson MC. FISH analysis in chromophobe renal-cell carcinoma. Diagn

Cytopathol 2000 Jan;22(1):3-6

Kovacs A, Kovacs G. Low chromosome number in chromophobe renal cell carcinomas. Genes Chromosomes Cancer 1992 Apr;4(3):267-8

Kovacs G, Soudah B, Hoene E. Binucleated cells in a human renal cell carcinoma with 34 chromosomes. Cancer Genet Cytogenet 1988 Apr;31(2):211-5

Speicher MR, Schoell B, du Manoir S, Schröck E, Ried T, Cremer T, Störkel S, Kovacs A, Kovacs G. Specific loss of chromosomes 1, 2, 6, 10, 13, 17, and 21 in chromophobe renal cell carcinomas revealed by comparative genomic hybridization. Am J Pathol 1994 Aug;145(2):356-64

Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, Hes O, Moch H, Montironi R, Tickoo SK, Zhou M, Argani P; ISUP Renal Tumor Panel

The International Society of Urological Pathology (ISUP) Vancouver Classification of Renal Neoplasia Am J Surg Pathol

Störkel S, Steart PV, Drenckhahn D, Thoenes W. The human chromophobe cell renal carcinoma: its probable relation to intercalated cells of the collecting ductVirchows Arch B Cell Pathol Incl Mol Pathol 1989;56(4):237-45

Tan PH, Cheng L, Rioux-Leclercq N, Merino MJ, Netto G, Reuter VE, Shen SS, Grignon DJ, Montironi R, Egevad L, Srigley JR, Delahunt B, Moch H; ISUP Renal Tumor PanelRenal tumors: diagnostic and prognostic biomarkers Am J Surg Pathol

Tickoo SK, Amin MB, Zarbo R. Colloidal iron staining in renal epithelial neoplasms, including chromophobe renal cell carcinoma: emphasis on technique and patterns of staining. Am J Surg Pathol 1998 Apr;22(4):419-24

This article should be referenced as such:

Dal Cin P, Hirsch MS. Chromophobe renal cell carcinoma. Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):472-474.

Page 42: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Cancer Prone Disease Section Review

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 475

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS OPEN ACCESS JOURNAL

Ataxia telangiectasia (A-T) Yossi Shiloh

The David and Inez Myers Chair in Cancer Research, Department of Human Molecular Genetics and

Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;

[email protected]

Published in Atlas Database: October 2016

Online updated version : http://AtlasGeneticsOncology.org/Kprones/ataxiaID10003.html

Printable original version : http://documents.irevues.inist.fr/bitstream/handle/2042/69014/10-2016-ataxiaID10003.pdf DOI: 10.4267/2042/69014

This article is an update of : Huret, JL. Ataxia telangiectasia. Atlas Genet Cytogenet Oncol Haematol. 1998;2(4):153-154. Uhrhammer, N ; Bay, JO ; Gatti, RA. Ataxia telangiectasia. Atlas Genet Cytogenet Oncol Haematol. 1999;3(4):209-211. Uhrhammer, N ; Bay, JO ; Gatti, RA. Ataxia telangiectasia. Atlas Genet Cytogenet Oncol Haematol. 2003;7(1):52-54.

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2017 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abstract

Review on Ataxia telangiectasia, with data on

clinics, and the gene involved.

Keywords

Review on Ataxia telangiectasia, with data on

clinics, and the gene involved.

Identity

Other names

Louis-Bar syndrome

Note

See also, in Deep Insight section: Ataxia-

Telangiectasia and variants.

Inheritance

Autosomal recessive disease. Genome instability

syndrome found worldwide with incidence of .0.5.

to 2.5/105 newborns in different human populations.

A founder effect is found in some isolated

population. Heterozygotes are estimated to be 1%

of the general population. The clinical phenotype of

A-T ranges from severe to milder variants of the

disease, but is usually portrayed by its classical,

severe form (Perlman SL et al., 2012; Lavin MF,

2008; Crawford TO, 1998; Chun HH et al., 2004;

Nissenkorn A et al., 2016). However, awareness is

growing of the broad clinical variability associated

with the causative mutations (Taylor AM et al.,

2015).

The primary cause of all variants of the disease is

mutations in the autosomal gene ATM (A-T,

mutated) at 11q22-23 (Gatti RA et al., 1988;

Savitsky K et al., 1995a), which encodes the ATM

protein (Savitsky K et al., 1995b; Ziv Y et al.,

1997) a multi-functional protein kinase (Shiloh Y et

al., 2013; Shiloh Y, 2014; Guleria A et al., 2016;

Ditch S et al., 2012).

Clinics Ataxia telangiectasia is a chromosome instability

syndrome (Perlman SL et al., 2012; Lavin MF,

2008; Crawford TO, 1998; Chun HH et al., 2004;

Taylor AM et al., 1982; Taylor AM et al., 2015;

Taylor AM, 1978; Butterworth SV et al., 1986;

Kennaugh AA et al., 1986) with cerebellar

degeneration, immunodeficiency, and an increased

risk of cancers; A-T cells are defective in

recognizing double-strand DNA damage to signal

for repair.

The cellular phenotype of A-T represents genome

instability, deficient DNA damage response (DDR),

and elevated oxidative stress, in addition to a

premature senescence component (Shiloh Y et al.,

1982). A-T patients show a striking sensitivity to

the cytotoxic effect of ionizing radiation (Gotoff SP

et al., 1967; Morgan JL et al., 1968). Cells from A-

T patients exhibit marked chromosomal instability

and sensitivity to ionizing radiations and

radiomimetic chemicals (Taylor AM, 1978; Taylor

AM et al., 1975; Taylor AM et al., 1979; Shiloh Y

Page 43: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 476

et al., 1982; Shiloh Y et al., 1983; Djuzenova CS et

al., 1999). This acute sensitivity results from a

profound defect in the cellular response to DNA

double-strand breaks (DSBs), whose chief

mobilizer is the ATM protein. It is important to

note, however, that A-T cells are also moderately

sensitive to a wide array of other DNA damaging

agents suggesting that these cells cope less

efficiently with many other DNA lesions besides

DSBs.

Phenotype and clinics

Onset of the disease is often noted during the

second year of life: there is progressive cerebellar

ataxia (initially truncal, with further peripheral

extension); ataxia is a constant feature in this

disease; oculomotor apraxia, dysarthria, and

dystonia; leading to muscular atrophia.

Cerebellar ataxia. The prominent symptom of

classical A-T is progressive cerebellar ataxia that

develops into a general motor dysfunction,

eventually confining most patients to a wheelchair

around the end of their first decade (Crawford TO,

1998; Chun HH et al., 2004; Nissenkorn A et al.,

2016; Boder E et al., 1958; Sedgwick RP et al.,

1960; Boder E, 1985; Crawford TO et al., 2000;

Gatti RA, 1995; Verhagen MM et al., 2012). The

main underlying pathology appears to be

progressive cerebellar cortical degeneration that

primarily affects Purkinje and granule neurons, but

also basket cells (Vinters HV et al., 1985; Gatti RA

et al., 1985).

Impairment of the extrapyramidal movement

system is common in A-T, as are oculomotor

abnormalities such as apraxia, strabismus and

nystagmus. Swallowing and articulation of speech

are often abnormal, and facial expression is limited.

Dysfunctional swallowing is often associated with a

general nutritional problem as well as clinically

unapparent aspiration, which is thought to play a

role in the increasing frequency of lower respiratory

tract infections in many patients (Lefton-Greif MA

et al., 2000; Bhatt JM et al., 2015). An absence of

deep reflexes and peripheral neuropathy are

common in A-T, but usually develop relatively later

than other neurological impairments (Nissenkorn A

et al., 2016).

Oculocutaneous telangiectasia (dilated blood

vessels) appear at various ages, usually in the eyes

(conjunctiva) and sometimes on the ears and facial

skin exposed to sunlight, (Perlman SL et al., 2012;

Greenberger S et al., 2013). Finally, telangiectasia

appear in the brain and other internal organs of

young adults with A-T, a peculiar finding seen in

people without A-T only as a late effect of

treatment with ionizing radiation for cancer therapy

(Lin DD et al., 2014).

Combined Immunodeficiency (in 70 %) is

another hallmark of A-T. Typically, IgA, IgE and

various IgG subclasses are reduced; a diminished

lymphocyte count is common, affecting B and T

but not natural killer cells, and many have impaired

antibody responses to vaccines (Gatti RA, 1995),

(Nowak-Wegrzyn A et al., 2004; Gatti RA et al.,

1982; Weaver M et al., 1985; Härtlova A et al.,

2015). The thymus is typically vestigial, as are the

gonads.

Growth/Puberty. Many children with A-T grow at

a diminished rate, and puberty is often delayed; this

growth retardation was suggested to result from a

primary endocrine defect (Ehlayel M et al., 2014;

Voss S et al., 2014; Pommerening H et al., 2015;

Ehlayel M et al., 2014), or a primary growth defect

(Nissenkorn A et al., 2016), but is probably also a

function of swallowing problems making eating an

inefficient and exhausting task.

Dyslipidemia and diabetes. There was also an

increased incidence of dyslipidemia (10/52 = 19%)

and diabetes (2/52 = 4%; Nissenkorn A et al.,

2016). These abnormalities together with elevated

levels of C-reactive protein suggest a diagnosis of

metabolic syndrome in a substantial number of

young A-T patients. Insulin-resistant diabetes is an

important endocrine abnormality in some patients

(Nissenkorn A et al., 2016; Schalch DS et al., 1970;

Morrell D et al., 1986; Blevins LS Jr et al., 1996).

Osteoporosis is common because of a lack of

weight bearing, nutritional deficiencies, and early

gonadal failure in females. Incapacitating fatigue

affects a majority of A-T patients over the age of

30. The etiology of this problem is likely to be

multifactorial, with contributions from the extra

effort required to function with neurodegeneration,

and central nervous system effects of elevated

levels of pro-inflammatory cytokines including IL-

6 and IL-8 (McGrath-Morrow SA et al., 2016) and

chronic, elevated levels of Type I interferons

(Härtlova A et al., 2015).

SenescenceA-T has recently emerged as a

premature aging disease. The broad immune system

defects in A-T have been regarded as a reflection of

premature ageing of this system in these patients

(Exley AR et al., 2011; Carney EF et al., 2012).

Finding striking similarities between the immune

system phenotypes of A-T patients and the elderly

(Carney EF et al., 2012), it was concluded that the

immune system of A-T patients is congenitally

aged, and A-T could be viewed as a model of

immune ageing (Exley AR et al., 2011). Similarly,

the resemblance between ageing-associated decline

of brain functionality and neurodegeneration

associated with genome instability has recently

been highlighted (Barzilai A et al., 2016).

Adolescents and young adults with A-T exhibit an

array of health problems that are typically not seen

until late middle age or later. Among 53 A-T

patients with mean age of 14.6 years (range 5.9 -

26.1), 43% had elevated serum transaminases, 39%

Page 44: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 477

of those patients had fatty liver detected by

ultrasound, and 33% of the latter group developed

steatohepatitis , fibrosis or cirrhosis (Weiss B et al.,

2016). Progeric features of skin include premature

greying and thinning of hair, thinning of skin, and

vitiligo (Reed WB et al., 1966).

Neoplastic risk

Another prominent clinical hallmark of A-T is

cancer predisposition; risk of cancers is X 100,

consisting mainly of T- cell malignancies (a 70-fold

and 250-fold increased risks of leukemia of both B

cell and T cell origin, and 250-fold increased risks

of non-Hodgkin's lymphoma and Hodgkin's

lymphoma), but not myeloid leukemia (Loeb DM et

al., 2000; Murphy RC et al., 1999; Olsen JH et al.,

2001; Taylor AM et al., 1982). There is a striking

incidence of gammopathy in A-T (Sadighi Akha

AA et al., 1999), another abnormality that is rarely

seen in people < 30 years old.

The most common malignancies in A-T patients of

all ages are of lymphocytic origin. However, among

those from 18-40 years old with cancer, 11/21

(52%) had cancers of solid organs (stomach,

esophagus,liver,parotid gland, thyroid, skin,breast

and lung) that are rarely seen in that age group

among people without A-T (HM Lederman, L

Chessa, unpublished observations).

Cancer treatment is complicated by radiation- and

chemo-sensitivity.

Evolution

Progressive cerebellar degeneration: patients are

usually in a wheelchair by the age of ten.

Prognosis

Respiratory infection is the common cause of death,

with cancer being the second most common.

Survival is often into fourth decade today where

optimal medical care is available.

Cytogenetics

Difficulty to grow cells with phytohemaglutinin:

karyotypes should be performed with interleukine 2

in 4 days cultures.

Lymphocyte cultures from A-T patients often

contain clonal translocations that mainly involve

the loci of the T-cell receptor and immunoglobulin

heavy-chain genes (Butterworth SV et al., 1986;

Kennaugh AA et al., 1986; Taylor AM et al., 1986;

Heppell A et al., 1988; Kojis TL et al., 1991),

pointing to a defect in the maturation of these genes

via V(D)J and class-switch recombination in the

adaptive immune system. Such clones usually

herald the onset of malignancy and expand as

malignancy progresses. Cultured A-T cell strains

exhibit elevated rates of chromosome end

associations and reduced telomere length (Pandita

TK et al., 1995; Smilenov LB et al., 1999; Wood

LD et al., 2001; Metcalfe JA et al., 1996; Vaziri H,

1997). A-T fibroblast strains exhibit similar growth

properties to wild-type cells at early passage levels

but senesce prematurely (Shiloh Y et al., 1982).

Inborn conditions

Spontaneous chromatid/chromosome breaks,

triradials, quadriradials (less prominent

phenomenon than in Fanconi anaemia); telomeric

associations.

The best diagnosis test is on the (pathognomonic)

highly elevated level (10% of mitoses) of

inv(7)(p14q35), t(14;14)(q11;q32), and other non

clonal stable chromosome rearrangements

involving 2p12, 7p14, 7q 35, 14q11, 14q32, and

22q11 (illegitimate recombinations between

immunoglobulin superfamilly genes Ig and TCR);

normal level of those rearrangements are: 1/500

(inv(14)), 1/200 (t(7;14)), 1/10 000 (inv(7)).

Clonal rearrangements further occur in 10% of

patients, but without manifestation of malignancy:

t(14;14), inv(14), or t(X;14).

Cytogenetics of cancer

Clonal rearrangements in T-cell ALL and T-PLL

(prolymphocytic leukaemia) in AT patients are

complex, with the frequent involvement of

t(14;14)(q11;q32)(q11;q32), or t(X;14)(q28;q11),

implicating the genes TCL1 or MTCP1

respectively, as is found in T-Pro Lymphocytic

Leukemia in non-AT patients.

Other findings

High sensitivity to ionizing radiations and to

radiomimetic drugs (diagnostic may in part be

based on the hypersensitivity of AT lymphocytes to

killing by gamma irradiation); cell irradiation does

not inhibit S phase (DNA synthesis): this is quite

pathognomonic of AT, and shows that G1

checkpoint is deficient; there is a lack of TP53,

GADD45 and CDKN1A (P21) induction, and a fall

in radiation-induced apoptosis; TP53

phosphorylation at ser15 is deficient.

Telomeres. The observation of accelerated

telomere shortening and telomere fusions in

peripheral blood lymphocytes (Metcalfe JA et al.,

1996) and cultured fibroblasts (Xia SJ et al., 1996;

Smilenov LB et al., 1997) from A-T patients and

cell lines expressing dominant-negative ATM

fragments (Smilenov LB et al., 1997) exposed an

important possible contributor to premature

senescence of ATM-deficient cells. The wealth of

information currently available on telomere

maintenance and the role of the DDR in telomere

dynamics (reviewed in (Webb CJ et al., 2013;

Doksani Y et al., 2014; Arnoult N et al., 2015) has

tightly linked ATM to telomere homeostasis and

added an important component to the ageing aspect

of A-T.

Page 45: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 478

Sporadic (rows 1 and 2) and clonal (row 3) rearrangements in ataxia telangiectasia (R- banding). Row 1, from left to right: inv(7)(p14q35), t(7;7)(p14;q35), t(14;14)(q11;q32), inv(14)(q11q32); Row 2:, from left to right: t(7;14)(p14;q11), t(7;14)(q35;q11), t(7;14)(p14;q32), t(7;14)(q35;q32); Row 3, from left to right: inv(14)(q11;q32), t(X;14)(q28;q11) (note the late replicating X on the

left ), t(14;14)(q11;q32) - Courtesy Alain Aurias (modified figure reprinted from Médecine/Sciences 1986; 2: 298-303., by permission of the publisher Masson).

Lenthening of the cell cycle.

Oxidative stress. Increasing numbers of reports

have described elevated readouts of oxidative stress

in plasma of A-T patients (Reichenbach J et al.,

2002), in cultured A-T fibroblasts (Gatei M et al.,

2001; Lee SA et al., 2001) and lymphocytes

(Ludwig LB et al., 2013), and in tissues and

cultured cells from Atm-deficient mice (Barlow C

et al., 1999; Kamsler A et al., 2001; Gage BM et

al., 2001; Ziv S et al., 2005; Chen P et al., 2003;

Reliene R et al., 2004; Reliene R et al., 2007; Liu N

et al., 2005; McDonald CJ et al., 2011). Notably,

the response of A-T fibroblast strains to induced

oxidative stress was found defective (Yi M et al.,

1990; Ward AJ et al., 1994). These observations

were later linked to the role of ATM in regulating

cellular oxidative stress.

Alpha fetoprotein/serum carcinoembryonic

antigen Notable laboratory findings are elevation

of serum alpha fetoprotein and serum

carcinoembryonic antigen. Further aspects of A-T,

which entail segmental premature ageing.

Genes involved and proteins

Gene

ATM (ataxia telangiectasia mutated)

Location

11q22.3

DNA/RNA

Description

66 exons spanning 184 kb of genomic DNA.

Transcription

Northern blot analysis shows two transcript: the

first-one, TRIM37a of about 4.5-kb and the second-

one, TRIM37b of approximately 3.9 kb.

Protein

Description

3056 amino acids; 350 kDa; contains a Pl 3-kinase-

like domain.

Page 46: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 479

Localisation

Mostly in the nucleus in replicating cells, cytoplasm

in differentiating cells.

Function

Homeostatic protein kinase involved in many

cellular circuits. A primary role in the DNA damage

response. Activated vigorously by DNA double-

strand breaks and activates a broad network of

responses. ATM initiates cell cycle checkpoints in

response to double-strand DNA breaks by

phosphorylating TP53, BRCA1, H2AFX ID:

40783,

Double strand breaks. The most widely

documented function of ATM, and the one

associated with its most vigorous activation, is the

mobilization of the complex signaling network that

responds to DSBs in the DNA (Shiloh Y et al.,

2013; Cremona CA et al., 2014; Awasthi P et al.,

2016; Thompson LH, 2012; McKinnon PJ, 2012).

DSBs are induced by exogenous DNA breaking

agents or endogenous reactive oxygen species

(Schieber M et al., 2014), and are an integral part of

physiological processes including meiotic

recombination (Borde V et al., 2013; Lange J et al.,

2011) and the rearrangement of antigen receptor

genes in the adaptive immune system (Alt FW et

al., 2013). DSBs are repaired via nonhomologous

end-joining (NHEJ), or homologous recombination

repair (HRR; Shibata A et al., 2014; Chapman JR et

al., 2012; Jasin M et al., 2013; Radhakrishnan SK et

al., 2014). Once ATM mobilizes the vast DDR

network in response to a DSB (McKinnon PJ, 2012;

Shiloh Y et al., 2013; Bhatti S et al., 2011), its

protein kinase activity is rapidly enhanced. ATM

subsequently phosphorylates key players in various

arms of the DSB response network (Shiloh Y et al.,

2013; Bensimon A et al., 2010; Matsuoka S et al.,

2007; Mu JJ et al., 2007; Bensimon A et al., 2011),

including other protein kinases that in turn

phosphorylate still other targets (Bensimon A et al.,

2011).

Single-strand break repair and base excision

repair. A broader, overarching role for ATM in

maintaining genome stability was recently

suggested in addition to mobilizing the DSB

response (Shiloh Y, 2014). According to this

conjecture, ATM supports other DNA repair

pathways that respond to various genotoxic

stresses, among them single-strand break repair

(SSBR; Khoronenkova SV et al., 2015) and base

excision repair (BER) - a cardinal pathway in

dealing with the daily nuclear and mitochondrial

DNA damage caused by endogenous agents

(Wallace SS, 2014; Bauer NC et al., 2015). ATM's

involvement in these processes is based on its

ability to phosphorylate proteins that function in

these pathways.

This ongoing role of ATM is its routine function in

the daily maintenance of genome stability, while its

powerful role in the DSB response is reserved for

when this harmful lesion interferes with the daily

life of a cell. Thus, when ATM is missing, not only

is there markedly reduced response to DSBs, the

ongoing modulation of numerous pathways in

response to occasional stresses becomes

suboptimal. All of these lesions are part of the daily

wear and tear on the genome that contributes to

ageing.

An additional role for ATM in genome dynamics

was proposed following evidence that ATM is

involved in shaping the epigenome in neurons by

regulating the localization of the histone

deacetylase 4 (HDAC4; Li J et al., 2012; Herrup K

et al., 2013; Herrup K, 2013), targeting the EZH2

component of the polycomb repressive complex 2

(Li J et al., 2013), and regulating the levels of 5-

hydroxymethylcytosine in Purkinje cells (Jiang D et

al., 2015).

Oxidative stress/Cellular homeostasis. Cytoplasmic fraction of ATM. ATM's role in

cellular homeostasis is further expanded by its

cytoplasmic fraction. Specifically, cytoplasmic

ATM was found to be associated with peroxisomes

(Watters D et al., 1999; Tripathi DN et al., 2016;

Zhang J et al., 2015) and mitochondria (Valentin-

Vega YA et al., 2012). In view of the evidence of

increased oxidative stress in ATM-deficient cells, it

has long been suspected that ATM senses and

responds to oxidative stress (Gatei M et al., 2001;

Rotman G et al., 1997; Rotman G et al., 1997;

Barzilai A et al., 2002; Watters DJ, 2003; Takao N

et al., 2000; Alexander A et al., 2010). This

conjecture was validated by work from the Paull lab

(Guo Z et al., 2010a), which identified an MRN-

independent mode of ATM activation,

differentiating it from DSB-induced activation,

stimulated by reactive oxygen species (ROS) and

leading to ATM oxidation (Paull TT, 2015; Guo Z

et al., 2010a; Guo Z et al., 2010b; Lee JH et al.,

2014).

Mitochondrial fraction of ATM. Still another

arm of the ATM-mediated response to oxidative

stress operates in the mitochondrial fraction of

ATM. ATM is thus emerging also as a regulator of

mitochondrial homeostasis. Evidence is

accumulating of its involvement in mitochondrial

function, mitophagy, and the integrity of

mitochondrial DNA (Valentin-Vega YA et al.,

2012; Ambrose M et al., 2007; Eaton JS et al.,

2007; Fu X et al., 2008; Valentin-Vega YA et al.,

2012; D'Souza AD et al., 2013; Sharma NK et al.,

2014) and further work is needed to identify its

substrates in mitochondria and the mechanistic

aspects of its action in this arena.

Page 47: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 480

Links between ATM and the SASP (senescence-

associated secretory phenotype). Several

laboratories recently described direct links between

ATM and the SASP - a cardinal feature of cell

senescence. Work from the Gamble lab (Chen H et

al., 2015) showed that the histone variant

macroH2A.1 is required for full transcriptional

activation of SASP-promoting genes, driving a

positive feedback loop that enhances cell

senescence. This response is countered by a

negative feedback loop that involves ATM

activation by endoplasmic reticulum stress, elevated

ROS levels or DNA damage. ATM's activity is

required for the removal of macroH2A.1 from sites

of SASP genes, thus leading to SASP gene

repression.

Insulin response and lipoprotein metabolism. IGF-1 receptor. Another pathway by which ATM

may impact on cellular senescence is the

dependence of IGF1R (IGF-1 receptor) expression

on ATM (Peretz S et al., 2001; Goetz EM et al.,

2011; Ching JK et al., 2013).

Beta-adrenergic receptor. Another series of

observations assigned ATM a protective role in

cardiac myocyte apoptosis stimulated by β-

adrenergic receptor and myocardial remodeling.

Mutations

Germinal

Various types of mutations, dispersed throughout

the gene, and therefore most patients are compound

heterozygotes; however, most mutations appear to

inactivate the ATM protein by truncation, large

deletions, or annulation of initiation or termination.

Missense mutations have been described in breast

cancer patients, but do not seem to contribute to

ataxia-telangiectasia.

Patients with the severe form of A-T are

homozygous or compound heterozygous for null

ATM alleles. The corresponding mutations usually

lead to truncation of the ATM protein and

subsequently to its loss due to instability of the

truncated derivatives; a smaller portion of the

mutations create amino acid substitutions that

abolish ATM's catalytic activity (Taylor AM et al.,

2015; Gilad S et al., 1996; Sandoval N et al., 1999;

Barone G et al., 2009) (see also

http://chromium.liacs.nl/LOVD2/home.php?select_

db=ATM).

Careful inspection of the neurological symptoms of

A-T patients reveals variability in their age of onset

and rate of progression among patients with

different combinations of null ATM alleles (Taylor

AM et al., 2015; Crawford TO et al., 2000;

Alterman N et al., 2007). Thus, despite the identical

outcome in terms of ATM function, additional

genes may affect the most cardinal symptom of A-

T. Other, milder types of ATM mutations further

extend this variability, and account for forms of the

disease with extremely variable severity and age of

onset of symptoms.

The corresponding ATM genotypes are

combinations of hypomorphic alleles or

combinations of null and hypomorphic ones. Many

of the latter are leaky splicing mutations and others

are missense mutations, eventually yielding low

amounts of active ATM (Taylor AM et al., 2015;

Alterman N et al., 2007; Soresina A et al., 2008;

Verhagen MM et al., 2009; Silvestri G et al., 2010;

Saunders-Pullman R et al., 2012; Verhagen MM et

al., 2012; Worth PF et al., 2013; Claes K et al.,

2013; Méneret A et al., 2014; Nakamura K et al.,

2014; Gilad S et al., 1998).

To be noted Heterozygote cancer risk: the relative risk of breast

cancer in A-T heterozygote women has been

estimated through epidemiological studies to be 3.9

(CI 2.1-7.1), and through haplotype analysis to be

3.32 (CI 1.75-6.38); since the A-T heterozygote

frequency is about 1 %, 2-4 % of breast cancer

cases may be due to ATM heterozygosity; the risk

of other types of cancer in A-T heterozygotes is

low.

The A-T variant Nijmegen breakage syndrome does

not involve the same gene, but, instead, NBN or

RAD50, involved in the MRE11/RAD50/NBN

double-strand break repair complex.

References Aird KM, Worth AJ, Snyder NW, Lee JV, Sivanand S, Liu Q, Blair IA, Wellen KE, Zhang R. ATM couples replication stress and metabolic reprogramming during cellular senescence. Cell Rep. 2015 May 12;11(6):893-901

Alagoz M, Chiang SC, Sharma A, El-Khamisy SF. ATM deficiency results in accumulation of DNA-topoisomerase I covalent intermediates in neural cells. PLoS One. 2013;8(4):e58239

Alayev A, Holz MK. mTOR signaling for biological control and cancer. J Cell Physiol. 2013 Aug;228(8):1658-64

Alexander A, Walker CL. Differential localization of ATM is correlated with activation of distinct downstream signaling pathways. Cell Cycle. 2010 Sep 15;9(18):3685-6

Alt FW, Zhang Y, Meng FL, Guo C, Schwer B. Mechanisms of programmed DNA lesions and genomic instability in the immune system. Cell. 2013 Jan 31;152(3):417-29

Alterman N, Fattal-Valevski A, Moyal L, Crawford TO, Lederman HM, Ziv Y, Shiloh Y. Ataxia-telangiectasia: mild neurological presentation despite null ATM mutation and severe cellular phenotype. Am J Med Genet A. 2007 Aug 15;143A(16):1827-34

Ambrose M, Goldstine JV, Gatti RA. Intrinsic mitochondrial dysfunction in ATM-deficient lymphoblastoid cells. Hum Mol Genet. 2007 Sep 15;16(18):2154-64

Arnoult N, Karlseder J. Complex interactions between the DNA-damage response and mammalian telomeres. Nat Struct Mol Biol. 2015 Nov;22(11):859-66

Page 48: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 481

Aurias A, Croquette MF, Nuyts JP, Griscelli C, Dutrillaux B. New data on clonal anomalies of chromosome 14 in ataxia telangiectasia: tct(14;14) and inv(14). Hum Genet. 1986 Jan;72(1):22-4

Aurias A, Dutrillaux B. Probable involvement of immunoglobulin superfamily genes in most recurrent chromosomal rearrangements from ataxia telangiectasia. Hum Genet. 1986 Mar;72(3):210-4

Awasthi P, Foiani M, Kumar A. ATM and ATR signaling at a glance. J Cell Sci. 2016 Mar 15;129(6):1285

BODER E, SEDGWICK RP. Ataxia-telangiectasia; a familial syndrome of progressive cerebellar ataxia, oculocutaneous telangiectasia and frequent pulmonary infection. Pediatrics. 1958 Apr;21(4):526-54

Bakkenist CJ, Kastan MB. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature. 2003 Jan 30;421(6922):499-506

Baretić D, Williams RL. PIKKs--the solenoid nest where partners and kinases meet. Curr Opin Struct Biol. 2014 Dec;29:134-42

Barfknecht TR, Little JB. Hypersensitivity of ataxia telangiectasia skin fibroblasts to DNA alkylating agents. Mutat Res. 1982 Jun;94(2):369-82

Barlow C, Brown KD, Deng CX, Tagle DA, Wynshaw-Boris A. Atm selectively regulates distinct p53-dependent cell-cycle checkpoint and apoptotic pathways. Nat Genet. 1997 Dec;17(4):453-6

Barlow C, Dennery PA, Shigenaga MK, Smith MA, Morrow JD, Roberts LJ 2nd, Wynshaw-Boris A, Levine RL. Loss of the ataxia-telangiectasia gene product causes oxidative damage in target organs. Proc Natl Acad Sci U S A. 1999 Aug 17;96(17):9915-9

Barlow C, Hirotsune S, Paylor R, Liyanage M, Eckhaus M, Collins F, Shiloh Y, Crawley JN, Ried T, Tagle D, Wynshaw-Boris A. Atm-deficient mice: a paradigm of ataxia telangiectasia. Cell. 1996 Jul 12;86(1):159-71

Barone G, Groom A, Reiman A, Srinivasan V, Byrd PJ, Taylor AM. Modeling ATM mutant proteins from missense changes confirms retained kinase activity. Hum Mutat. 2009 Aug;30(8):1222-30

Barzilai A, Rotman G, Shiloh Y. ATM deficiency and oxidative stress: a new dimension of defective response to DNA damage. DNA Repair (Amst). 2002 Jan 22;1(1):3-25

Bauer NC, Corbett AH, Doetsch PW. The current state of eukaryotic DNA base damage and repair. Nucleic Acids Res. 2015 Dec 2;43(21):10083-101

Bensimon A, Aebersold R, Shiloh Y. Beyond ATM: the protein kinase landscape of the DNA damage response. FEBS Lett. 2011 Jun 6;585(11):1625-39

Bhatt JM, Bush A, van Gerven M, Nissenkorn A, Renke M, Yarlett L, Taylor M, Tonia T, Warris A, Zielen S, Zinna S, Merkus PJ. ERS statement on the multidisciplinary respiratory management of ataxia telangiectasia. Eur Respir Rev. 2015 Dec;24(138):565-81

Bhatti S, Kozlov S, Farooqi AA, Naqi A, Lavin M, Khanna KK. ATM protein kinase: the linchpin of cellular defenses to stress. Cell Mol Life Sci. 2011 Sep;68(18):2977-3006

Blevins LS Jr, Gebhart SS. Insulin-resistant diabetes mellitus in a black woman with ataxia-telangiectasia. South Med J. 1996 Jun;89(6):619-21

Boder E. Ataxia-telangiectasia: an overview. Kroc Found Ser. 1985;19:1-63

Borde V, de Massy B. Programmed induction of DNA double strand breaks during meiosis: setting up communication between DNA and the chromosome structure. Curr Opin Genet Dev. 2013 Apr;23(2):147-55

Broeks A, Urbanus JH, Floore AN, Dahler EC, Klijn JG, Rutgers EJ, Devilee P, Russell NS, van Leeuwen FE, van 't Veer LJ. ATM-heterozygous germline mutations contribute to breast cancer-susceptibility. Am J Hum Genet. 2000 Feb;66(2):494-500

Burma S, Chen BP, Murphy M, Kurimasa A, Chen DJ. ATM phosphorylates histone H2AX in response to DNA double-strand breaks. J Biol Chem. 2001 Nov 9;276(45):42462-7

Butterworth SV, Taylor AM. A subpopulation of t(2;14)(p11;q32) cells in ataxia telangiectasia B lymphocytes. Hum Genet. 1986 Aug;73(4):346-9

Carney EF, Srinivasan V, Moss PA, Taylor AM. Classical ataxia telangiectasia patients have a congenitally aged immune system with high expression of CD95. J Immunol. 2012 Jul 1;189(1):261-8

Chapman JR, Taylor MR, Boulton SJ. Playing the end game: DNA double-strand break repair pathway choice. Mol Cell. 2012 Aug 24;47(4):497-510

Chen BP, Li M, Asaithamby A. New insights into the roles of ATM and DNA-PKcs in the cellular response to oxidative stress. Cancer Lett. 2012 Dec 31;327(1-2):103-10

Chen H, Ruiz PD, McKimpson WM, Novikov L, Kitsis RN, Gamble MJ. MacroH2A1 and ATM Play Opposing Roles in Paracrine Senescence and the Senescence-Associated Secretory Phenotype. Mol Cell. 2015 Sep 3;59(5):719-31

Chen P, Peng C, Luff J, Spring K, Watters D, Bottle S, Furuya S, Lavin MF. Oxidative stress is responsible for deficient survival and dendritogenesis in purkinje neurons from ataxia-telangiectasia mutated mutant mice. J Neurosci. 2003 Dec 10;23(36):11453-60

Ching JK, Luebbert SH, Collins RL 4th, Zhang Z, Marupudi N, Banerjee S, Hurd RD, Ralston L, Fisher JS. Ataxia telangiectasia mutated impacts insulin-like growth factor 1 signalling in skeletal muscle. Exp Physiol. 2013 Feb;98(2):526-35

Chun HH, Gatti RA. Ataxia-telangiectasia, an evolving phenotype. DNA Repair (Amst). 2004 Aug-Sep;3(8-9):1187-96

Claes K, Depuydt J, Taylor AM, Last JI, Baert A, Schietecatte P, Vandersickel V, Poppe B, De Leeneer K, D'Hooghe M, Vral A. Variant ataxia telangiectasia: clinical and molecular findings and evaluation of radiosensitive phenotypes in a patient and relatives. Neuromolecular Med. 2013 Sep;15(3):447-57

Cornu M, Albert V, Hall MN. mTOR in aging, metabolism, and cancer. Curr Opin Genet Dev. 2013 Feb;23(1):53-62

Cosentino C, Grieco D, Costanzo V. ATM activates the pentose phosphate pathway promoting anti-oxidant defence and DNA repair. EMBO J. 2011 Feb 2;30(3):546-55

Crawford TO. Ataxia telangiectasia. Semin Pediatr Neurol. 1998 Dec;5(4):287-94

Crawford TO, Mandir AS, Lefton-Greif MA, Goodman SN, Goodman BK, Sengul H, Lederman HM. Quantitative neurologic assessment of ataxia-telangiectasia. Neurology. 2000 Apr 11;54(7):1505-9

Cremona CA, Behrens A. ATM signalling and cancer.

Page 49: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 482

Oncogene. 2014 Jun 26;33(26):3351-60

D'Souza AD, Parish IA, Krause DS, Kaech SM, Shadel GS. Reducing mitochondrial ROS improves disease-related pathology in a mouse model of ataxia-telangiectasia. Mol Ther. 2013 Jan;21(1):42-8

Daniel LL, Daniels CR, Harirforoosh S, Foster CR, Singh M, Singh K. Deficiency of ataxia telangiectasia mutated kinase delays inflammatory response in the heart following myocardial infarction. J Am Heart Assoc. 2014 Dec;3(6):e001286

Davis AJ, Chen BP, Chen DJ. DNA-PK: a dynamic enzyme in a versatile DSB repair pathway. DNA Repair (Amst). 2014 May;17:21-9

Ditch S, Paull TT. The ATM protein kinase and cellular redox signaling: beyond the DNA damage response. Trends Biochem Sci. 2012 Jan;37(1):15-22

Djuzenova CS, Schindler D, Stopper H, Hoehn H, Flentje M, Oppitz U. Identification of ataxia telangiectasia heterozygotes, a cancer-prone population, using the single-cell gel electrophoresis (Comet) assay. Lab Invest. 1999 Jun;79(6):699-705

Doksani Y, de Lange T. The role of double-strand break repair pathways at functional and dysfunctional telomeres. Cold Spring Harb Perspect Biol. 2014 Sep 16;6(12):a016576

Eaton JS, Lin ZP, Sartorelli AC, Bonawitz ND, Shadel GS. Ataxia-telangiectasia mutated kinase regulates ribonucleotide reductase and mitochondrial homeostasis. J Clin Invest. 2007 Sep;117(9):2723-34

Ehlayel M, Soliman A, De Sanctis V. Linear growth and endocrine function in children with ataxia telangiectasia. Indian J Endocrinol Metab. 2014 Nov;18(Suppl 1):S93-6

Errico A, Costanzo V. Mechanisms of replication fork protection: a safeguard for genome stability. Crit Rev Biochem Mol Biol. 2012 May-Jun;47(3):222-35

Espach Y, Lochner A, Strijdom H, Huisamen B. ATM protein kinase signaling, type 2 diabetes and cardiovascular disease. Cardiovasc Drugs Ther. 2015 Feb;29(1):51-8

Exley AR, Buckenham S, Hodges E, Hallam R, Byrd P, Last J, Trinder C, Harris S, Screaton N, Williams AP, Taylor AM, Shneerson JM. Premature ageing of the immune system underlies immunodeficiency in ataxia telangiectasia. Clin Immunol. 2011 Jul;140(1):26-36

Fedier A, Schlamminger M, Schwarz VA, Haller U, Howell SB, Fink D. Loss of atm sensitises p53-deficient cells to topoisomerase poisons and antimetabolites. Ann Oncol. 2003 Jun;14(6):938-45

Foster CR, Daniel LL, Daniels CR, Dalal S, Singh M, Singh K. Deficiency of ataxia telangiectasia mutated kinase modulates cardiac remodeling following myocardial infarction: involvement in fibrosis and apoptosis. PLoS One. 2013;8(12):e83513

Foster CR, Singh M, Subramanian V, Singh K. Ataxia telangiectasia mutated kinase plays a protective role in β-adrenergic receptor-stimulated cardiac myocyte apoptosis and myocardial remodeling. Mol Cell Biochem. 2011 Jul;353(1-2):13-22

Foster CR, Zha Q, Daniel LL, Singh M, Singh K. Lack of ataxia telangiectasia mutated kinase induces structural and functional changes in the heart: role in β-adrenergic receptor-stimulated apoptosis. Exp Physiol. 2012 Apr;97(4):506-15

Fu X, Wan S, Lyu YL, Liu LF, Qi H. Etoposide induces ATM-dependent mitochondrial biogenesis through AMPK activation. PLoS One. 2008 Apr 23;3(4):e2009

Gage BM, Alroy D, Shin CY, Ponomareva ON, Dhar S, Sharma GG, Pandita TK, Thayer MJ, Turker MS. Spontaneously immortalized cell lines obtained from adult Atm null mice retain sensitivity to ionizing radiation and exhibit a mutational pattern suggestive of oxidative stress. Oncogene. 2001 Jul 19;20(32):4291-7

Gatei M, Shkedy D, Khanna KK, Uziel T, Shiloh Y, Pandita TK, Lavin MF, Rotman G. Ataxia-telangiectasia: chronic activation of damage-responsive functions is reduced by alpha-lipoic acid. Oncogene. 2001 Jan 18;20(3):289-94

Gatti RA. Ataxia-telangiectasia. Dermatol Clin. 1995 Jan;13(1):1-6

Gatti RA, Berkel I, Boder E, Braedt G, Charmley P, Concannon P, Ersoy F, Foroud T, Jaspers NG, Lange K. Localization of an ataxia-telangiectasia gene to chromosome 11q22-23. Nature. 1988 Dec 8;336(6199):577-80

Gatti RA, Bick M, Tam CF, Medici MA, Oxelius VA, Holland M, Goldstein AL, Boder E. Ataxia-Telangiectasia: a multiparameter analysis of eight families. Clin Immunol Immunopathol. 1982 May;23(2):501-16

Gatti RA, Vinters HV. Cerebellar pathology in ataxia-telangiectasia: the significance of basket cells. Kroc Found Ser. 1985;19:225-32

Gilad S, Chessa L, Khosravi R, Russell P, Galanty Y, Piane M, Gatti RA, Jorgensen TJ, Shiloh Y, Bar-Shira A. Genotype-phenotype relationships in ataxia-telangiectasia and variants. Am J Hum Genet. 1998 Mar;62(3):551-61

Gilad S, Khosravi R, Shkedy D, Uziel T, Ziv Y, Savitsky K, Rotman G, Smith S, Chessa L, Jorgensen TJ, Harnik R, Frydman M, Sanal O, Portnoi S, Goldwicz Z, Jaspers NG, Gatti RA, Lenoir G, Lavin MF, Tatsumi K, Wegner RD, Shiloh Y, Bar-Shira A. Predominance of null mutations in ataxia-telangiectasia. Hum Mol Genet. 1996 Apr;5(4):433-9

Gobbini E, Cesena D, Galbiati A, Lockhart A, Longhese MP. Interplays between ATM/Tel1 and ATR/Mec1 in sensing and signaling DNA double-strand breaks. DNA Repair (Amst). 2013 Oct;12(10):791-9

Goetz EM, Shankar B, Zou Y, Morales JC, Luo X, Araki S, Bachoo R, Mayo LD, Boothman DA. ATM-dependent IGF-1 induction regulates secretory clusterin expression after DNA damage and in genetic instability. Oncogene. 2011 Sep 1;30(35):3745-54

Goodarzi AA, Jeggo PA. The repair and signaling responses to DNA double-strand breaks. Adv Genet. 2013;82:1-45

Gotoff SP, Amirmokri E, Liebner EJ. Ataxia telangiectasia. Neoplasia, untoward response to x-irradiation, and tuberous sclerosis. Am J Dis Child. 1967 Dec;114(6):617-25

Greenberger S, Berkun Y, Ben-Zeev B, Levi YB, Barziliai A, Nissenkorn A. Dermatologic manifestations of ataxia-telangiectasia syndrome. J Am Acad Dermatol. 2013 Jun;68(6):932-6

Guleria A, Chandna S. ATM kinase: Much more than a DNA damage responsive protein. DNA Repair (Amst). 2016 Mar;39:1-20

Guo Z, Deshpande R, Paull TT. ATM activation in the presence of oxidative stress. Cell Cycle. 2010 Dec

Page 50: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 483

15;9(24):4805-11

Guo Z, Kozlov S, Lavin MF, Person MD, Paull TT. ATM activation by oxidative stress. Science. 2010 Oct 22;330(6003):517-21

Härtlova A, Erttmann SF, Raffi FA, Schmalz AM, Resch U, Anugula S, Lienenklaus S, Nilsson LM, Kröger A, Nilsson JA, Ek T, Weiss S, Gekara NO. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity. 2015 Feb 17;42(2):332-343

Halaby MJ, Hibma JC, He J, Yang DQ. ATM protein kinase mediates full activation of Akt and regulates glucose transporter 4 translocation by insulin in muscle cells. Cell Signal. 2008 Aug;20(8):1555-63

Hannan MA, Hellani A, Al-Khodairy FM, Kunhi M, Siddiqui Y, Al-Yussef N, Pangue-Cruz N, Siewertsen M, Al-Ahdal MN, Aboussekhra A. Deficiency in the repair of UV-induced DNA damage in human skin fibroblasts compromised for the ATM gene. Carcinogenesis. 2002 Oct;23(10):1617-24

Henderson EE, Ribecky R. DNA repair in lymphoblastoid cell lines established from human genetic disorders. Chem Biol Interact. 1980 Dec;33(1):63-81

Heppell A, Butterworth SV, Hollis RJ, Kennaugh AA, Beatty DW, Taylor AM. Breakage of the T cell receptor alpha chain locus in non malignant clones from patients with ataxia telangiectasia. Hum Genet. 1988 Aug;79(4):360-4

Herrup K. ATM and the epigenetics of the neuronal genome. Mech Ageing Dev. 2013 Oct;134(10):434-9

Herrup K, Li J, Chen J. The role of ATM and DNA damage in neurons: upstream and downstream connections. DNA Repair (Amst). 2013 Aug;12(8):600-4

Hoar DI, Sargent P. Chemical mutagen hypersensitivity in ataxia telangiectasia. Nature. 1976 Jun 17;261(5561):590-2

Jasin M, Rothstein R. Repair of strand breaks by homologous recombination. Cold Spring Harb Perspect Biol. 2013 Nov 1;5(11):a012740

Jaspers NG, de Wit J, Regulski MR, Bootsma D. Abnormal regulation of DNA replication and increased lethality in ataxia telangiectasia cells exposed to carcinogenic agents. Cancer Res. 1982 Jan;42(1):335-41

Jeong I, Patel AY, Zhang Z, Patil PB, Nadella ST, Nair S, Ralston L, Hoormann JK, Fisher JS. Role of ataxia telangiectasia mutated in insulin signalling of muscle-derived cell lines and mouse soleus. Acta Physiol (Oxf). 2010 Apr;198(4):465-75

Jette N, Lees-Miller SP. The DNA-dependent protein kinase: A multifunctional protein kinase with roles in DNA double strand break repair and mitosis. Prog Biophys Mol Biol. 2015 Mar;117(2-3):194-205

Jiang D, Zhang Y, Hart RP, Chen J, Herrup K, Li J. Alteration in 5-hydroxymethylcytosine-mediated epigenetic regulation leads to Purkinje cell vulnerability in ATM deficiency. Brain. 2015 Dec;138(Pt 12):3520-36

Kaidi A, Jackson SP. KAT5 tyrosine phosphorylation couples chromatin sensing to ATM signalling. Nature. 2013 Jun 6;498(7452):70-4

Kamsler A, Daily D, Hochman A, Stern N, Shiloh Y, Rotman G, Barzilai A. Increased oxidative stress in ataxia telangiectasia evidenced by alterations in redox state of

brains from Atm-deficient mice. Cancer Res. 2001 Mar 1;61(5):1849-54

Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, Lu T, Yankner BA, Campisi J, Elledge SJ. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science. 2015 Sep 25;349(6255):aaa5612

Katyal S, Lee Y, Nitiss KC, Downing SM, Li Y, Shimada M, Zhao J, Russell HR, Petrini JH, Nitiss JL, McKinnon PJ. Aberrant topoisomerase-1 DNA lesions are pathogenic in neurodegenerative genome instability syndromes. Nat Neurosci. 2014 Jun;17(6):813-21

Kennaugh AA, Butterworth SV, Hollis R, Baer R, Rabbitts TH, Taylor AM. The chromosome breakpoint at 14q32 in an ataxia telangiectasia t(14;14) T cell clone is different from the 14q32 breakpoint in Burkitts and an inv(14) T cell lymphoma. Hum Genet. 1986 Jul;73(3):254-9

Khoronenkova SV, Dianov GL. ATM prevents DSB formation by coordinating SSB repair and cell cycle progression. Proc Natl Acad Sci U S A. 2015 Mar 31;112(13):3997-4002

Kojis TL, Gatti RA, Sparkes RS. The cytogenetics of ataxia telangiectasia. Cancer Genet Cytogenet. 1991 Oct 15;56(2):143-56

Kozlov SV, Graham ME, Peng C, Chen P, Robinson PJ, Lavin MF. Involvement of novel autophosphorylation sites in ATM activation. EMBO J. 2006 Aug 9;25(15):3504-14

Kozlov SV, Waardenberg AJ, Engholm-Keller K, Arthur JW, Graham ME, Lavin M. Reactive Oxygen Species (ROS)-Activated ATM-Dependent Phosphorylation of Cytoplasmic Substrates Identified by Large-Scale Phosphoproteomics Screen. Mol Cell Proteomics. 2016 Mar;15(3):1032-47

Krüger A, Ralser M. ATM is a redox sensor linking genome stability and carbon metabolism. Sci Signal. 2011 Apr 5;4(167):pe17

Lange J, Pan J, Cole F, Thelen MP, Jasin M, Keeney S. ATM controls meiotic double-strand-break formation. Nature. 2011 Oct 16;479(7372):237-40

Lavin MF. Ataxia-telangiectasia: from a rare disorder to a paradigm for cell signalling and cancer. Nat Rev Mol Cell Biol. 2008 Oct;9(10):759-69

Lee JH, Guo Z, Myler LR, Zheng S, Paull TT. Direct activation of ATM by resveratrol under oxidizing conditions. PLoS One. 2014;9(6):e97969

Lee JH, Kim KH, Morio T, Kim H. Ataxia-telangiectasia-mutated-dependent activation of Ku in human fibroblasts exposed to hydrogen peroxide. Ann N Y Acad Sci. 2006 Dec;1091:76-82

Lee SA, Dritschilo A, Jung M. Role of ATM in oxidative stress-mediated c-Jun phosphorylation in response to ionizing radiation and CdCl2. J Biol Chem. 2001 Apr 13;276(15):11783-90

Lefton-Greif MA, Crawford TO, Winkelstein JA, Loughlin GM, Koerner CB, Zahurak M, Lederman HM. Oropharyngeal dysphagia and aspiration in patients with ataxia-telangiectasia. J Pediatr. 2000 Feb;136(2):225-31

Leonard JC, Mullinger AM, Schmidt J, Cordell HJ, Johnson RT. Genome instability in ataxia telangiectasia (A-T) families: camptothecin-induced damage to replicating DNA discriminates between obligate A-T heterozygotes, A-T homozygotes and controls. Biosci Rep. 2004

Page 51: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 484

Dec;24(6):617-29

Li J, Chen J, Ricupero CL, Hart RP, Schwartz MS, Kusnecov A, Herrup K. Nuclear accumulation of HDAC4 in ATM deficiency promotes neurodegeneration in ataxia telangiectasia. Nat Med. 2012 May;18(5):783-90

Li J, Hart RP, Mallimo EM, Swerdel MR, Kusnecov AW, Herrup K. EZH2-mediated H3K27 trimethylation mediates neurodegeneration in ataxia-telangiectasia. Nat Neurosci. 2013 Dec;16(12):1745-53

Lin DD, Barker PB, Lederman HM, Crawford TO. Cerebral abnormalities in adults with ataxia-telangiectasia. AJNR Am J Neuroradiol. 2014 Jan;35(1):119-23

Liu N, Stoica G, Yan M, Scofield VL, Qiang W, Lynn WS, Wong PK. ATM deficiency induces oxidative stress and endoplasmic reticulum stress in astrocytes. Lab Invest. 2005 Dec;85(12):1471-80

Loeb DM, Lederman HM, Winkelstein JA. Lymphoid malignancy as a presenting sign of ataxia-telangiectasia. J Pediatr Hematol Oncol. 2000 Sep-Oct;22(5):464-7

Lovejoy CA, Cortez D. Common mechanisms of PIKK regulation. DNA Repair (Amst). 2009 Sep 2;8(9):1004-8

Ludwig LB, Valiati VH, Palazzo RP, Jardim LB, da Rosa DP, Bona S, Rodrigues G, Marroni NP, Prá D, Maluf SW. Chromosome instability and oxidative stress markers in patients with ataxia telangiectasia and their parents. Biomed Res Int. 2013;2013:762048

Luo X, Suzuki M, Ghandhi SA, Amundson SA, Boothman DA. ATM regulates insulin-like growth factor 1-secretory clusterin (IGF-1-sCLU) expression that protects cells against senescence. PLoS One. 2014;9(6):e99983

Méneret A, Ahmar-Beaugendre Y, Rieunier G, Mahlaoui N, Gaymard B, Apartis E, Tranchant C, Rivaud-Péchoux S, Degos B, Benyahia B, Suarez F, Maisonobe T, Koenig M, Durr A, Stern MH, Dubois d'Enghien C, Fischer A, Vidailhet M, Stoppa-Lyonnet D, Grabli D, Anheim M. The pleiotropic movement disorders phenotype of adult ataxia-telangiectasia. Neurology. 2014 Sep 16;83(12):1087-95

Maréchal A, Zou L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect Biol. 2013 Sep 1;5(9)

Matsuoka S, Ballif BA, Smogorzewska A, McDonald ER 3rd, Hurov KE, Luo J, Bakalarski CE, Zhao Z, Solimini N, Lerenthal Y, Shiloh Y, Gygi SP, Elledge SJ. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science. 2007 May 25;316(5828):1160-6

McDonald CJ, Ostini L, Wallace DF, John AN, Watters DJ, Subramaniam VN. Iron loading and oxidative stress in the Atm-/- mouse liver. Am J Physiol Gastrointest Liver Physiol. 2011 Apr;300(4):G554-60

McGrath-Morrow SA, Collaco JM, Detrick B, Lederman HM. Serum Interleukin-6 Levels and Pulmonary Function in Ataxia-Telangiectasia. J Pediatr. 2016 Apr;171:256-61.e1

McKinnon PJ. ATM and the molecular pathogenesis of ataxia telangiectasia. Annu Rev Pathol. 2012;7:303-21

Mercer JR, Cheng KK, Figg N, Gorenne I, Mahmoudi M, Griffin J, Vidal-Puig A, Logan A, Murphy MP, Bennett M. DNA damage links mitochondrial dysfunction to atherosclerosis and the metabolic syndrome. Circ Res. 2010 Oct 15;107(8):1021-31

Mercer JR, Yu E, Figg N, Cheng KK, Prime TA, Griffin JL,

Masoodi M, Vidal-Puig A, Murphy MP, Bennett MR. The mitochondria-targeted antioxidant MitoQ decreases features of the metabolic syndrome in ATM+/-/ApoE-/- mice. Free Radic Biol Med. 2012 Mar 1;52(5):841-9

Metcalfe JA, Parkhill J, Campbell L, Stacey M, Biggs P, Byrd PJ, Taylor AM. Accelerated telomere shortening in ataxia telangiectasia. Nat Genet. 1996 Jul;13(3):350-3

Miles PD, Treuner K, Latronica M, Olefsky JM, Barlow C. Impaired insulin secretion in a mouse model of ataxia telangiectasia. Am J Physiol Endocrinol Metab. 2007 Jul;293(1):E70-4

Mirzayans R, Smith BP, Paterson MC. Hypersensitivity to cell killing and faulty repair of 1-beta-D-arabinofuranosylcytosine-detectable sites in human (ataxia-telangiectasia) fibroblasts treated with 4-nitroquinoline 1-oxide. Cancer Res. 1989 Oct 15;49(20):5523-9

Morgan JL, Holcomb TM, Morrissey RW. Radiation reaction in ataxia telangiectasia. Am J Dis Child. 1968 Nov;116(5):557-8

Morrell D, Chase CL, Kupper LL, Swift M. Diabetes mellitus in ataxia-telangiectasia, Fanconi anemia, xeroderma pigmentosum, common variable immune deficiency, and severe combined immune deficiency families. Diabetes. 1986 Feb;35(2):143-7

Mu JJ, Wang Y, Luo H, Leng M, Zhang J, Yang T, Besusso D, Jung SY, Qin J. A proteomic analysis of ataxia telangiectasia-mutated (ATM)/ATM-Rad3-related (ATR) substrates identifies the ubiquitin-proteasome system as a regulator for DNA damage checkpoints. J Biol Chem. 2007 Jun 15;282(24):17330-4

Murphy RC, Berdon WE, Ruzal-Shapiro C, Hall EJ, Kornecki A, Daneman A, Brunelle F, Campbell JB. Malignancies in pediatric patients with ataxia telangiectasia. Pediatr Radiol. 1999 Apr;29(4):225-30

Nakamura K, Fike F, Haghayegh S, Saunders-Pullman R, Dawson AJ, Dörk T, Gatti RA. A-TWinnipeg: Pathogenesis of rare ATM missense mutation c.6200C>A with decreased protein expression and downstream signaling, early-onset dystonia, cancer, and life-threatening radiotoxicity. Mol Genet Genomic Med. 2014 Jul;2(4):332-40

Nissenkorn A, Levy-Shraga Y, Banet-Levi Y, Lahad A, Sarouk I, Modan-Moses D. Endocrine abnormalities in ataxia telangiectasia: findings from a national cohort. Pediatr Res. 2016 Jun;79(6):889-94

Nowak-Wegrzyn A, Crawford TO, Winkelstein JA, Carson KA, Lederman HM. Immunodeficiency and infections in ataxia-telangiectasia. J Pediatr. 2004 Apr;144(4):505-11

Olsen JH, Hahnemann JM, Børresen-Dale AL, Brøndum-Nielsen K, Hammarström L, Kleinerman R, Kääriäinen H, Lönnqvist T, Sankila R, Seersholm N, Tretli S, Yuen J, Boice JD Jr, Tucker M. Cancer in patients with ataxia-telangiectasia and in their relatives in the nordic countries. J Natl Cancer Inst. 2001 Jan 17;93(2):121-7

Pandita TK, Pathak S, Geard CR. Chromosome end associations, telomeres and telomerase activity in ataxia telangiectasia cells. Cytogenet Cell Genet. 1995;71(1):86-93

Panier S, Durocher D. Push back to respond better: regulatory inhibition of the DNA double-strand break response. Nat Rev Mol Cell Biol. 2013 Oct;14(10):661-72

Paterson MC, Smith BP, Lohman PH, Anderson AK,

Page 52: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 485

Fishman L. Defective excision repair of gamma-ray-damaged DNA in human (ataxia telangiectasia) fibroblasts. Nature. 1976 Apr 1;260(5550):444-7

Paull TT. Mechanisms of ATM Activation. Annu Rev Biochem. 2015;84:711-38

Peretz S, Jensen R, Baserga R, Glazer PM. ATM-dependent expression of the insulin-like growth factor-I receptor in a pathway regulating radiation response. Proc Natl Acad Sci U S A. 2001 Feb 13;98(4):1676-81

Perlman SL, Boder Deceased E, Sedgewick RP, Gatti RA. Ataxia-telangiectasia. Handb Clin Neurol. 2012;103:307-32

Platzer M, Rotman G, Bauer D, Uziel T, Savitsky K, Bar-Shira A, Gilad S, Shiloh Y, Rosenthal A. Ataxia-telangiectasia locus: sequence analysis of 184 kb of human genomic DNA containing the entire ATM gene. Genome Res. 1997 Jun;7(6):592-605

Polo SE, Jackson SP. Dynamics of DNA damage response proteins at DNA breaks: a focus on protein modifications. Genes Dev. 2011 Mar 1;25(5):409-33

Pommerening H, van Dullemen S, Kieslich M, Schubert R, Zielen S, Voss S. Body composition, muscle strength and hormonal status in patients with ataxia telangiectasia: a cohort study. Orphanet J Rare Dis. 2015 Dec 9;10:155

Radhakrishnan SK, Jette N, Lees-Miller SP. Non-homologous end joining: emerging themes and unanswered questions. DNA Repair (Amst). 2014 May;17:2-8

Reed WB, Epstein WL, Boder E, Sedgwick R. Cutaneous manifestations of ataxia-telangiectasia. JAMA. 1966 Feb 28;195(9):746-53

Reichenbach J, Schubert R, Schindler D, Müller K, Böhles H, Zielen S. Elevated oxidative stress in patients with ataxia telangiectasia. Antioxid Redox Signal. 2002 Jun;4(3):465-9

Reliene R, Fischer E, Schiestl RH. Effect of N-acetyl cysteine on oxidative DNA damage and the frequency of DNA deletions in atm-deficient mice. Cancer Res. 2004 Aug 1;64(15):5148-53

Reliene R, Schiestl RH. Antioxidants suppress lymphoma and increase longevity in Atm-deficient mice. J Nutr. 2007 Jan;137(1 Suppl):229S-232S

Rotman G, Shiloh Y. The ATM gene and protein: possible roles in genome surveillance, checkpoint controls and cellular defence against oxidative stress. Cancer Surv. 1997;29:285-304

SEDGWICK RP, BODER E. Progressive ataxia in childhood with particular reference to ataxia-telangiectasia. Neurology. 1960 Jul;10:705-15

Sadighi Akha AA, Humphrey RL, Winkelstein JA, Loeb DM, Lederman HM. Oligo-/monoclonal gammopathy and hypergammaglobulinemia in ataxia-telangiectasia. A study of 90 patients. Medicine (Baltimore). 1999 Nov;78(6):370-81

Saunders-Pullman R, Raymond D, Stoessl AJ, Hobson D, Nakamura K, Pullman S, Lefton D, Okun MS, Uitti R, Sachdev R, Stanley K, San Luciano M, Hagenah J, Gatti R, Ozelius LJ, Bressman SB. Variant ataxia-telangiectasia presenting as primary-appearing dystonia in Canadian Mennonites. Neurology. 2012 Feb 28;78(9):649-57

Savitsky K, Sfez S, Tagle DA, Ziv Y, Sartiel A, Collins FS, Shiloh Y, Rotman G. The complete sequence of the coding region of the ATM gene reveals similarity to cell cycle

regulators in different species. Hum Mol Genet. 1995 Nov;4(11):2025-32

Schalch DS, McFarlin DE, Barlow MH. An unusual form of diabetes mellitus in ataxia telangiectasia. N Engl J Med. 1970 Jun 18;282(25):1396-402

Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol. 2014 May 19;24(10):R453-62

Schneider JG, Finck BN, Ren J, Standley KN, Takagi M, Maclean KH, Bernal-Mizrachi C, Muslin AJ, Kastan MB, Semenkovich CF. ATM-dependent suppression of stress signaling reduces vascular disease in metabolic syndrome. Cell Metab. 2006 Nov;4(5):377-89

Scudiero DA. Decreased DNA repair synthesis and defective colony-forming ability of ataxia telangiectasia fibroblast cell strains treated with N-methyl-N'-nitro-N-nitrosoguanidine. Cancer Res. 1980 Apr;40(4):984-90

Segal-Raz H, Mass G, Baranes-Bachar K, Lerenthal Y, Wang SY, Chung YM, Ziv-Lehrman S, Ström CE, Helleday T, Hu MC, Chen DJ, Shiloh Y. ATM-mediated phosphorylation of polynucleotide kinase/phosphatase is required for effective DNA double-strand break repair. EMBO Rep. 2011 Jul 1;12(7):713-9

Semlitsch M, Shackelford RE, Zirkl S, Sattler W, Malle E. ATM protects against oxidative stress induced by oxidized low-density lipoprotein. DNA Repair (Amst). 2011 Aug 15;10(8):848-60

Sharma NK, Lebedeva M, Thomas T, Kovalenko OA, Stumpf JD, Shadel GS, Santos JH. Intrinsic mitochondrial DNA repair defects in Ataxia Telangiectasia. DNA Repair (Amst). 2014 Jan;13:22-31

Shibata A, Jeggo PA. DNA double-strand break repair in a cellular context. Clin Oncol (R Coll Radiol). 2014 May;26(5):243-9

Shiloh Y. ATM: expanding roles as a chief guardian of genome stability. Exp Cell Res. 2014 Nov 15;329(1):154-61

Shiloh Y, Tabor E, Becker Y. Cells from patients with ataxia telangiectasia are abnormally sensitive to the cytotoxic effect of a tumor promoter, phorbol-12-myristate-13-acetate. Mutat Res. 1985 Apr;149(2):283-6

Shiloh Y, Ziv Y. The ATM protein kinase: regulating the cellular response to genotoxic stress, and more. Nat Rev Mol Cell Biol. 2013 Apr;14(4):197-210

Silvestri G, Masciullo M, Piane M, Savio C, Modoni A, Santoro M, Chessa L. Homozygosity for c 6325T>G transition in the ATM gene causes an atypical, late-onset variant form of ataxia-telangiectasia. J Neurol. 2010 Oct;257(10):1738-40

Sirbu BM, Cortez D. DNA damage response: three levels of DNA repair regulation. Cold Spring Harb Perspect Biol. 2013 Aug 1;5(8):a012724

Smilenov LB, Morgan SE, Mellado W, Sawant SG, Kastan MB, Pandita TK. Influence of ATM function on telomere metabolism. Oncogene. 1997 Nov 27;15(22):2659-65

Smith PJ, Makinson TA, Watson JV. Enhanced sensitivity to camptothecin in ataxia-telangiectasia cells and its relationship with the expression of DNA topoisomerase I. Int J Radiat Biol. 1989 Feb;55(2):217-31

Smith PJ, Paterson MC. Defective DNA repair and increased lethality in ataxia telangiectasia cells exposed to 4-nitroquinoline-1-oxide. Nature. 1980 Oct

Page 53: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 486

23;287(5784):747-9

Soresina A, Meini A, Lougaris V, Cattaneo G, Pellegrino S, Piane M, Darra F, Plebani A. Different clinical and immunological presentation of ataxia-telangiectasia within the same family. Neuropediatrics. 2008 Feb;39(1):43-5

Speit G, Trenz K, Schütz P, Bendix R, Dörk T. Mutagen sensitivity of human lymphoblastoid cells with a BRCA1 mutation in comparison to ataxia telangiectasia heterozygote cells. Cytogenet Cell Genet. 2000;91(1-4):261-6

Stern MH, Zhang FR, Thomas G, Griscelli C, Aurias A. Molecular characterization of ataxia telangiectasia T cell clones. III. Mapping the 14q32.1 distal breakpoint. Hum Genet. 1988 Dec;81(1):18-22

Su Y, Swift M. Mortality rates among carriers of ataxia-telangiectasia mutant alleles. Ann Intern Med. 2000 Nov 21;133(10):770-8

Sun Y, Xu Y, Roy K, Price BD. DNA damage-induced acetylation of lysine 3016 of ATM activates ATM kinase activity. Mol Cell Biol. 2007 Dec;27(24):8502-9

Swift M, Chase C. Cancer and cardiac deaths in obligatory ataxia-telangiectasia heterozygotes. Lancet. 1983 May 7;1(8332):1049-50

Swift M, Morrell D, Massey RB, Chase CL. Incidence of cancer in 161 families affected by ataxia-telangiectasia. N Engl J Med. 1991 Dec 26;325(26):1831-6

Takao N, Li Y, Yamamoto K. Protective roles for ATM in cellular response to oxidative stress. FEBS Lett. 2000 Apr 21;472(1):133-6

Taylor AM. Unrepaired DNA strand breaks in irradiated ataxia telangiectasia lymphocytes suggested from cytogenetic observations. Mutat Res. 1978 Jun;50(3):407-18

Taylor AM, Butterworth SV. Clonal evolution of T-cell chronic lymphocytic leukaemia in a patient with ataxia telangiectasia. Int J Cancer. 1986 Apr 15;37(4):511-6

Taylor AM, Edwards MJ. Malignancy, DNA damage and chromosomal aberrations in ataxia telangiectasia. IARC Sci Publ. 1982;(39):119-26

Taylor AM, Harnden DG, Arlett CF, Harcourt SA, Lehmann AR, Stevens S, Bridges BA. Ataxia telangiectasia: a human mutation with abnormal radiation sensitivity. Nature. 1975 Dec 4;258(5534):427-9

Taylor AM, Lam Z, Last JI, Byrd PJ. Ataxia telangiectasia: more variation at clinical and cellular levels. Clin Genet. 2015 Mar;87(3):199-208

Taylor AM, Metcalfe JA, Thick J, Mak YF. Leukemia and lymphoma in ataxia telangiectasia. Blood. 1996 Jan 15;87(2):423-38

Taylor AM, Rosney CM, Campbell JB. Unusual sensitivity of ataxia telangiectasia cells to bleomycin. Cancer Res. 1979 Mar;39(3):1046-50

Teo IA, Arlett CF. The response of a variety of human fibroblast cell strains to the lethal effects of alkylating agents. Carcinogenesis. 1982;3(1):33-7

Thompson LH. Recognition, signaling, and repair of DNA double-strand breaks produced by ionizing radiation in mammalian cells: the molecular choreography. Mutat Res. 2012 Oct-Dec;751(2):158-246

Till A, Lakhani R, Burnett SF, Subramani S. Pexophagy: the selective degradation of peroxisomes. Int J Cell Biol.

2012;2012:512721

Tripathi DN, Chowdhury R, Trudel LJ, Tee AR, Slack RS, Walker CL, Wogan GN. Reactive nitrogen species regulate autophagy through ATM-AMPK-TSC2-mediated suppression of mTORC1. Proc Natl Acad Sci U S A. 2013 Aug 6;110(32):E2950-7

Uziel T, Savitsky K, Platzer M, Ziv Y, Helbitz T, Nehls M, Boehm T, Rosenthal A, Shiloh Y, Rotman G. Genomic Organization of the ATM gene. Genomics. 1996 Apr 15;33(2):317-20

Valentin-Vega YA, Kastan MB. A new role for ATM: regulating mitochondrial function and mitophagy. Autophagy. 2012 May 1;8(5):840-1

Vaziri H. Critical telomere shortening regulated by the ataxia-telangiectasia gene acts as a DNA damage signal leading to activation of p53 protein and limited life-span of human diploid fibroblasts. A review. Biochemistry (Mosc). 1997 Nov;62(11):1306-10

Verhagen MM, Last JI, Hogervorst FB, Smeets DF, Roeleveld N, Verheijen F, Catsman-Berrevoets CE, Wulffraat NM, Cobben JM, Hiel J, Brunt ER, Peeters EA, Gómez Garcia EB, van der Knaap MS, Lincke CR, Laan LA, Tijssen MA, van Rijn MA, Majoor-Krakauer D, Visser M, van 't Veer LJ, Kleijer WJ, van de Warrenburg BP, Warris A, de Groot IJ, de Groot R, Broeks A, Preijers F, Kremer BH, Weemaes CM, Taylor MA, van Deuren M, Willemsen MA. Presence of ATM protein and residual kinase activity correlates with the phenotype in ataxia-telangiectasia: a genotype-phenotype study. Hum Mutat. 2012 Mar;33(3):561-71

Viniegra JG, Martínez N, Modirassari P, Hernández Losa J, Parada Cobo C, Sánchez-Arévalo Lobo VJ, Aceves Luquero CI, Alvarez-Vallina L, Ramón y Cajal S, Rojas JM, Sánchez-Prieto R. Full activation of PKB/Akt in response to insulin or ionizing radiation is mediated through ATM. J Biol Chem. 2005 Feb 11;280(6):4029-36

Vinters HV, Gatti RA, Rakic P. Sequence of cellular events in cerebellar ontogeny relevant to expression of neuronal abnormalities in ataxia-telangiectasia. Kroc Found Ser. 1985;19:233-55

Voss S, Pietzner J, Hoche F, Taylor AM, Last JI, Schubert R, Zielen S. Growth retardation and growth hormone deficiency in patients with Ataxia telangiectasia. Growth Factors. 2014 Jun;32(3-4):123-9

Wallace SS. Base excision repair: a critical player in many games. DNA Repair (Amst). 2014 Jul;19:14-26

Ward AJ, Olive PL, Burr AH, Rosin MP. Response of fibroblast cultures from ataxia-telangiectasia patients to reactive oxygen species generated during inflammatory reactions. Environ Mol Mutagen. 1994;24(2):103-11

Watters D, Kedar P, Spring K, Bjorkman J, Chen P, Gatei M, Birrell G, Garrone B, Srinivasa P, Crane DI, Lavin MF. Localization of a portion of extranuclear ATM to peroxisomes. J Biol Chem. 1999 Nov 26;274(48):34277-82

Watters DJ. Oxidative stress in ataxia telangiectasia. Redox Rep. 2003;8(1):23-9

Weaver M, Gatti RA. Lymphocyte subpopulations in ataxia-telangiectasia. Kroc Found Ser. 1985;19:309-14

Webb CJ, Wu Y, Zakian VA. DNA repair at telomeres: keeping the ends intact. Cold Spring Harb Perspect Biol. 2013 Jun 1;5(6)

Weiss B, Krauthammer A, Soudack M, Lahad A, Sarouk I, Somech R, Heimer G, Ben-Zeev B, Nissenkorn A. Liver

Page 54: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Ataxia telangiectasia (A-T) Shiloh Y

Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12) 487

Disease in Pediatric Patients With Ataxia Telangiectasia: A Novel Report. J Pediatr Gastroenterol Nutr. 2016 Apr;62(4):550-5

Westphal CH. Cell-cycle signaling: Atm displays its many talents. Curr Biol. 1997 Dec 1;7(12):R789-92

Wood LD, Halvorsen TL, Dhar S, Baur JA, Pandita RK, Wright WE, Hande MP, Calaf G, Hei TK, Levine F, Shay JW, Wang JJ, Pandita TK. Characterization of ataxia telangiectasia fibroblasts with extended life-span through telomerase expression. Oncogene. 2001 Jan 18;20(3):278-88

Worth PF, Srinivasan V, Smith A, Last JI, Wootton LL, Biggs PM, Davies NP, Carney EF, Byrd PJ, Taylor AM. Very mild presentation in adult with classical cellular phenotype of ataxia telangiectasia. Mov Disord. 2013 Apr;28(4):524-8

Wu D, Yang H, Xiang W, Zhou L, Shi M, Julies G, Laplante JM, Ballard BR, Guo Z. Heterozygous mutation of ataxia-telangiectasia mutated gene aggravates hypercholesterolemia in apoE-deficient mice. J Lipid Res. 2005 Jul;46(7):1380-7

Xia SJ, Shammas MA, Shmookler Reis RJ. Reduced telomere length in ataxia-telangiectasia fibroblasts. Mutat Res. 1996 Sep 2;364(1):1-11

Yamashita A. Role of SMG-1-mediated Upf1 phosphorylation in mammalian nonsense-mediated mRNA decay. Genes Cells. 2013 Mar;18(3):161-75

Yang DQ, Kastan MB. Participation of ATM in insulin signalling through phosphorylation of eIF-4E-binding protein 1. Nat Cell Biol. 2000 Dec;2(12):893-8

Yi M, Rosin MP, Anderson CK. Response of fibroblast cultures from ataxia-telangiectasia patients to oxidative stress. Cancer Lett. 1990 Oct 8;54(1-2):43-50

Zhang J, Kim J, Alexander A, Cai S, Tripathi DN, Dere R, Tee AR, Tait-Mulder J, Di Nardo A, Han JM, Kwiatkowski

E, Dunlop EA, Dodd KM, Folkerth RD, Faust PL, Kastan MB, Sahin M, Walker CL. A tuberous sclerosis complex signalling node at the peroxisome regulates mTORC1 and autophagy in response to ROS. Nat Cell Biol. 2013 Oct;15(10):1186-96

Zhang J, Tripathi DN, Jing J, Alexander A, Kim J, Powell RT, Dere R, Tait-Mulder J, Lee JH, Paull TT, Pandita RK, Charaka VK, Pandita TK, Kastan MB, Walker CL. ATM functions at the peroxisome to induce pexophagy in response to ROS. Nat Cell Biol. 2015 Oct;17(10):1259-1269

Zhang N, Song Q, Lu H, Lavin MF. Induction of p53 and increased sensitivity to cisplatin in ataxia-telangiectasia cells. Oncogene. 1996 Aug 1;13(3):655-9

Ziv S, Brenner O, Amariglio N, Smorodinsky NI, Galron R, Carrion DV, Zhang W, Sharma GG, Pandita RK, Agarwal M, Elkon R, Katzin N, Bar-Am I, Pandita TK, Kucherlapati R, Rechavi G, Shiloh Y, Barzilai A. Impaired genomic stability and increased oxidative stress exacerbate different features of Ataxia-telangiectasia. Hum Mol Genet. 2005 Oct 1;14(19):2929-43

Ziv Y, Bar-Shira A, Pecker I, Russell P, Jorgensen TJ, Tsarfati I, Shiloh Y. Recombinant ATM protein complements the cellular A-T phenotype. Oncogene. 1997 Jul 10;15(2):159-67

Zolner AE, Abdou I, Ye R, Mani RS, Fanta M, Yu Y, Douglas P, Tahbaz N, Fang S, Dobbs T, Wang C, Morrice N, Hendzel MJ, Weinfeld M, Lees-Miller SP. Phosphorylation of polynucleotide kinase/ phosphatase by DNA-dependent protein kinase and ataxia-telangiectasia mutated regulates its association with sites of DNA damage. Nucleic Acids Res. 2011 Nov;39(21):9224-37

This article should be referenced as such:

Shiloh Y. Ataxia telangiectasia (A-T). Atlas Genet Cytogenet Oncol Haematol. 2017; 21(12):475-487.

Page 55: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS

OPEN ACCESS JOURNAL

Instructions to Authors

Policies- Instructions to Authors

See http://documents.irevues.inist.fr/bitstream/handle/2042/48486/Instructions-to-authors.pdf

Manuscripts submitted to the Atlas must be submitted solely to the Atlas.

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).

Deep Insights are written as traditional papers, made of paragraphs with headings, at the author's convenience.

Case Reports in haematological malignancies are dedicated to recurrent –but rare- chromosomes abnormalities in

leukaemias/lymphomas; see http://atlasgeneticsoncology.org//BackpageAuthors.html#CASE_REPORTS .

It is mandatory to use the specific "Submissionformfor Case reports":

see http://AtlasGeneticsOncology.org/Reports/Case_Report_Submission.html.

Educational Items must be didactic, give full information and be accompanied with iconography.

Research articlesThe Atlas of Genetics and Cytogenetics in Oncology and Haematology does not publish research articles.

Authorship All authors should qualify for authorship according to the ICMJE criteria.

Editorial Ethics see http://documents.irevues.inist.fr/bitstream/handle/2042/56068/Policies-editorial-ethics.pdf for: Peer Review Process /

Commissioned papers vs Unsolicited papers /Responsibility for the reviewers / Editorial responsibilities / Conflict of interest-

Competing interests/ Privacy and Confidentiality - Iconography / Protection of Human Subjects and Animals in Research /

Duplicate Publication/ Plagiarism / Retracting a publication

Subscription The Atlas is FREE!

Costs/Page Charge There is NO page charge.

PubMed Central Once the paper online, authors are encouraged to send their manuscript to PubMed Central

http://www.ncbi.nlm.nih.gov/pmc/ with reference to the original paper in the Atlas in

http://documents.irevues.inist.fr/handle/2042/15655

Corporate patronage, sponsorship and advertising

Enquiries should be addressed to [email protected].

Rules, Copyright Notice and Disclaimer

http://documents.irevues.inist.fr/bitstream/handle/2042/48487/Copyright-sponsorship.pdf

Property As "cards" are to evolve with further improvements and updates from various contributors, the property of the

cards belongs to the editor, and modifications will be made without authorization from the previous contributor (who may,

nonetheless, be asked for refereeing); contributors are listed in an edit history manner. Authors keep the rights to use further

the content of their papers published in the Atlas, provided that the source is cited.

Copyright The information in the Atlas of Genetics and Cytogenetics in Oncology and Haematology is issued for general

distribution. All rights are reserved. The information presented is protected under international conventions and under

national laws on copyright and neighbouring rights. Commercial use is totally forbidden. Information extracted from the

Atlas may be reviewed, reproduced or translated for research or private study but not for sale or for use in conjunction with

commercial purposes. Any use of information from the Atlas should be accompanied by an acknowledgment of the Atlas as

the source, citing the uniform resource locator (URL) of the article and/or the article reference, according to the Vancouver

convention. Reference to any specific commercial products, process, or service by trade name, trademark, manufacturer, or

otherwise, does not necessarily constitute or imply its endorsement, recommendation, or favouring. The views and opinions

of contributors and authors expressed herein do not necessarily state or reflect those of the Atlas editorial staff or of the web

site holder, and shall not be used for advertising or product endorsement purposes. The Atlas does not make any warranty,

express or implied, including the warranties of merchantability and fitness for a particular purpose, or assumes any legal

liability or responsibility for the accuracy, completeness, or usefulness of any information, and shall not be liable whatsoever

for any damages incurred as a result of its use. In particular, information presented in the Atlas is only for research purpose,

and shall not be used for diagnosis or treatment purposes. No responsibility is assumed for any injury and/or damage to

persons or property for any use or operation of any methods products, instructions or ideas contained in the material herein.

See also "Uniform Requirements for Manuscripts Submitted to Biomedical Journals: Writing and Editing for Biomedical

Publication - Updated October 2004": http://www.icmje.org.

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

Page 56: vol 21 12 2017 - documents.irevues.inist.frdocuments.irevues.inist.fr/bitstream/handle/2042/69025/vol_21_12_2017.pdfin Oncology and Haematology The PDF version of the Atlas of Genetics

Atlas of Genetics and Cytogenetics

in Oncology and Haematology

INIST-CNRS

OPEN ACCESS JOURNAL