volume 16 - volume 1 -number 3number 1 may - september 1997...

91
Volume 1 - Number 1 May - September 1997 Volume 16 - Number 3 March 2012

Upload: hoanghuong

Post on 19-Jul-2018

226 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Volume 1 - Number 1 May - September 1997

Volume 16 - Number 3 March 2012

Page 2: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16
Page 3: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

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

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

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

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

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Scope

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

open access, devoted to genes, cytogenetics, and clinical entities in cancer, and cancer-prone diseases.

It presents structured review articles ("cards") on genes, leukaemias, solid tumours, cancer-prone diseases, more

traditional review articles on these and also on surrounding topics ("deep insights"), case reports in hematology,

and educational items in the various related topics for students in Medicine and in Sciences.

Editorial correspondance

Jean-Loup Huret Genetics, Department of Medical Information,

University Hospital

F-86021 Poitiers, France

tel +33 5 49 44 45 46 or +33 5 49 45 47 67

[email protected] or [email protected]

Staff Mohammad Ahmad, Mélanie Arsaban, Marie-Christine Jacquemot-Perbal, Maureen Labarussias, Vanessa Le

Berre, Anne Malo, Catherine Morel-Pair, Laurent Rassinoux, Alain Zasadzinski.

Philippe Dessen is the Database Director, and Alain Bernheim the Chairman of the on-line version (Gustave

Roussy Institute – Villejuif – France).

The Atlas of Genetics and Cytogenetics in Oncology and Haematology (ISSN 1768-3262) is published 12 times

a year by ARMGHM, a non profit organisation, and by the INstitute for Scientific and Technical Information of

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

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

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

Page 4: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16
Page 5: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3)

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Editor

Jean-Loup Huret

(Poitiers, France)

Editorial Board

Sreeparna Banerjee (Ankara, Turkey) Solid Tumours Section

Alessandro Beghini (Milan, Italy) Genes Section

Anne von Bergh (Rotterdam, The Netherlands) Genes / Leukaemia Sections

Judith Bovée (Leiden, The Netherlands) Solid Tumours Section

Vasantha Brito-Babapulle (London, UK) Leukaemia Section

Charles Buys (Groningen, The Netherlands) Deep Insights Section

Anne Marie Capodano (Marseille, France) Solid Tumours Section

Fei Chen (Morgantown, West Virginia) Genes / Deep Insights Sections

Antonio Cuneo (Ferrara, Italy) Leukaemia Section

Paola Dal Cin (Boston, Massachussetts) Genes / Solid Tumours Section

Louis Dallaire (Montreal, Canada) Education Section

Brigitte Debuire (Villejuif, France) Deep Insights Section

François Desangles (Paris, France) Leukaemia / Solid Tumours Sections

Enric Domingo-Villanueva (London, UK) Solid Tumours Section

Ayse Erson (Ankara, Turkey) Solid Tumours Section

Richard Gatti (Los Angeles, California) Cancer-Prone Diseases / Deep Insights Sections

Ad Geurts van Kessel (Nijmegen, The Netherlands) Cancer-Prone Diseases Section

Oskar Haas (Vienna, Austria) Genes / Leukaemia Sections

Anne Hagemeijer (Leuven, Belgium) Deep Insights Section

Nyla Heerema (Colombus, Ohio) Leukaemia Section

Jim Heighway (Liverpool, UK) Genes / Deep Insights Sections

Sakari Knuutila (Helsinki, Finland) Deep Insights Section

Lidia Larizza (Milano, Italy) Solid Tumours Section

Lisa Lee-Jones (Newcastle, UK) Solid Tumours Section

Edmond Ma (Hong Kong, China) Leukaemia Section

Roderick McLeod (Braunschweig, Germany) Deep Insights / Education Sections

Cristina Mecucci (Perugia, Italy) Genes / Leukaemia Sections

Yasmin Mehraein (Homburg, Germany) Cancer-Prone Diseases Section

Fredrik Mertens (Lund, Sweden) Solid Tumours Section

Konstantin Miller (Hannover, Germany) Education Section

Felix Mitelman (Lund, Sweden) Deep Insights Section

Hossain Mossafa (Cergy Pontoise, France) Leukaemia Section

Stefan Nagel (Braunschweig, Germany) Deep Insights / Education Sections

Florence Pedeutour (Nice, France) Genes / Solid Tumours Sections

Elizabeth Petty (Ann Harbor, Michigan) Deep Insights Section

Susana Raimondi (Memphis, Tennesse) Genes / Leukaemia Section

Mariano Rocchi (Bari, Italy) Genes Section

Alain Sarasin (Villejuif, France) Cancer-Prone Diseases Section

Albert Schinzel (Schwerzenbach, Switzerland) Education Section

Clelia Storlazzi (Bari, Italy) Genes Section

Sabine Strehl (Vienna, Austria) Genes / Leukaemia Sections

Nancy Uhrhammer (Clermont Ferrand, France) Genes / Cancer-Prone Diseases Sections

Dan Van Dyke (Rochester, Minnesota) Education Section

Roberta Vanni (Montserrato, Italy) Solid Tumours Section

Franck Viguié (Paris, France) Leukaemia Section

José Luis Vizmanos (Pamplona, Spain) Leukaemia Section

Thomas Wan (Hong Kong, China) Genes / Leukaemia Sections

Page 6: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3)

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Volume 16, Number 3, March 2012

Table of contents

Editorial

Why breast cancer and prostate cancer are so frequent? A new genetic mechanism, involving hormones and viruses 185 Jean-Loup Huret

Gene Section

CUX1 (cut-like homeobox 1) 189 Benjamin Kühnemuth, Patrick Michl

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) 194 June L Traicoff, Stephen M Hewitt, Joon-Yong Chung

MYEOV (myeloma overexpressed (in a subset of t(11;14) positive multiple myelomas)) 203 Jérôme Moreaux

PCNA (proliferating cell nuclear antigen) 206 Ivaylo Stoimenov, Thomas Helleday

RASSF5 (Ras association (RalGDS/AF-6) domain family member 5) 210 Lee Schmidt, Geoffrey J Clark

RGS17 (regulator of G-protein signaling 17) 214 Chenguang Li, Lei Wang, Yihua Sun, Haiquan Chen

SLC39A1 (solute carrier family 39 (zinc transporter), member 1) 216 Renty B Franklin, Leslie C Costello

CBX7 (chromobox homolog 7) 218 Ana O'Loghlen, Jesus Gil

RPRM (reprimo, TP53 dependent G2 arrest mediator candidate) 221 Alejandro H Corvalan, Veronica A Torres

VMP1 (vacuole membrane protein 1) 223 Alejandro Ropolo, Andrea Lo Ré, María Inés Vaccaro

XPO1 (exportin 1 (CRM1 homolog, yeast)) 226 Alessandra Ruggiero, Maria Giubettini, Patrizia Lavia

Leukaemia Section

t(11;18)(p15;q12) 231 Jean-Loup Huret

t(11;21)(q21;q22) 232 Jean-Loup Huret

t(8;17)(q24;q22) ???BCL3/MYC 234 Jean-Loup Huret

Page 7: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

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

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3)

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Deep Insight Section

Plasticity and Tumorigenicity 236 Elena Campos-Sanchez, Isidro Sanchez-Garcia, Cesar Cobaleda

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function 251 Xiaodong Lu, Wenxin Qin

Case Report Section

A case of Acute Lymphoblastic Leukemia with rare t(11;22)(q23;q13) 259 Jill D Kremer, Anwar N Mohamed

Insertion as an alternative mechanism of CBFB-MYH11 gene fusion in a new case of acute myeloid leukemia with an abnormal chromosome 16 262 Yaser Hussein, Vandana Kulkarni, Anwar N Mohamed

Page 8: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16
Page 9: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Editorial

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 185

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Why breast cancer and prostate cancer are so frequent? A new genetic mechanism, involving hormones and viruses Jean-Loup Huret

Atlas of Genetics and Cytogenetics in Oncology and Haematology Unit, University of Poitiers,

Department of Medical Information, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH)

This work was presented at the 8th European Cytogenetics Conference, Porto, 2-5 July 2011

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Abbreviated title: Hormones and viruses in breast and prostate cancers

Abstract Prostate and breast cancers, which are hormone-dependant cancers, are highly frequent (up to 1/3 of cancers in

male, 1/3 of cancers in female patients) and often multifocal. Multifocality, in particular, rings the bell of a

specific carcinogenetic agent (such as heritability is in retinoblastoma). Here we point a highly uneven

distribution of genetic events (translocation breakpoints) in prostate and breast cancers, which favours the

hypothesis of cooperation between viruses and hormone receptors to cut DNA at high rates, delete parts of it,

facilitating oncogene translocations and oncogenesis. If our hypothesis turns out to be right, vaccination against

breast cancer and prostate cancer might notably diminish the frequency of these cancers.

Looking at chromosomal rearrangements in

prostate adenocarcinoma, using the Atlas of

Genetics and Cytogenetics in Oncology and

Haematology (Huret et al., 2003), the Mitelman

Database (Mitelman et al., 2012), and Goldenpath

(Fujita et al., 2011)

(http://atlasgeneticsoncology.org/, http://cgap.nci.

nih.gov/Chromosomes/Mitelman, and

http://genome.ucsc.edu/ respectively), we noted

that, out of 42 relevant rearrangements available in

early 2011, 10 exhibited the two partner

breakpoints in the same chromosome band (e.g.

5q31 fused to 5q31). Given that 312 chromosome

bands are at risk of rearrangement, the probability

of the observed distribution is p=1.5 x 10-16

(binomial distribution). Such a non random close

proximity of the two partner breakpoints is even

more striking at the base level (e.g. 6 kb in the

5q31-5q31 rearrangement; see Table 1).

The situation is very similar with breast

adenocarcinoma: Of 39 rearrangements, 20 have

breakpoints in close proximity (Table 2). The

probability of such an event is p=6 x 10-39

. We

herein uncover a highly significant non random

distribution of breakpoints in breast and prostate

cancer DNA rearragements and microdeletions (by

comparison, only 1 of 10 rearrangements in lung

adenocarcinoma exhibit both breakpoints in the

same band (R3HDM2/NFE2 in the

del(12)(q13q13)). A bias of publications may exist,

but cannot account for such a highly unexpected

finding.

Page 10: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Why breast cancer and prostate cancer are so frequent? A new genetic mechanism, involving hormones and viruses

Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 186

Table 1. Distance in base pairs in rearrangements with the two breakpoints on the same chromosomal

band in prostate adenocarcinoma

fusion gene rearrangement coordinates 1st gene coordinates 2nd gene distance

WDR55/DND1 t(5;5)(q31;q31) 140044384 140050382 6 kb

MBTPS2/YY2 t(X;X)(p22;p22) 21857656 21874603 17 kb

ZNF649/ZNF577 t(19;19)(q13;q13) 52392489 52374553 18 kb

C19ORF25/APC2 t(19;19)(p13;p13) 1473201 1450148 23 kb

SLC45A3/ELK4 t(1;1)(q32;q32) 205626981 205588398 39 kb

USP10/ZDHHC7 del(16)(q24q24) 84733555 85008067 275 kb

RERE/PIK3CD del(1)(p36p36) 8412466 9711790 1,3 Mb

HJURP/EIF4E2 t(2;2)(q37;q37) 234745487 233415357 1,3 Mb

TMPRSS2/ERG t(21;21)(q22;q22) 42836479 39751952 3 Mb

PIK3C2A/TEAD1 del(11)(p15p15) 17108126 12695969 4,4 Mb

Note. These fusion genes were first described in Tomlins et al., 2005; Maher et al., 2009a; et al., 2009b;

Rickman et al., 2009.

Table 2. Distance in base pairs in rearrangements with the two breakpoints on the same chromosomal

band in breast adenocarcinoma

fusion gene rearrangement coordinates 1st gene coordinates 2nd gene distance

EFTUD2/KIF18B del(17)(q21q21) 42927655 43003449 76 kb

PLXND1/TMCC1 del(3)(q22q22) 129274056 129366637 93 kb

PAPOLA/AK7 del(14)(q32q32) 96968720 96858448 110 kb

SEPT8/AFF4 del(5)(q31q31) 132086509 132211072 125 kb

SLC26A6/PRKAR2A del(3)(p21p21) 48663158 48788093 125 kb

AC141586/CCNF del(16)(p13p13) 2653351 2479395 174 kb

ERO1L/FERMT2 del(14)(q22q22) 53108607 53323990 215 kb

HN1/USH1G del(17)(q25q25) 73131344 72912176 219 kb

HMGXB3/PPARGC1B del(5)(q32q32) 149380169 149109864 270 kb

INTS4/GAB2 del(11)(q14q14) 77589768 77926343 337 kb

PLA2R1/RBMS1 del(2)(q24q24) 160798012 161128663 330 kb

RASA2/ACPL2 del(3)(q23q23) 141205926 140950682 255 kb

BC017255/TMEM49 t(17;17)(q22;q23) 57183959 57784863 600 kb

LDHC/SERGEF del(11)(p15p15) 18433853 17809599 624 kb

KCNQ5/RIMS1 del(6)(q13q13) 73331571 72596650 735 kb

MYO9B/FCHO1 del(19)(p13p13) 17186591 17858527 672 kb

STRADB/NOP58 del(2)(q33q33) 202316392 203130515 814 kb

SMYD3/ZNF695 del(1)(q44q44) 245912645 247148625 1,2 Mb

CYTH1/PRPSAP1 del(17)(q25q25) 76670131 74306868 2,4 Mb

RAF1/DAZL t(3;3)(p24;p25) 12625102 16628303 4 Mb

Note. These fusion genes were first described in Maher et al., 2009b; Stephens, P.J. et al., 2009.

Page 11: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Why breast cancer and prostate cancer are so frequent? A new genetic mechanism, involving hormones and viruses

Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 187

Prostate and breast share two specificities: high

frequency of cancer, cancers that are frequently

multifocal. Prostate cancer represents up to 1/3 of

cancers in men, breast cancer 1/3 of cancers in

female patients in western countries. Prostate

cancer is a multicentric tumour in 75-80% of cases.

At autopsy, up to 30 to 70 % of men aged 70-80

years have cancerous foci in the prostate; after 80,

90% have hyperplasia, and more than 70% have a

neoplastic disease. Breast adenocarcinoma is a

multifocal tumour in at least 10-15% of cases.

Multifocality, in particular, rings the bell of a

specific "helper" carcinogenetic agent (such as

heritability in retinoblastoma).

Androgen and estrogen receptors are crucial for the

normal development as well as for cancer

progression of the target organs, respectively the

androgen receptor (AR) for the prostate and the

estrogen receptor (ER) for the breast. Hormone

receptors bind DNA at specific motifs called

hormone responsive elements: AR binds to a

specific TGT/AGGGA/T motif and ER binds to the

consensus core element AGGGTCA.

Some retroviruses, such as the mouse mammary

tumour virus (MMTV) and its human homolog

HMTV contain hormone responsive elements (Cato

et al., 1987; Pogo et al., 2010). A retrovirus

containing androgen response elements remains to

be found, since the candidate, XMRV, appears now

to be of experimental recombinant origin (Paprotka

et al., 2011).

Both androgen and estrogen receptors induce DNA

double strands breaks (DSBs) (Lin et al., 2009,

Williamson and Lees-Miller, 2011); such DSBs can

seed the formation of genomic/chromosome

rearrangements, and, at random, the possible

junction of oncogene loci normally separated (Lin

et al., 2009, Mani et al., 2009). It has been found

that androgen receptor signaling and topoisomerase

II mediate DSBs and TMPRSS2-ERG

rearrangements in prostate cancer (Haffner et al.,

2010). In case of a TMPRSS2-ERG rearrangement,

a deletion of 3 mb occurs (Table 1).

HMTV is found in 40% of breast cancers in

American women, in 60% of milk from patients

with a history of breast cancer and in 5% of milk

from normal subjects (cited in Pogo et al., 2010).

HMTV/MMTV sequences and enhanced Wnt-1

expression were found in breast ductal carcinoma

(Lawson et al., 2010). However, other viruses may

be implicated, and it must be kept in mind that

HBV, HPV, EBV, CMV have also been found to be

associated with breast cancer.

We Hypothesize that HMTV and other viruses can

integrate in the cell genome of breast or prostatic

cells as proviruses in multiple sites, at random.

Hormone receptors (ER or AR) would bind DNA at

hormone responsive elements sites, including those

added in numerous copies by the proviruses.

Hormone receptors would induce DNA breaks, as

usually, DNA deletions would occur in a certain

percentage of cases, facilitating oncogene

translocations. Not all breaks, not all fusion genes

are pathogenetically significant, but the cooperation

of viruses and hormone receptors (together with

other factors: genotoxic stress, inflammation...) to

cut and saw DNA would greatly enhance the risk

for an oncogenic event to occur.

There is no reason a priori for an organ to fall "too"

frequently into a cancerous process in the absence

of an additional carcinogenetic agent. In skin

cancer, ultraviolet radiation is the known major

helper, and lung cancer is a rare cancer without a

history of smoking. The "abnormally" high

frequency of prostate and breast cancers may well

be due to this association in crime of viruses and

hormone receptors.

If our hypothesis turns to be right, inhibitors for

androgens/estrogens receptors and vaccination

against breast cancer prostate cancer (as it is now

available for cervix cancer / papilloma virus) might

abrogate an important proportion of DSBs, and

notably diminish the frequency of these cancers

and/or facilitate their cure.

References Cato AC, Henderson D, Pont H. The hormone response element of the mouse mammary tumour virus DNA mediates the progestin and androgen induction of transcription in the proviral long terminal repeat region. EMBO J 1987; 6:363-638.

Fujita PA, Rhead B, Zweig AS, Hinrichs AS, Karolchik D, Cline MS, Goldman M, Barber GP, Clawson H, Coelho A, Diekhans M, Dreszer TR, Giardine BM, Harte RA, Hillman-Jackson J, Hsu F, Kirkup V, Kuhn RM, Learned K, Li CH, Meyer LR, Pohl A, Raney BJ, Rosenbloom KR, Smith KE, Haussler D, Kent WJ. The UCSC Genome Browser database: update 2011. Nucleic Acids Res 2011; 39:D876-882

Haffner MC, Aryee MJ, Toubaji A, Esopi DM, Albadine R, Gurel B, Isaacs WB, Bova GS, Liu W, Xu J, Meeker AK, Netto G, De Marzo AM, Nelson WG, Yegnasubramanian S. Androgen-induced TOP2B-mediated double-strand breaks and prostate cancer gene rearrangements. Nat Genet 2010; 42:668-675.

Huret JL, Dessen P, Bernheim A. An Internet database on genetics in oncology. Oncogene 2003; 22:1907.

Lawson JS, Glenn WK, Salmons B, Ye Y, Heng B, Moody P, Johal H, Rawlinson WD, Delprado W, Lutze-Mann L, Whitaker NJ. Mouse mammary tumor virus-like sequences in human breast cancer. Cancer Res 2010; 70:3576-3585.

Lin C, Yang L, Tanasa B, Hutt K, Ju BG, Ohgi K, Zhang J, Rose DW, Fu XD, Glass CK, Rosenfeld MG. Nuclear receptor-induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell 2009; 139:1069-1083.

Maher CA, Kumar-Sinha C, Cao X, Kalyana-Sundaram S, Han B, Jing X, Sam L, Barrette T, Palanisamy N, Chinnaiyan AM. Transcriptome sequencing to detect gene fusions in cancer. Nature 2009a; 458:97-101.

Maher CA, Palanisamy N, Brenner JC, Cao X, Kalyana-Sundaram S, Luo S, Khrebtukova I, Barrette TR, Grasso C, Yu J, Lonigro RJ, Schroth G, Kumar-Sinha C, Chinnaiyan AM. Chimeric transcript discovery by paired-

Page 12: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Why breast cancer and prostate cancer are so frequent? A new genetic mechanism, involving hormones and viruses

Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 188

end transcriptome sequencing. Proc Natl Acad Sci USA 2009b; 106:12353-12358.

Mani RS, Tomlins SA, Callahan K, Ghosh A, Nyati MK, Varambally S, Palanisamy N, Chinnaiyan AM. Induced chromosomal proximity and gene fusions in prostate cancer. Science 2009; 326:1230-1232.

Mitelman F, Johansson B. and Mertens F. (Eds.). 2011. Mitelman Database of Chromosome Aberrations and Gene Fusions in Cancer.

Paprotka T, Delviks-Frankenberry KA, Cingöz O, Martinez A, Kung HJ, Tepper CG, Hu WS, Fivash MJ Jr, Coffin JM, Pathak VK. Recombinant Origin of the Retrovirus XMRV. Science 2011; 333:97-101.

Pogo BG, Holland JF, Levine PH. Human mammary tumor virus in inflammatory breast cancer. Cancer 2010; 116:2741-2744.

Rickman DS, Pflueger D, Moss B, VanDoren VE, Chen CX, de la Taille A, Kuefer R, Tewari AK, Setlur SR, Demichelis F, Rubin MA. SLC45A3-ELK4 is a novel and frequent erythroblast transformation-specific fusion transcript in prostate cancer. Cancer Res 2009; 69: 2734-2738.

Stephens PJ, McBride DJ, Lin ML,Varela I, Pleasance ED, Simpson JT, Stebbings LA, Leroy C, Edkins S, Mudie LJ,

Greenman CD, Jia M, Latimer C, Teague JW, Lau KW, Burton J, Quail MA, Swerdlow H, Churcher C, Natrajan R, Sieuwerts AM, Martens JW, Silver DP, Langerød A, Russnes HE, Foekens JA, Reis-Filho JS, van 't Veer L, Richardson AL, Børresen-Dale AL, Campbell PJ, Futreal PA, Stratton MR. Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature 2009; 462:1005-1010.

Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, Varambally S, Cao X, Tchinda J, Kuefer R, Lee C, Montie JE, Shah RB, Pienta KJ, Rubin MA, Chinnaiyan AM. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 2005; 310:644-648.

Williamson LM, Lees-Miller SP. Estrogen receptor α-mediated transcription induces cell cycle-dependent DNA double-strand breaks. Carcinogenesis 2011; 32:279-285.

This article should be referenced as such:

Huret JL. Why breast cancer and prostate cancer are so frequent? A new genetic mechanism, involving hormones and viruses. Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):185-188.

Page 13: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 189

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

CUX1 (cut-like homeobox 1) Benjamin Kühnemuth, Patrick Michl

Department of Gastroenterology and Endocrinology, University of Marburg, Marburg, Germany (BK,

PM)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/CUX1ID403ch7q22.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI CUX1ID403ch7q22.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: CASP, CDP, CDP/Cut, CDP1,

COY1, CUTL1, CUX, Clox, Cux/CDP, FLJ31745,

GOLIM6, Nbla10317, p100, p110, p200, p75

HGNC (Hugo): CUX1

Location: 7q22.1

DNA/RNA

Description

The human CUX1 gene is located on chromosome

7q22 (Scherer et al., 1993). It comprises 33 exons

and spans 468 kb.

Five alternative splice variants have been identified.

Most of the splicing sites are located in the regions

downstream of exon 14 and 15 (Rong Zeng et al.,

2000). Two alternative sites for transcript

termination have been identified. Termination at

UGA in exon 24 leads to production of CUX1

mRNA comprising exon 1-24. Elongation up to

exon 33 results in alternative splicing and the

production of CASP mRNA comprising exon 1-15

and 25-33 (Lievens et al., 1997; Rong Zeng et al.,

2000).

The first transcriptional start site is located in exon

1 but transcription can be initiated at several sites in

a 200 bp region upstream of exon 1 (Rong Zeng et

al., 2000). Initiation within intron 20 leads to

production of an mRNA coding for the shortened

p75 isoform (Goulet et al., 2002).

Several putative translation initiation codons can be

found in exon 1 but ATG at position 550 has been

described as the predominant initiation site (Rong

Zeng et al., 2000).

Protein

Description

The human full length CUX1 protein (p200)

consists of 1505 amino acids and contains four

DNA binding domains: three CUT-repeats and one

CUT-homeodomain (Harada et al., 1994).

Several shortened CUX1 isoforms have been

described that are named according to their

molecular weight. CUX1 p75 is the product of a

shortened mRNA that is generated by the use of an

alternative transcription start site in exon 20 (Rong

Zeng et al., 2000; Goulet et al., 2002). CUX1 p150,

p110, p90 and p80 are generated by proteolytic

processing of the full length protein by a nuclear

isoform of Cathepsin L and other not yet identified

proteases such as caspases (Goulet et al., 2004;

Goulet et al., 2006; Maitra et al., 2006; Truscott et

al., 2007).

The presence of DNA binding domains in the

CUX1 isoforms determines their interaction with

DNA and their transcriptional activity. The full

length protein p200 shows unstable DNA binding,

carries the CCAAT-displacement activity and

functions predominantly as a transcriptional

repressor. In contrast, the p110, p90, p80 and p75

isoforms show stable DNA binding and function

both as transcriptional repressors or activators

(Truscott et al., 2004; Goulet et al., 2002; Goulet et

al., 2006; Moon et al., 2001). According to Maitra

et al., the p150 isoform is incapable of DNA

binding (Maitra et al., 2006).

Several posttranslational modifications are known

to modulate the DNA binding activities of the

CUX1 proteins.

Page 14: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

CUX1 (cut-like homeobox 1) Kühnemuth B, Michl P

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 190

Cux1 isoforms. The p75 isoform is the product of a shortened mRNA that is generated by the use of an alternative

transcriptional start site. In contrast, the p150, p110, p90 and p80 isoforms are produced by proteolytic processing of the full length protein (p200). CR = cut repeat; HD = homeodomain.

Protein kinase C and Casein kinase II are able to

phosphorylate serine or threonine residues within

the cut repeats (Coqueret et al., 1998b; Li et al.,

2007). Protein kinase A and cyclin A/Cdk1

phosphorylate specific serine residues in a region

between the Cut repeat 3 and the homeodomain

(Michl et al., 2006; Santaguida et al., 2001). PCAF

acetyl-transferase is able to acetylate CUX1 on a

lysine residue in the homeodomain (Li et al., 2000).

Both, phosphorylation and acetylation have been

shown to inhibit CUX1 DNA binding (Sansregret et

al., 2010; Li et al., 2000). Consistent with this,

dephosphorylation by Cdc 25A phosphatase is able

to increase DNA binding of CUX1 (Coqueret et al,

1998a).

Expression

Early studies suggested that in mammalian cells,

CUX1 represses genes that are upregulated in

differentiated tissues. Furthermore, the expression

of CUX1 might be restricted to proliferating and

undifferentiated cells and is inversely related to the

degree of differentiation (vanden Heuvel et al.,

1996; Pattison et al., 1997; van Gurp et al., 1999).

More recently however, studies in mice revealed

that CUX1 is also expressed in terminally

differentiated cells of many tissues (Khanna-Gupta

et al., 2001; Ellis et al., 2001).

Increased CUX1 expression was found in various

tumour types including multiple myelomas, acute

lymphoblastic leukaemia, breast carcinoma and

pancreatic cancer (De Vos et al., 2002; Tsutsumi et

al., 2003; Michl et al., 2005; Ripka et al., 2007).

It has been shown that the cellular expression of

CUX1 mRNA and protein is elevated following

TGF-beta stimulation in many cell types including

fibroblasts, pancreatic cancer cells, breast cancer

cells and malignant plasma cells (Fragiadaki et al.,

2011; Michl et al., 2005; De Vos et al., 2002). This

regulation of CUX1 expression by TGF-beta is

probably mediated by p38MAPK and Smad4

signalling (Michl et al., 2005).

Localisation

Studies indicate that phosphorylated CUX1 is

preferentially localized in the cytoplasm whereas

dephosphorylation leads to translocation into the

nucleus (Sansregret et al., 2010).

Function

The vast majority of studies describes CUX1 as a

transcriptional repressor (Lievens et al., 1995; Ai et

al., 1999; Catt et al., 1999a; Catt et al., 1999b; Ueda

et al., 2007). The repressor activity can be mediated

by competition for DNA binding sites with

transcriptional activators (Kim et al., 1997; Stünkel

et al., 2000), by recruitment of histone deacetylases

(Li et al., 1999) or by recruitment of histone lysine

methyltransferases (Nishio and Walsh, 2004).

CUX1 may also negatively regulate gene

expression by binding to matrix attachment regions

and by modulating their association with the

nuclear scaffold (Banan et al., 1997; Stünkel et al.,

2000; Goebel et al., 2002; Kaul-Ghanekar et al.,

2004). In contrast, the mechanisms underlying its

effects on transcriptional activation are less well

understood.

CUX1 is involved in at least three cellular

processes important for cancer progression: cell

proliferation, cell motility/invasiveness and

apoptosis.

Proliferation Studies indicate that the pro-proliferative effects of

CUX1 are mainly mediated by the p110 isoform.

This isoform is produced by proteolytic cleavage of

the full length protein occuring during G1/S-

transition in the cell cycle (Goulet et al., 2004;

Moon et al., 2001). Cells stably transfected with

p110 CUX1 showed increased proliferation due to a

shortened G1-phase whereas embryonic fibroblasts

obtained from CUX1 knockout mice showed

elongated G1-phase and less proliferation compared

to cells isolated from wild-type mice (Sansregret et

al., 2006).

A genome-wide location array for p110 CUX1

binding sites in transformed and non-transformed

cell lines identified numerous CUX1 target genes

that are related to proliferation and cell cycle

progression (Harada et al., 2008). Most of these

genes are activated by p110 CUX1 including DNA

polymerase-alpha, cyclin A2 and cyclin E2. In

Page 15: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

CUX1 (cut-like homeobox 1) Kühnemuth B, Michl P

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 191

contrast, other genes are repressed such as the

CDK-inhibitor p21 (Truscott et al., 2003; Nishio

and Walsh, 2004; Harada et al., 2008).

Cell motility First evidence that CUX1 plays a role in cell

motility originates from knockdown studies in

fibroblasts and a panel of human cancer cell lines

that revealed that depletion of CUX1 leads to

decreased cell migration and invasion (Michl et al.,

2005). In agreement with this, cells stably

expressing p110 and p75 CUX1 show increased

cell migration and invasion (Kedinger et al., 2009;

Cadieux et al., 2009). Additionally, tail vein

injection of cells stably expressing shRNA against

CUX1 resulted in reduced formation of lung

metastases, whereas injection of cells stably

overexpressing CUX1 led to increased lung

metastases (Michl et al., 2005; Cadieux et al.,

2009).

The molecular basis for these effects on cell

motility was in part elucidated in a genome-wide

location analysis in several cell lines (Kedinger et

al., 2009). In this study, CUX1 was found to inhibit

the expression of genes that repress cell migration

(e.g. E-cadherin, occludin) and to turn on the

expression of genes that promote cell migration

(e.g. FAK, N-cadherin, vimentin) (Kedinger et al.,

2009). The regulation of these genes seems to be

mediated both directly by binding of CUX1 to the

gene promoters but also indirectly by modulation of

transcription factors and signaling proteins involved

in EMT (e.g. SNAI1, SNAI2, Src, Wnt5a)

(Kedinger et al., 2009; Aleksic et al., 2007; Ripka

et al., 2007). Additionally, several of the CUX1

target genes are known GTPases important for

actin-cytoskeleton polymerization (Kedinger et al.,

2009).

Apoptosis Studies in pancreatic cancer cell lines showed that

depletion of CUX1 by siRNA increases TNFalpha-

and TRAIL-induced apoptosis whereas

overexpression of CUX1 rescues from apoptosis.

Additionally, treatment of xenograft tumours with

siRNA for CUX1 lead to retarded tumour growth

and increased apoptosis. These effects are at least in

part explained by a positive regulation of the

antiapoptotic protein BCL2 by CUX1 (Ripka et al.,

2010a). Subsequently, the glutamate receptor

GRIA3 was identified as another downstream target

of CUX1 able to mediate its antiapoptotic effects

(Ripka et al., 2010b).

Homology

Cut homeodomain proteins are highly conserved in

evolution of metazoans. Homologues of the

Drosophila melanogaster Cut protein have been

described at least in human, dog and mouse

(Neufeld et al., 1992; Andres et al., 1992; Valarché

et al., 1993). In humans, a homologue gene, called

CUX2, was described (Jacobsen et al., 2001).

Mutations

Note

A missense mutation affecting the homeodomain

has been described in one patient suffering from

acute myeloid leukaemia, the significance of which

remains to be elucidated (Thoennissen et al., 2011).

Implicated in

Pancreatic cancer

Note

In pancreatic cancer CUX1 expression is elevated

compared to normal pancreas tissue (Ripka et al.,

2010a). Furthermore, an increased expression in

high-grade tumours compared to low grade tumours

was described (Michl et al., 2005).

The expression of CUX1 is accompanied by the

overexpression of its downstream targets WNT5a

and GRIA3 that, at least in part, mediate the

proinvasive and proproliferative effects of CUX1

(Ripka et al., 2006; Ripka et al., 2010b).

Antiapoptotic effects of CUX1 in pancreatic cancer,

that have been shown in in vitro studies and in

xenograft models, are associated with a positive

regulation of BCL2 and downregulation of tumour

necrosis factor alpha and are, at least in part,

mediated by the glutamate receptor GRIA3 (Ripka

et al., 2010a; Ripka et al., 2010b).

Breast cancer

Note

In mammary carcinoma the CUX1 expression is

increased in high-grade tumours compared to low

grade tumours and a reverse correlation between

CUX1 mRNA levels and the relapse free- and

overall-survival was shown (Michl et al., 2005).

Furthermore, is has been shown that the expression

levels of the intron 20-initiated mRNA, that leads to

the synthesis of the p75 CUX1 isoform, is

specifically expressed in breast cancer and

positively correlated with a diffuse infiltrative

growth pattern (Goulet et al., 2002). Transgenic

mice expressing p75 and p110 CUX1 under the

control of the mouse mammary tumour virus-long

terminal repeat developed breast cancer after a long

latency period. This tumour development was

accompanied by an increased activity of WNT-β-

catenin signalling (Cadieux et al., 2009).

References Neufeld EJ, Skalnik DG, Lievens PM, Orkin SH. Human CCAAT displacement protein is homologous to the Drosophila homeoprotein, cut. Nat Genet. 1992 Apr;1(1):50-5

Andres V, Nadal-Ginard B, Mahdavi V. Clox, a mammalian homeobox gene related to Drosophila cut, encodes DNA-binding regulatory proteins differentially expressed during development. Development. 1992 Oct;116(2):321-34

Page 16: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

CUX1 (cut-like homeobox 1) Kühnemuth B, Michl P

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 192

Scherer SW, Neufeld EJ, Lievens PM, Orkin SH, Kim J, Tsui LC. Regional localization of the CCAAT displacement protein gene (CUTL1) to 7q22 by analysis of somatic cell hybrids. Genomics. 1993 Mar;15(3):695-6

Valarché I, Tissier-Seta JP, Hirsch MR, Martinez S, Goridis C, Brunet JF. The mouse homeodomain protein Phox2 regulates Ncam promoter activity in concert with Cux/CDP and is a putative determinant of neurotransmitter phenotype. Development. 1993 Nov;119(3):881-96

Harada R, Dufort D, Denis-Larose C, Nepveu A. Conserved cut repeats in the human cut homeodomain protein function as DNA binding domains. J Biol Chem. 1994 Jan 21;269(3):2062-7

Lievens PM, Donady JJ, Tufarelli C, Neufeld EJ. Repressor activity of CCAAT displacement protein in HL-60 myeloid leukemia cells. J Biol Chem. 1995 May 26;270(21):12745-50

Vanden Heuvel GB, Bodmer R, McConnell KR, Nagami GT, Igarashi P. Expression of a cut-related homeobox gene in developing and polycystic mouse kidney. Kidney Int. 1996 Aug;50(2):453-61

Banan M, Rojas IC, Lee WH, King HL, Harriss JV, Kobayashi R, Webb CF, Gottlieb PD. Interaction of the nuclear matrix-associated region (MAR)-binding proteins, SATB1 and CDP/Cux, with a MAR element (L2a) in an upstream regulatory region of the mouse CD8a gene. J Biol Chem. 1997 Jul 18;272(29):18440-52

Kim EC, Lau JS, Rawlings S, Lee AS. Positive and negative regulation of the human thymidine kinase promoter mediated by CCAAT binding transcription factors NF-Y/CBF, dbpA, and CDP/cut. Cell Growth Differ. 1997 Dec;8(12):1329-38

Lievens PM, Tufarelli C, Donady JJ, Stagg A, Neufeld EJ. CASP, a novel, highly conserved alternative-splicing product of the CDP/cut/cux gene, lacks cut-repeat and homeo DNA-binding domains, and interacts with full-length CDP in vitro. Gene. 1997 Sep 15;197(1-2):73-81

Pattison S, Skalnik DG, Roman A. CCAAT displacement protein, a regulator of differentiation-specific gene expression, binds a negative regulatory element within the 5' end of the human papillomavirus type 6 long control region. J Virol. 1997 Mar;71(3):2013-22

Coqueret O, Bérubé G, Nepveu A. The mammalian Cut homeodomain protein functions as a cell-cycle-dependent transcriptional repressor which downmodulates p21WAF1/CIP1/SDI1 in S phase. EMBO J. 1998a Aug 17;17(16):4680-94

Coqueret O, Martin N, Bérubé G, Rabbat M, Litchfield DW, Nepveu A. DNA binding by cut homeodomain proteins is down-modulated by casein kinase II. J Biol Chem. 1998b Jan 30;273(5):2561-6

Ai W, Toussaint E, Roman A. CCAAT displacement protein binds to and negatively regulates human papillomavirus type 6 E6, E7, and E1 promoters. J Virol. 1999 May;73(5):4220-9

Catt D, Hawkins S, Roman A, Luo W, Skalnik DG. Overexpression of CCAAT displacement protein represses the promiscuously active proximal gp91(phox) promoter. Blood. 1999a Nov 1;94(9):3151-60

Catt D, Luo W, Skalnik DG. DNA-binding properties of CCAAT displacement protein cut repeats. Cell Mol Biol (Noisy-le-grand). 1999b Dec;45(8):1149-60

Li S, Moy L, Pittman N, Shue G, Aufiero B, Neufeld EJ, LeLeiko NS, Walsh MJ. Transcriptional repression of the cystic fibrosis transmembrane conductance regulator gene, mediated by CCAAT displacement protein/cut

homolog, is associated with histone deacetylation. J Biol Chem. 1999 Mar 19;274(12):7803-15

van Gurp MF, Pratap J, Luong M, Javed A, Hoffmann H, Giordano A, Stein JL, Neufeld EJ, Lian JB, Stein GS, van Wijnen AJ. The CCAAT displacement protein/cut homeodomain protein represses osteocalcin gene transcription and forms complexes with the retinoblastoma protein-related protein p107 and cyclin A. Cancer Res. 1999 Dec 1;59(23):5980-8

Li S, Aufiero B, Schiltz RL, Walsh MJ. Regulation of the homeodomain CCAAT displacement/cut protein function by histone acetyltransferases p300/CREB-binding protein (CBP)-associated factor and CBP. Proc Natl Acad Sci U S A. 2000 Jun 20;97(13):7166-71

O'Connor MJ, Stünkel W, Koh CH, Zimmermann H, Bernard HU. The differentiation-specific factor CDP/Cut represses transcription and replication of human papillomaviruses through a conserved silencing element. J Virol. 2000 Jan;74(1):401-10

Rong Zeng W, Soucie E, Sung Moon N, Martin-Soudant N, Bérubé G, Leduy L, Nepveu A. Exon/intron structure and alternative transcripts of the CUTL1 gene. Gene. 2000 Jan 4;241(1):75-85

Stünkel W, Huang Z, Tan SH, O'Connor MJ, Bernard HU. Nuclear matrix attachment regions of human papillomavirus type 16 repress or activate the E6 promoter, depending on the physical state of the viral DNA. J Virol. 2000 Mar;74(6):2489-501

Ellis T, Gambardella L, Horcher M, Tschanz S, Capol J, Bertram P, Jochum W, Barrandon Y, Busslinger M. The transcriptional repressor CDP (Cutl1) is essential for epithelial cell differentiation of the lung and the hair follicle. Genes Dev. 2001 Sep 1;15(17):2307-19

Jacobsen NJ, Elvidge G, Franks EK, O'Donovan MC, Craddock N, Owen MJ. CUX2, a potential regulator of NCAM expression: genomic characterization and analysis as a positional candidate susceptibility gene for bipolar disorder. Am J Med Genet. 2001 Apr 8;105(3):295-300

Khanna-Gupta A, Zibello T, Sun H, Lekstrom-Himes J, Berliner N. C/EBP epsilon mediates myeloid differentiation and is regulated by the CCAAT displacement protein (CDP/cut). Proc Natl Acad Sci U S A. 2001 Jul 3;98(14):8000-5

Moon NS, Premdas P, Truscott M, Leduy L, Bérubé G, Nepveu A. S phase-specific proteolytic cleavage is required to activate stable DNA binding by the CDP/Cut homeodomain protein. Mol Cell Biol. 2001 Sep;21(18):6332-45

Santaguida M, Ding Q, Bérubé G, Truscott M, Whyte P, Nepveu A. Phosphorylation of the CCAAT displacement protein (CDP)/Cux transcription factor by cyclin A-Cdk1 modulates its DNA binding activity in G(2). J Biol Chem. 2001 Dec 7;276(49):45780-90

De Vos J, Thykjaer T, Tarte K, Ensslen M, Raynaud P, Requirand G, Pellet F, Pantesco V, Rème T, Jourdan M, Rossi JF, Ørntoft T, Klein B. Comparison of gene expression profiling between malignant and normal plasma cells with oligonucleotide arrays. Oncogene. 2002 Oct 3;21(44):6848-57

Goebel P, Montalbano A, Ayers N, Kompfner E, Dickinson L, Webb CF, Feeney AJ. High frequency of matrix attachment regions and cut-like protein x/CCAAT-displacement protein and B cell regulator of IgH transcription binding sites flanking Ig V region genes. J Immunol. 2002 Sep 1;169(5):2477-87

Goulet B, Watson P, Poirier M, Leduy L, Bérubé G, Meterissian S, Jolicoeur P, Nepveu A. Characterization of

Page 17: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

CUX1 (cut-like homeobox 1) Kühnemuth B, Michl P

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 193

a tissue-specific CDP/Cux isoform, p75, activated in breast tumor cells. Cancer Res. 2002 Nov 15;62(22):6625-33

Truscott M, Raynal L, Premdas P, Goulet B, Leduy L, Bérubé G, Nepveu A. CDP/Cux stimulates transcription from the DNA polymerase alpha gene promoter. Mol Cell Biol. 2003 Apr;23(8):3013-28

Tsutsumi S, Taketani T, Nishimura K, Ge X, Taki T, Sugita K, Ishii E, Hanada R, Ohki M, Aburatani H, Hayashi Y. Two distinct gene expression signatures in pediatric acute lymphoblastic leukemia with MLL rearrangements. Cancer Res. 2003 Aug 15;63(16):4882-7

Goulet B, Baruch A, Moon NS, Poirier M, Sansregret LL, Erickson A, Bogyo M, Nepveu A. A cathepsin L isoform that is devoid of a signal peptide localizes to the nucleus in S phase and processes the CDP/Cux transcription factor. Mol Cell. 2004 Apr 23;14(2):207-19

Kaul-Ghanekar R, Jalota A, Pavithra L, Tucker P, Chattopadhyay S. SMAR1 and Cux/CDP modulate chromatin and act as negative regulators of the TCRbeta enhancer (Ebeta). Nucleic Acids Res. 2004;32(16):4862-75

Nishio H, Walsh MJ. CCAAT displacement protein/cut homolog recruits G9a histone lysine methyltransferase to repress transcription. Proc Natl Acad Sci U S A. 2004 Aug 3;101(31):11257-62

Truscott M, Raynal L, Wang Y, Bérubé G, Leduy L, Nepveu A. The N-terminal region of the CCAAT displacement protein (CDP)/Cux transcription factor functions as an autoinhibitory domain that modulates DNA binding. J Biol Chem. 2004 Nov 26;279(48):49787-94

Michl P, Ramjaun AR, Pardo OE, Warne PH, Wagner M, Poulsom R, D'Arrigo C, Ryder K, Menke A, Gress T, Downward J. CUTL1 is a target of TGF(beta) signaling that enhances cancer cell motility and invasiveness. Cancer Cell. 2005 Jun;7(6):521-32

Goulet B, Truscott M, Nepveu A. A novel proteolytically processed CDP/Cux isoform of 90 kDa is generated by cathepsin L. Biol Chem. 2006 Sep;387(9):1285-93

Maitra U, Seo J, Lozano MM, Dudley JP. Differentiation-induced cleavage of Cutl1/CDP generates a novel dominant-negative isoform that regulates mammary gene expression. Mol Cell Biol. 2006 Oct;26(20):7466-78

Michl P, Downward J. CUTL1: a key mediator of TGFbeta-induced tumor invasion. Cell Cycle. 2006 Jan;5(2):132-4

Sansregret L, Goulet B, Harada R, Wilson B, Leduy L, Bertoglio J, Nepveu A. The p110 isoform of the CDP/Cux transcription factor accelerates entry into S phase. Mol Cell Biol. 2006 Mar;26(6):2441-55

Aleksic T, Bechtel M, Krndija D, von Wichert G, Knobel B, Giehl K, Gress TM, Michl P. CUTL1 promotes tumor cell migration by decreasing proteasome-mediated Src degradation. Oncogene. 2007 Aug 30;26(40):5939-49

Li J, Wang E, Dutta S, Lau JS, Jiang SW, Datta K, Mukhopadhyay D. Protein kinase C-mediated modulation of FIH-1 expression by the homeodomain protein CDP/Cut/Cux. Mol Cell Biol. 2007 Oct;27(20):7345-53

Ripka S, König A, Buchholz M, Wagner M, Sipos B, Klöppel G, Downward J, Gress T, Michl P. WNT5A--target of CUTL1 and potent modulator of tumor cell migration and invasion in pancreatic cancer. Carcinogenesis. 2007 Jun;28(6):1178-87

Truscott M, Denault JB, Goulet B, Leduy L, Salvesen GS, Nepveu A. Carboxyl-terminal proteolytic processing of CUX1 by a caspase enables transcriptional activation in proliferating cells. J Biol Chem. 2007 Oct 12;282(41):30216-26

Ueda Y, Su Y, Richmond A. CCAAT displacement protein regulates nuclear factor-kappa beta-mediated chemokine transcription in melanoma cells. Melanoma Res. 2007 Apr;17(2):91-103

Harada R, Vadnais C, Sansregret L, Leduy L, Bérubé G, Robert F, Nepveu A. Genome-wide location analysis and expression studies reveal a role for p110 CUX1 in the activation of DNA replication genes. Nucleic Acids Res. 2008 Jan;36(1):189-202

Cadieux C, Kedinger V, Yao L, Vadnais C, Drossos M, Paquet M, Nepveu A. Mouse mammary tumor virus p75 and p110 CUX1 transgenic mice develop mammary tumors of various histologic types. Cancer Res. 2009 Sep 15;69(18):7188-97

Kedinger V, Sansregret L, Harada R, Vadnais C, Cadieux C, Fathers K, Park M, Nepveu A. p110 CUX1 homeodomain protein stimulates cell migration and invasion in part through a regulatory cascade culminating in the repression of E-cadherin and occludin. J Biol Chem. 2009 Oct 2;284(40):27701-11

Ripka S, Neesse A, Riedel J, Bug E, Aigner A, Poulsom R, Fulda S, Neoptolemos J, Greenhalf W, Barth P, Gress TM, Michl P. CUX1: target of Akt signalling and mediator of resistance to apoptosis in pancreatic cancer. Gut. 2010a Aug;59(8):1101-10

Ripka S, Riedel J, Neesse A, Griesmann H, Buchholz M, Ellenrieder V, Moeller F, Barth P, Gress TM, Michl P. Glutamate receptor GRIA3--target of CUX1 and mediator of tumor progression in pancreatic cancer. Neoplasia. 2010b Aug;12(8):659-67

Sansregret L, Gallo D, Santaguida M, Leduy L, Harada R, Nepveu A. Hyperphosphorylation by cyclin B/CDK1 in mitosis resets CUX1 DNA binding clock at each cell cycle. J Biol Chem. 2010 Oct 22;285(43):32834-43

Fragiadaki M, Ikeda T, Witherden A, Mason RM, Abraham D, Bou-Gharios G. High doses of TGF-β potently suppress type I collagen via the transcription factor CUX1. Mol Biol Cell. 2011 Jun 1;22(11):1836-44

Thoennissen NH, Lasho T, Thoennissen GB, Ogawa S, Tefferi A, Koeffler HP. Novel CUX1 missense mutation in association with 7q- at leukemic transformation of MPN. Am J Hematol. 2011 Aug;86(8):703-5

This article should be referenced as such:

Kühnemuth B, Michl P. CUX1 (cut-like homeobox 1). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):189-193.

Page 18: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 194

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) June L Traicoff, Stephen M Hewitt, Joon-Yong Chung

Center for Peer Review and Science Management, SRA International, Inc Maryland, USA (JLT),

Applied Molecular Pathology Laboratory & Tissue Array Research Program, Laboratory of

Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health,

Bethesda, MD, USA (SMH), Applied Molecular Pathology Laboratory, Laboratory of Pathology,

Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD,

USA (JYC)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/DNAJA3ID40342ch16p13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI DNAJA3ID40342ch16p13.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: FLJ45758, TID1, hTid-1

HGNC (Hugo): DNAJA3

Location: 16p13.3

Local order: According to NCBI Map Viewer,

genes flanking DNAJA3 are COR07-PAM16,

NMRAL1, and HMOX2.

Note

DNAJA3 was first identified by its ability to form

complexes with the human papillomavirus E7

oncoprotein (Schilling et al., 1998) in a yeast-two

hybrid screen. Sequence analysis revealed that

DNAJA3 was the human homolog of the

Drosophila tumor suppressor protein Tid56.

Furthermore, DNAJA3 contained a J-domain which

is characteristic of the family of DnaJ proteins

which interact with and stimulate the ATPase

activity of heat shock cognate 70 (hsc70) family

members (Schilling et al., 1998).

DNA/RNA

Note

DNAJA3 belongs to the evolutionarily conserved

DNAJ/HSP40 family of proteins. There are 41

known DnaJ/Hsp40 proteins in the human genome

(Qiu et al., 2006).

According to NCBI Gene, the DNAJA3 gene is

conserved in human chimpanzee, cow, mouse, rat,

chicken, zebrafish, fruit fly, mosquito, C. elegans,

S. pombe, S. cerevisiae, K. lactis, E. gossypii, M.

grisea, N. crassa, and rice.

Description

The DNAJA3 gene is located on chromosome

16p13.3 between markers D16S521 and D16S418.

This chromosomal region carries several loci

implicated in human proliferation disorders,

including the tuberous sclerosis 2 gene (TSC2),

polycystic kidney disease 1 gene (PKD1), and the

CREB binding protein (CBP) locus (Yin and

Rozakis-Adcock, 2001).

Chromosome 16 - NC_000016.9. Modified from NCBI Map Viewer.

Page 19: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 195

DNAJA3 is approximately 34 kb and is composed

of 12 exons separated by 11 introns. Exon sizes

vary from 64 to 232 nucleotides, with the exception

of exon 12 corresponding to the 3' untranslated

region of DNAJA3, which extends over 1.1 kb.

Intron sizes vary from 618 to 8291 nucleotides (Yin

and Rozakis-Adcock, 2001).

Sequence encoding the DNAJ domain is present in

exons 2, 3 and 4, sequence encoding the Cys-rich

domain is found in exons 5 an 6, and the COOH-

terminal region is found in exons 7 through 11 (Yin

and Rozakis-Adcock, 2001).

Transcription

Promoter elements. DNAJA3 contains a putative

transcriptional start site 21 nucleotides upstream of

the initiating methionine. The presumptive

promoter is characterized by the lack of TATA and

CAAT motifs, and a high G+C content. The 5'

flanking region contains several consensus binding

sites for transcription factors that regulate gene

expression during tissue and organ development,

such as myeloid zinc finger (MZF1), Ikaros 2 and

homeodomain proteins, as well as factors

implicated in cell growth and survival responses,

including AP-1, PEA3, E2F and NF-kB.

Splice variants. Alternative splicing of a single

heteronuclear RNA (hnRNA) species generates the

three DNAJA3 isoforms. The long form DNAJA3L

(hTID1L) fully incorporates all exons. The

intermediate form DNAJA3I (hTID1I) is generated

by splicing of exon 10 to exon 12. This results in

the loss of the 34 C-terminal-most amino acids as

well as the stop codon; these are replaced with six

amino acids KRSTGN from exon 12. The short

form DNAJA3S (hTID1S) results from an in-frame

deletion of 50 amino acids that correspond

precisely to exon 5 (Yin and Rozakis-Adcock,

2001).

RNA expression. DNAJA3 mRNA was detected in

50 different human fetal and adult tissues. However

the relative abundance correlated with metabolic

activity of the tissues, with the highest levels

observed in liver and skeletal muscle (Kurzik-

Dumke and Czaja, 2007).

Human tissues and cell lines showed differential

expression of the three DNAJA3 splice variant

mRNAs. Fetal brain tissue predominantly expressed

DNAJA3I, while breast tissues and T-cells

predominantly expressed DNAJA3L. Cell lines

derived from prostate epithelia, skin and lung

fibroblasts, normal astrocytes, and an osteosarcoma

predominantly expressed DNAJA3I with low levels

of DNAJA3L also present. DNAJA3S transcript

was undetectable in all samples (Yin and Rozakis-

Adcock, 2001).

DNAJA3 transcripts showed differential expression

during development. Expression of DNAJA3

transcripts in mouse neonatal cardiomyocytes

increased as development of the heart proceeded

and reached a maximal level at 4 weeks of age,

when cardiac myocytes have matured (Hayashi et

al., 2006). DNAJA3 expression also increased in

pathological cardiac hypertrophic states (Hayashi et

al., 2006).

Pseudogene

Paralogs. According to GeneCards, DNAJC16 is a

paralog for DNAJA3. DNAJC16 is located on

chromosome 1p36.1.

Protein

Note

The DNAJA3 gene encodes three cytosolic (Tid50,

Tid48, Tid46) proteins and three mitochondrial

(Tid43, Tid40, Tid38) proteins. Proteins encoded

by the longer splice variant DNAJA3L have often

been designated in the literature as Tid1L. Proteins

encoded by the shorter splice variant DNAJA3S

have often been named Tid1S. In this review,

Tid1L will be designated DNAJA3L, and Tid1S

will be designated DNAJA3S. Specific isoforms

will be designated by size, e.g., Tid 50 will be

designated as DNAJA3 (50 kD).

Description

DNAJA3 protein is present in two isoforms,

corresponding to splice variants encoding them.

The longer DNAJA3L isoform is a 480 amino acid

protein with a predicted size of 52 kD. The shorter

DNAJA3S isoform is a 453 amino acid protein with

a predicted size of 49 kD (Lu et al., 2006; UniProt).

Expression

DNAJA3 protein has been detected in human

breast, colon, ovarian, lung, and head and neck

squamous cell carcinoma (HNSCC) tissues

(Traicoff et al., 2007; Kurzik-Dumke et al., 2008;

Chen et al., 2009).

Localisation

DNAJA3 localizes to human mitochondrial

nucleoids, which are large protein complexes bound

to mitochondrial DNA. Unlike other DnaJs,

DNAJA3L and DNAJA3S form heterocomplexes;

both unassembled and complexed DNAJA3 are

observed in human cells. DNAJA3L showed a

longer residency time in the cytosol prior to

mitochondrial import as compared with DNAJA3S;

DNAJA3L was also significantly more stable in the

cytosol than DNAJA3S, which is rapidly degraded

(Lu et al., 2006).

Function

I. Binding partners Human DNAJA3 protein has been shown to

interact with diverse partners, including viral

proteins, heat shock proteins, and key regulators of

cell signaling and growth.

Viral proteins Hepatitis B virus core protein: DNAJA3

associated with the hepatitis B virus core protein,

Page 20: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 196

specifically with the carboxyl-terminal region

(amino acids 94-185). The N-terminal end of

DNAJA3 (amino acids 1-447) was required for this

interaction. Furthermore, the DNAJA3S precursor

co-sedimented with viral capsid-like particles

composed of the full-length core protein (Sohn et

al., 2006). Interaction between DNAJA3 and the

HBV core protein was confirmed in co-

immunoprecipitation experiments using transfected

hepatoma cells (Sohn et al., 2006).

Epstein-Barr virus-encoded BARF1 protein: DNAJA3 (amino acids 149-320) associated with

the Epstein-Barr virus-encoded BARF1 protein

(amino acids 21-221). Interaction between

DNAJA3 and BARF1 was confirmed in co-

immunoprecipitation experiments using transfected

HeLa cells (Wang et al., 2006).

Herpes simplex virus type 1 UL9 protein: DNAJA3 associated with the herpes simplex virus

type 1 (HSV-1) UL9 protein. UL9 protein is an

origin-binding protein. Interaction between

DNAJA3 and UL9 was confirmed by in vitro co-

immunoprecipitation (Eom and Lehman, 2002).

Human T cell leukemia virus type 1 (HTLV-1)

Tax protein: DNAJA3 associated with HTLV-1

Tax. The interaction occurred through a central

cysteine-rich zinc finger-like region of DNAJA3

(amino acids 236 to 300). Interaction between

DNAJA3 and Tax was confirmed by co-

immunoprecipitation experiments using transfected

human embryonic kidney cells (HEK) (Cheng et

al., 2001). Furthermore, the DNAJA3 and Tax

interaction occurred through a complex comprised

of DNAJA3, Tax, and heat shock protein 70

(Hsp70), in which the cysteine-rich region of

DNAJA3 interacted with Tax, while the J domain

of DNAJA3 interacted with Hsp70 (Cheng et al.,

2001).

Human papilloma virus-16 (HPV-16) E7

oncoprotein: DNAJA3 was initially characterized

through its interaction with the HPV-16 E7

oncoprotein. DNAJA3 amino acids 1 to 235 and

297 to 342 independently interacted with HPV-16

E7. Interaction between DNAJA3 and HPV-16 E7

was confirmed by in vitro binding assays and co-

immunoprecipitation experiments using transfected

human osteosarcoma (U2OS) cells (Schilling et al.,

1998).

Heat shock proteins Hsp70 and Hsc70: endogenous DNAJA3

(specifically the cytosolic form)

immunoprecipitated with the heat shock proteins

Hsp70 and Hsc70 in normal colon epithelium and

colon cancer cell lines (Kurzik-Dumke and Czaja,

2007). Endogenous DNAJA3 also interacted with

Hsp70/Hsc70 in HEp2 cells, and this interaction

was reduced in cells treated with interferon-gamma

(Sarkar et al., 2001). The J domain of DNAJA3 was

shown to be required for interaction with Hsp70 in

HEK cells (Cheng et al., 2001).

Proteins encoded by the long and short splice forms

of DNAJA3, DNAJA3L and DNAJA3S,

respectively, showed differential interactions with

heat shock proteins. Unassembled DNAJA3L (the

long splice variant) was shown to interact with

Hsc70 specifically in the cytosol (Lu et al., 2006).

The unique carboxyl terminus of DNAJA3L was

required for this interaction (Lu et al., 2006). Both

DNAJA3S and DNAJA3L could interact with

Hsp70 (Kim et al., 2004). Endogenous DNAJA3L

and DNAJA3S coimmunoprecipitated with

mitochondrial Hsp70, but not Hsc70, in U2OS

osteosarcoma cells (Syken et al., 1999).

Tumor suppressor proteins Adenomatous polyposis coli (APC): endogenous

cytosolic DNAJA3 proteins interacted with APC in

normal colon epithelium and colorectal cancer cell

lines (HT-29, Caco-2, and HRT-18). The N-

terminal Armadillo domain of APC was sufficient

for binding to DNAJA3. The DNAJA3 and APC

interaction comprised part of a larger multi-

component complex that also contained Hsp70,

Hsc70, Actin, Dvl, and Axin. This complex

functions independently of the known roles of APC

in beta-catenin degradation and proliferation

mediated by Wg/Wnt signaling (Kurzik-Dumke and

Czaja, 2007).

Endogenous DNAJA3 proteins were shown to

interact with the caspase-cleaved N-terminus of

APC in HCT116 cells (Qian et al., 2010). The

caspase-cleaved APC protein has an important

physiological role in mediating apoptosis (Qian et

al., 2010).

Patched: endogenous human Patched interacted

with the cytosolic forms of the DNAJA3 proteins in

human colon epithelium and colon tumor cells

(Kurzik-Dumke and Czaja, 2007). The tumor-

associated polymorphism in Patched (Ptch FVB

allele) was associated with poorer binding to

DNAJA3 (Wakabayashi et al., 2007).

INT6: endogenous human DNAJA3 interacted with

INT6 (the p48 subunit of the eIF3 translation

initiation factor) in log phase, but not confluent,

Jurkat T-cells (Traicoff et al., 2007).

Von Hippel-Lindau protein (VHL): endogenous

pVHL co-immunoprecipitated with DNAJA3L

protein in HEK293 cells (Bae et al., 2005).

p53: DNAJA3 directly interacts with p53 through

the DNAJA3 DNAJ domain. Either the N- or C-

terminal domains of p53 was sufficient for the

interaction (Trinh et al., 2010).

Receptors

Interferon-gamma receptor (IFN-gammaR)

subunit IFN-gammaR2: DNAJA3 interacted with

IFN-gamma R2 in transfected COS cells.

Furthermore, DNAJA3 bound more efficiently to a

IFN-gammaR2 chimera with an active kinase

domain than to a similar construct with an inactive

kinase domain (Sarkar et al., 2001).

Page 21: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 197

ErbB-2 (HER2/neu): endogenous ErbB-2 and

DNAJA3 co-immunoprecipitated in SK-BR-3

breast cancer cells (Kim et al., 2004). The

cytoplasmic domains of ErbB-2 and DNAJA3 were

sufficient for this interaction (Kim et al., 2004).

ErbB-2 co-immunoprecipiated with DNAJA3 and

the carboxyl terminus of heat shock cognate 70

interacting protein (CHIP). This complex was

demonstrated in tissue extracts from breast tumor

specimens as well as in transfected cell lines (Jan et

al., 2011).

Trk receptor tyrosine kinases: the carboxyl-

terminal end of DNAJA3 (residues 224-429) bound

to Trk at its activation loop in a phosphotyrosine-

dependent manner (Liu et al., 2005).

Muscle-specific kinase (MuSK) component of

the agrin receptor: DNAJA3S, but not DNAJA3L,

associated with the cytoplasmic portion of MuSK in

mouse skeletal muscle cells (Linnoila et al., 2008).

Signaling proteins NF-kappaB: DNAJA3 strongly associated with the

cytoplasmic protein complex of NF-kappaB-

IkappaB through direct interaction with

IkappaBalpha/IkappaBbeta and the IKKalpha/beta

subunits of the IkappaB kinase complex. The

endogenous interaction was observed in Jurkat,

SAOS-2, and HEK293 cells (Cheng et al., 2005).

JAK/STAT: Jak2 interacted with DNAJA3S as

well as DNAJA3L as shown by

immunoprecipitation from transfected COS-1 cells

expressing these proteins. Endogenous DNAJA3

and Jak2 were shown to interact in HEp2 cells

(Sarkar et al., 2001).

The carboxyl terminus of endogenous DNAJA3L,

but not DNAJA3S, co-immunoprecipitated with

STAT1 and with STAT3 in U2OS cells (Lu et al.,

2006). DNAJA3L remained associated with

activated phosphorylated STAT1 upon treatment

with interferon-gamma (Lu et al., 2006).

The DNAJ domain of DNAJA3 interacted with the

transactivation domain of Stat5b in hematopoietic

cell lines (Dhennin-Duthille et al., 2011).

p120 GTPase-activating protein (GAP): both the

cytoplasmic precursor and mitochondrial mature

forms of murine DNAJA3 associated with GAP in

vivo in rodent cells. GAP selectively bound to the

unphosphorylated form of murine DNAJA3

(Trentin et al., 2001).

DNA replication proteins

DNA polymerase gamma (Polga) alpha subunit: endogenous DNAJA3 interacted with the alpha

subunit of Polga in HEK293 cells. Polga is the only

mitochondrial DNA polymerase responsible for all

mitochondrial DNA synthetic reactions (Hayashi et

al., 2006).

II. Signaling pathways and cellular effects DNAJA3 modulates diverse signaling pathways

and cellular effects that are vital for cell growth and

differentiation.

Neural pathways Neuromuscular synaptogenesis: DNAJA3 is an

essential component of the agrin signaling pathway

that is crucial for synaptic development.

Motoneuron-derived agrin clusters nicotinic

acetylcholine receptors (AChRs) in mammalian

cells. DNAJA3 binds to the cytoplasmic domain of

muscle-specific kinase (MuSK), a component of the

agrin receptor and colocalizes with AchRs at

developing, adult, and denvervated motor

endplates. DNAJA3 transduces signals from MuSK

activation to AchR clustering, culmintating in

cross-linking to the subsynaptic cytoskeleton, as

demonstrated by knockdown and overexpression

experiments.

Knockdown of DNAJA3 in skeletal muscle fibers

resulted in dispersed synaptic AchR clusters and

impaired neuromuscular transmission. Knockdown

of DNAJA3 in myotubes resulted in inhibition of

AchR clustering, inhibition of agrin-induced

activation of the Rac and Rho small GTPases and

tyrosine phosphorylation of AchR, and decreased

Dok-7-induced clustering of AChRs. In contrast,

overexpression of the N-terminal half of DNAJA3

induced agrin-and MuSK-independent

phosphorylation and clustering of AChRs (Linnoila

et al., 2008; Song and Balice-Gordon, 2008).

Neurite outgrowth: DNAJA3 regulated nerve

growth factor (NGF)-induced neurite outgrowth in

PC12-derived nnr5 cells. DNAJA3 bound to Trk at

the activation loop and DNAJA3 was tyrosine

phosphorylated by Trk in yeast cells, transfected

cells, and in neurotophin-stimulated primary rat

hippocampal neurons. Overexpression of DNAJA3

led to NGF-induced neurite outgrowth in TrkA-

expressing nnr5 cells. In contrast, knockdown of

DNAJA3 resulted in reduced NGF-induced neurite

growth in nnr5-TrkA cells (Liu et al., 2005).

Viral pathways Hepatitis B virus replication: expression of

DNAJA3 suppressed replication of HBV in human

hepatoma cells, while knockdown of DNAJA3 led

to increased HBV replication. The mechanism for

inhibited replication was through accelerated

degradation and destabilization of the viral core and

HBx proteins (Sohn et al., 2006).

Herpes simplex virus type 1 replication: the

HSV-1 UL9 protein is an origin-binding protein

that is essential for viral DNA replication. DNAJA3

modulates DNA replication by enhancing the

binding of UL9 protein to an HSV-1 origin and

facilitating formation of the multimer from the

dimeric UL9 protein, perhaps through a chaperone

function. However, DNAJA3 had no effect on the

DNA-dependent ATPase or helicase activities

associated with the UL9 protein (Eom and Lehman,

2002).

Epstein-Barr virus secretion: the EBV BARF1

gene encodes a secretory protein with transforming

and mitogenic activities. Coexpression experiments

Page 22: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 198

with BARF1 and DNAJA3 showed that DNAJA3

could promote secretion of BARF1, perhaps

through chaperone functions (Wang et al., 2006).

Motility and metastasis DNAJA3 was shown to negatively regulate the

motility and metastasis of breast cancer cells

through attenuation of nuclear factor kappaB

activity on the promoter of the IL8 gene (Kim et al.,

2005). Reductions of DNAJA3 levels in MDA-

MB231 breast cancer cells increased their migration

as a result of increased interleukin-8 (IL-8)

secretion without affecting survival or growth rate.

Furthermore, DNAJA3 was shown to negatively

modulate de novo synthesis of IL-8 through

regulating NFkappaB activity. Additionally,

DNAJA3 knockdown enhanced the metastasis of

breast cancer cells in animals (Kim et al., 2005).

Other studies also indicate a potential role for

DNAJA3 in inhibiting transformation and

metastasis. Stable DNAJA3 knockdown cells

exhibited an enhanced ability for anchorage-

independent growth, as measured by an increase in

soft-agar colony formation (Edwards and Münger,

2004). In contrast, ectopic expression of DNAJA3

in HNSCC cells was shown to significantly inhibit

cell proliferation, migration, invasion, anchorage-

independent growth, and xenotransplantation

tumorigenicity (Chen et al., 2009). Expression of

DNAJA3 inhibited the transformation phenotype of

two human lung adenocarcinoma cell lines (Cheng

et al., 2001).

Apoptosis DNAJA3 encodes two mitochondrial matrix

localized splice variants: DNAJA3 (43 kD) and

DNAJA3 (40 kD). DNAJA3 (43 kD) and DNAJA3

(40 kD) do not themselves induce apoptosis;

instead they have opposing effects on apoptosis

induced by exogenous stimuli.

Expression of DNAJA3 (43 kD) increases apoptosis

induced by both the DNA-damaging agent

mitomycin c and tumor necrosis factor-alpha. This

activity is J domain-dependent, since a J domain

mutant of DNAJA3 (43 kD) suppressed apoptosis.

Conversely, expression of DNAJA3 (40 kD)

suppressed apoptosis, while expression of a J

domain mutant of DNAJA3 (40 kD) increased

apoptosis (Syken et al., 1999).

Cells lacking expression of DNAJA3 proteins were

protected from cell death in response to multiple

stimuli, including cisplatin, tumor necrosis factor

alpha/cycloheximide and mitomycin C (Edwards

and Münger, 2004).

DNAJA3 regulates activation-induced cell death

(AICD) in the Th2 subset of helper T cells. AICD is

an apoptotic process induced by stimulation

through the T-cell receptor and Th2 cells are

significantly less prone to AICD than Th1 cells are.

The antiapoptotic variant, Tid-1S was shown to be

selectively induced in murine Th2 cells following

activation. Expression of a dominant-negative

mutant of hTid-1S in a Th2 cell line strikingly

enhanced activation of caspase 3 in response to

CD3 stimulation, and caused the cells to become

sensitive to AICD. Therefore, the accumulation of

Tid-1S in Th2 cells following activation may

contribute to the induction of apoptosis resistance

during the activation of Th2 cells (Syken et al.,

2003).

DNAJA3 mediates apoptosis through the

nuclear factor kappaB (NF-kappaB) pathway. DNAJA3 repressed the activity of NF-kappaB

through physical and functional interactions with

the IKK complex and IkappaB. Overexpression of

DNAJA3 led to inhibition of cell proliferation and

induction of apoptosis of human osteosarcoma cells

and human melanoma cells regardless of the p53

expression status. In contrast, cells transduced with

a DNAJA3 mutant that has an N-terminal J domain

deletion and that lost suppressive activity on IKK,

continued to proliferate (Cheng et al., 2005).

DNAJA3 mediates apoptosis through the Bcl-2

pathway. DNAJA3 induced apoptosis in SF767

glioma cells that contained a tumor-associated

mutation at the DNAJA3 locus. Apoptosis resulted

from caspase activation and cytochrome c release

from mitochondria. However, Bcl-XL protected

cells from hTid-1S-induced cell death and

cytochrome c release. However, hTid1S caused S

and G2/M arrest in cells with wild type Tid1.

Interestingly, hTid1L had no effect on growth of

glioma cells (Trentin et al., 2004).

Immature dnaja3(-/-) DN4 thymocytes exhibited

significantly reduced expression of the

antiapoptotic bcl-2 gene (Lo et al., 2005).

Expression of constitutively active AKT (pAKT)

counteracted and inhibited DNAJA3-induced

apoptosis in HNSCC cells (Chen et al., 2009).

DNAJA3 mediates apoptosis through APC. DNAJA3 (40 kD) isoform inhibited apoptosis

through antagonizing the apoptotic function of the

N-terminal region of the APC protein (Qian et al.,

2010).

DNAJA3 mediates apoptosis through p53. Overexpression of DNAJA3 enhanced p53-

dependent apoptosis, and restored pro-apoptotic

activity of mutant p53 in colon, breast, and glioma

cell lines (Ahn et al., 2010). The mechanism is

through direct interaction of the DNAJ domain of

DNAJA3 and p53 (Trinh et al., 2010). In contrast,

depletion of DNAJA3 resulted in the inhibition of

hypoxia or genotoxic stress-induced p53

mitochondrial localization and apoptosis (Trinh et

al., 2010).

Mitochondrial functions Although DNAJA3 has many cellular functions,

DNAJA3 often localizes to the mitochondria and

also has important functions in the mitochondria.

Epidermal growth factor (EGF) response: GAP

and DNAJA3 were shown to colocalize at

Page 23: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 199

perinuclear mitochondrial membranes in response

to EGF stimulation (Trentin et al., 2001).

p53 localization and apoptotic function: depletion of DNAJA3 prevented p53 accumulation

at the mitochondria and resulted in resistance to

apoptosis under hypoxic or genotoxic stresses

(Trinh et al., 2010). DNAJA3 formed a complex

with p53 under hypoxic conditions that directed

p53 translocation to the mitochondria and the

subsequent initiation of apoptosis (Ahn et al.,

2010). Loss of DNAJA3 expression abrogated p53

translocation to the mitochondria and inhibited

apoptosis (Ahn et al., 2010). Conversely,

overexpression of DNAJA3 promoted p53

mitochondrial localization and apoptosis (Ahn et

al., 2010).

Viral protein localization: in the absence of Tax,

expression of the DNAJA3/Hsp70 molecular

complex was targeted to perinuclear mitochondrial

clusters. In the presence of Tax, DNAJA3 and its

associated Hsp70 are sequestered within a

cytoplasmic "hot spot" structure, a subcellular

distribution that is characteristic of Tax in HEK

cells (Cheng et al., 2001).

APC interaction: the amino terminus of APC

interacted with DNAJA3 at the mitochondria in

vivo in colorectal cancer cell lines (Qian et al.,

2010).

Chaperone function: DNAJA3 isoforms were also

shown to exhibit a conserved mitochondrial DnaJ-

like function substituting for the yeast

mitochondrial DnaJ-like protein Mdj1p (Lu et al.,

2006).

Mitochondrial biogenesis: DNAJA3 was shown to

be crucial for mitochondrial biogenesis partly

through chaperone activity on DNA polymerase

gamma (Hayashi et al., 2006). Mice deficient in

Dnaja3 developed dilated cardiomyopathy (DCM)

and died before 10 weeks of age (Hayashi et al.,

2006). Progressive respiratory chain deficiency and

decreased copy number of mitochondrial DNA

were observed in cardiomyocytes lacking Dnaja3

(Hayashi et al., 2006).

Tumor suppressor pathways APC: DNAJA3 directly bound to the APC tumor

suppressor protein and promoted a physiological

function for APC that was independent of APC's

involvement in beta-catenin degradation or

regulation of the actin cytoskeleton (Kurzik-Dumke

and Czaja, 2007).

pVHL: TID1L directly interacted with von Hippel-

Lindau protein and enhanced the interaction

between HIF-1 alpha and pVHL. This resulted in

destabilization of HIF-1 alpha protein, decreased

vascular endothelial growth factor expression, and

inhibition of angiogenesis (Bae et al., 2005).

Interferon-gamma: DNAJA3L and DNAJA3S

interacted with the interferon-gamma receptor chain

IFN-gammaR2 and modulated IFN-gamma-

mediated transcriptional activity. Furthermore, IFN-

gamma treatment reduced the interaction between

Hsp70/Hsc70 and DNAJA3 (Sarkar et al., 2001).

Oncogenic pathways Erb-B2/HER2: DNAJA3 physically interacted

with the signaling domain of ErbB-2 and ErbB-2

were shown to colocalize in mammary carcinoma

cells (SK-BR-3). Overexpression of DNAJA3

induced growth arrest and apoptosis in ErbB-2-

overexpressing breast cancer cells; the DNAJ and

C-terminal domains of DNAJA3 were critical for

mediating apoptosis. Downregulation of

ERK1/ERK2 and BMK1 MAPK pathways also

contributed to apoptosis. DNAJA3S negatively

regulated ErbB-2 signaling pathways by enhancing

the degradation of ErbB-2. Finally, increased

cellular DNAJA3 inhibited the growth of ErbB-2-

dependent tumors in mice (Kim et al., 2004).

Mammary tumor tissue from breast cancer patients

and transgenic mice carrying the rat HER-2/neu

oncogene suggest that DNAJA3 suppresses ErbB-2

in breast cancers (Kurzik-Dumke et al., 2010).

NF-kappaB: expression of DNAJA3 was

upregulated upon cellular senescence in rat and

mouse embryo fibroblasts, as well as in premature

senescence of REF52 cells triggered by activated

ras. Conversely, spontaneous immortalization of rat

embryo fibroblasts was suppressed upon ectopic

expression of DNAJA3. Suppression of endogenous

DNAJA3 activity alleviated the suppression of

tumor necrosis factor alpha-induced NF-kappaB

activity by DNAJA3. These results suggest that

DNAJA3 contributes to senescence by repressing

NF-kappa B signaling (Tarunina et al., 2004).

DNAJA3 repressed NF-kappaB activity induced by

Tax, tumor necrosis factor alpha (TNFalpha), and

Bcl10. DNAJA3 specifically suppressed serine

phosphorylation of IkappaBalpha by activated

IkappaB kinase beta (IKKbeta).

The suppressive activity of DNAJA3 on IKKbeta

required a functional J domain that mediates

association with heat shock proteins and resulted in

prolonging the half-life of the NF-kappaB inhibitors

IkappaBalpha and IkappaBbeta (Cheng et al.,

2002).

AKT: overexpression of DNAJA3 in HNSCC cells

inhibited cell proliferation, migration, invasion,

anchorage-independent growth, and

xenotransplantation tumorigenicity. Overexpression

of DNAJA3 attenuated EGFR activity and blocked

the activation of AKT in HNSCC cells, which are

known to be involved in the regulation of survival

in HNSCC cells. Conversely, ectopic expression of

constitutively active AKT greatly reduced apoptosis

induced by DNAJA3 overexpression (Chen et al.,

2009).

JAK2: DNAJA3L and DNAJA3S interacted with

Jak2 in vivo in COS-1 cells. Interaction was

primarily in the cytoplasm and predominantly with

the active kinase domain of Jak2 (Sarkar et al.,

2001).

Page 24: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 200

c-MET receptor tyrosine kinase (MetR): MetR

interacted with DNAJA3L and DNAJA3S, but

showed preferential binding to DNAJA3S.

Interaction occurred through the J domain. In RCC

cells, overexpression of DNAJA3S enhanced HGF-

mediated MetR autophosphorylation, while

DNAJA3L showed modest inhibition of MetR

activity. Modulation of MetR phosphorylation

levels was independent of pVHL. DNAJA3S

enhanced HGF-mediated cell migration and

modulated HGF-mediated MAPK phosphorylation.

DNAJA3 knockdown inhibited MetR activation

and migration in response to HGF (Copeland et al.,

2011).

Signal transducers and activators of

transcription (STAT) 5b: DNAJA3 specifically

interacted with STAT5b but not STAT5a in

hematopoietic cell lines. Interaction involved the

DNAJ domain. DNAJA3 negatively regulated the

expression and transcriptional activity of STAT5b

and suppressed the growth of hematopoietic cells

transformed by an oncogenic form of STAT5b

(Dhennin-Duthille et al., 2011).

Cell Fate DNAJA3 was shown to be required for the T-cell

transition from double-negative 3 to double-

positive stages. Mice with dnaja3 specifically

deleted in T cells developed thymic atrophy, with

dramatic reduction of double-positive and single-

positive thymocytes in the dnaja3(-/-) thymus.

DNAJA3 deficiency inhibited cell proliferation and

enhanced cell death of DN4 cells. The expression

profile of genes involved in cytokine receptor

signaling was altered in DN4 T-cells. Expression of

human bcl-2 transgene restored T lymphocyte

proliferation and differentiation in the dnaja3

knockout mice. These results suggest that dnaja3 is

critical in early thymocyte development, especially

during transition from the DN3 to double-positive

stages, possibly through its regulation of bcl-2

expression, which provides survival signals.

Homology

Mouse (laboratory): Dnaja3

Rat: Dnaja3

Cattle: DNAJA3

Chimpanzee: DNAJA3

Dog (domestic): DNAJA3

Mutations

Note

The SF767 glioma cell line exhibits an aberrant 52

kD molecular weight protein. Sequence analysis of

cDNA generated from this line showed two

mutations: an additional thymine at nucleotide

position 1438 and an additional cytosine at

nucleotide position 1449. These mutations alter the

reading frame of the DNAJA3 sequence,

introducing an additional 71 amino acids following

the penultimate threonine residue at position 479.

The mutations appear to increase the steady-state

abundance of the mutant protein, resulting in

aberrantly high levels of the DNAJA3 mutant

variant (Trentin et al., 2004).

Implicated in

Colon cancer

Disease

DNAJA3 and INT6 protein levels, as well as

DNAJA3 and Patched protein levels, were

positively correlated in human colon tumor tissues

(Traicoff et al., 2007). However, there were no

correlations between DNAJA3 and p53, c-Jun, or

phospho-c-Jun protein levels (Traicoff et al., 2007).

These results were demonstrated by multiplex

tissue immunoblotting of tissue microarrays

(Traicoff et al., 2007).

Progression of colorectal cancers correlated with

overexpression and loss of polarization of

expression of DNAJA3. These changes were

associated with upregulation of Hsp70 and loss of

compartmentalization of APC (Kurzik-Dumke et

al., 2008).

Breast cancer

Disease

DNAJA3 protein expression showed a strong

correlation with negative or weakly positive

expression of ErbB2 in human breast cancer tissue

samples. High DNAJA3 levels were strongly

correlated with high levels of CHIP (carboxyl

terminus of heat shock cognate 70 interacting

protein). Lower expression of DNAJA3 had a

higher risk of unfavorable tumor grade, later

pathological stage, larger tumor size, and

microscopic features of a more malignant histology

(Jan et al., 2011). Higher expression of DNAJA3

correlated with increased 10-year overall and

disease-free survival rate (Jan et al., 2011).

The expression of the three DNAJA3 isoform

transcripts was examined in human breast cancer

carcinomas by RT-PCR. Aberrant expression of all

three forms correlated with malignant

transformation. Furthermore, elevated DNAJA3L

expression was associated with less aggressive

tumors (Kurzik-Dumke et al., 2010).

Immunohistochemical analysis demonstrated high

levels of DNAJA3 protein in tumors of the luminal

A subtype, but significantly lower levels of

DNAJA3 protein in the luminal B subtype, triple

negative tumors, and the HER-2 subtype which

overexpresses HER-2 (Kurzik-Dumke et al., 2010).

Multiplex tissue immunoblotting of human breast

tumor tissue microarrays was used to test

correlations between DNAJA3 protein levels and a

set of tumor suppressor proteins. DNAJA3 protein

levels showed strongly positive correlations with

p53, Patched, and INT6 proteins (Traicoff et al.,

2007). Additionally, DNAJA3 protein levels

Page 25: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 201

showed moderate positive correlations with c-Jun

and phospho-c-Jun proteins (Traicoff et al., 2007).

Head and neck squamous cell carcinoma (HNSCC)

Disease

The clinical association between DNAJA3

expression and progression of HNSCC was

explored using immunohistochemical analysis of

primary HNSCC patient tumor tissue. DNAJA3

expression was negatively associated with tumor T

stage, overall stage, survival, and recurrence.

Patients with higher expression of DNAJA3 were

predicted to have better overall survival than those

with low or undetectable expression of DNAJA3

protein (Chen et al., 2009). Highly malignant

HNSCC cell lines also demonstrated low or

undetectable levels of DNAJA3, in contrast to less

aggressive lines where DNAJA3 protein was easily

detected (Chen et al., 2009).

Ovarian cancer

Disease

Multiplex tissue immunoblotting of ovarian tumor

tissues demonstrated that DNAJA3 protein levels

showed moderate positive correlations with INT6,

c-Jun, phospho-c-Jun, and p53. No correlations

were observed between DNAJA3 and Patched

(Traicoff et al., 2007).

Lung cancer

Disease

Multiplex tissue immunoblotting of lung tumor

tissues demonstrated that DNAJA3 protein levels

were strongly correlated with INT6. DNAJA3

protein levels were moderately correlated with

Patched, c-Jun, and p53. However, DNAJA3

proteins showed negative correlation with phospho-

c-Jun in these samples (Traicoff et al., 2007).

Cardiomyopathy

Note

Mice deficient in Dnaja3 developed dilated

cardiomyopathy (DCM) and died before 10 weeks

of age (Hayashi et al., 2006). Progressive

respiratory chain deficiency and decreased copy

number of mitochondrial DNA were observed in

cardiomyocytes lacking Dnaja3 (Hayashi et al.,

2006).

References Schilling B, De-Medina T, Syken J, Vidal M, Münger K. A novel human DnaJ protein, hTid-1, a homolog of the Drosophila tumor suppressor protein Tid56, can interact with the human papillomavirus type 16 E7 oncoprotein. Virology. 1998 Jul 20;247(1):74-85

Syken J, De-Medina T, Münger K. TID1, a human homolog of the Drosophila tumor suppressor l(2)tid, encodes two mitochondrial modulators of apoptosis with opposing functions. Proc Natl Acad Sci U S A. 1999 Jul 20;96(15):8499-504

Cheng H, Cenciarelli C, Shao Z, Vidal M, Parks WP, Pagano M, Cheng-Mayer C. Human T cell leukemia virus type 1 Tax associates with a molecular chaperone complex containing hTid-1 and Hsp70. Curr Biol. 2001 Nov 13;11(22):1771-5

Sarkar S, Pollack BP, Lin KT, Kotenko SV, Cook JR, Lewis A, Pestka S. hTid-1, a human DnaJ protein, modulates the interferon signaling pathway. J Biol Chem. 2001 Dec 28;276(52):49034-42

Trentin GA, Yin X, Tahir S, Lhotak S, Farhang-Fallah J, Li Y, Rozakis-Adcock M. A mouse homologue of the Drosophila tumor suppressor l(2)tid gene defines a novel Ras GTPase-activating protein (RasGAP)-binding protein. J Biol Chem. 2001 Apr 20;276(16):13087-95

Yin X, Rozakis-Adcock M. Genomic organization and expression of the human tumorous imaginal disc (TID1) gene. Gene. 2001 Oct 31;278(1-2):201-10

Cheng H, Cenciarelli C, Tao M, Parks WP, Cheng-Mayer C. HTLV-1 Tax-associated hTid-1, a human DnaJ protein, is a repressor of Ikappa B kinase beta subunit. J Biol Chem. 2002 Jun 7;277(23):20605-10

Eom CY, Lehman IR. The human DnaJ protein, hTid-1, enhances binding of a multimer of the herpes simplex virus type 1 UL9 protein to oris, an origin of viral DNA replication. Proc Natl Acad Sci U S A. 2002 Feb 19;99(4):1894-8

Sehgal PB. Plasma membrane rafts and chaperones in cytokine/STAT signaling. Acta Biochim Pol. 2003;50(3):583-94

Syken J, Macian F, Agarwal S, Rao A, Münger K. TID1, a mammalian homologue of the drosophila tumor suppressor lethal(2) tumorous imaginal discs, regulates activation-induced cell death in Th2 cells. Oncogene. 2003 Jul 24;22(30):4636-41

Edwards KM, Münger K. Depletion of physiological levels of the human TID1 protein renders cancer cell lines resistant to apoptosis mediated by multiple exogenous stimuli. Oncogene. 2004 Nov 4;23(52):8419-31

Kim SW, Chao TH, Xiang R, Lo JF, Campbell MJ, Fearns C, Lee JD. Tid1, the human homologue of a Drosophila tumor suppressor, reduces the malignant activity of ErbB-2 in carcinoma cells. Cancer Res. 2004 Nov 1;64(21):7732-9

Tarunina M, Alger L, Chu G, Munger K, Gudkov A, Jat PS. Functional genetic screen for genes involved in senescence: role of Tid1, a homologue of the Drosophila tumor suppressor l(2)tid, in senescence and cell survival. Mol Cell Biol. 2004 Dec;24(24):10792-801

Trentin GA, He Y, Wu DC, Tang D, Rozakis-Adcock M. Identification of a hTid-1 mutation which sensitizes gliomas to apoptosis. FEBS Lett. 2004 Dec 17;578(3):323-30

Bae MK, Jeong JW, Kim SH, Kim SY, Kang HJ, Kim DM, Bae SK, Yun I, Trentin GA, Rozakis-Adcock M, Kim KW. Tid-1 interacts with the von Hippel-Lindau protein and modulates angiogenesis by destabilization of HIF-1alpha. Cancer Res. 2005 Apr 1;65(7):2520-5

Cheng H, Cenciarelli C, Nelkin G, Tsan R, Fan D, Cheng-Mayer C, Fidler IJ. Molecular mechanism of hTid-1, the human homolog of Drosophila tumor suppressor l(2)Tid, in the regulation of NF-kappaB activity and suppression of tumor growth. Mol Cell Biol. 2005 Jan;25(1):44-59

Kim SW, Hayashi M, Lo JF, Fearns C, Xiang R, Lazennec G, Yang Y, Lee JD. Tid1 negatively regulates the migratory potential of cancer cells by inhibiting the production of interleukin-8. Cancer Res. 2005 Oct 1;65(19):8784-91

Kittler R, Pelletier L, Ma C, Poser I, Fischer S, Hyman AA, Buchholz F. RNA interference rescue by bacterial artificial

Page 26: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3) Traicoff JL, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 202

chromosome transgenesis in mammalian tissue culture cells. Proc Natl Acad Sci U S A. 2005 Feb 15;102(7):2396-401

Liu HY, MacDonald JI, Hryciw T, Li C, Meakin SO. Human tumorous imaginal disc 1 (TID1) associates with Trk receptor tyrosine kinases and regulates neurite outgrowth in nnr5-TrkA cells. J Biol Chem. 2005 May 20;280(20):19461-71

Lo JF, Zhou H, Fearns C, Reisfeld RA, Yang Y, Lee JD. Tid1 is required for T cell transition from double-negative 3 to double-positive stages. J Immunol. 2005 May 15;174(10):6105-12

Hayashi M, Imanaka-Yoshida K, Yoshida T, Wood M, Fearns C, Tatake RJ, Lee JD. A crucial role of mitochondrial Hsp40 in preventing dilated cardiomyopathy. Nat Med. 2006 Jan;12(1):128-32

Lu B, Garrido N, Spelbrink JN, Suzuki CK. Tid1 isoforms are mitochondrial DnaJ-like chaperones with unique carboxyl termini that determine cytosolic fate. J Biol Chem. 2006 May 12;281(19):13150-8

Qiu XB, Shao YM, Miao S, Wang L. The diversity of the DnaJ/Hsp40 family, the crucial partners for Hsp70 chaperones. Cell Mol Life Sci. 2006 Nov;63(22):2560-70

Sohn SY, Kim SB, Kim J, Ahn BY. Negative regulation of hepatitis B virus replication by cellular Hsp40/DnaJ proteins through destabilization of viral core and X proteins. J Gen Virol. 2006 Jul;87(Pt 7):1883-91

Wang L, Tam JP, Liu DX. Biochemical and functional characterization of Epstein-Barr virus-encoded BARF1 protein: interaction with human hTid1 protein facilitates its maturation and secretion. Oncogene. 2006 Jul 20;25(31):4320-31

Kurzik-Dumke U, Czaja J. Htid-1, the human homolog of the Drosophila melanogaster l(2)tid tumor suppressor, defines a novel physiological role of APC. Cell Signal. 2007 Sep;19(9):1973-85

Traicoff JL, Chung JY, Braunschweig T, Mazo I, Shu Y, Ramesh A, D'Amico MW, Galperin MM, Knezevic V, Hewitt SM. Expression of EIF3-p48/INT6, TID1 and Patched in cancer, a profiling of multiple tumor types and correlation of expression. J Biomed Sci. 2007 May;14(3):395-405

Wakabayashi Y, Mao JH, Brown K, Girardi M, Balmain A. Promotion of Hras-induced squamous carcinomas by a polymorphic variant of the Patched gene in FVB mice. Nature. 2007 Feb 15;445(7129):761-5

Kurzik-Dumke U, Hörner M, Czaja J, Nicotra MR, Simiantonaki N, Koslowski M, Natali PG. Progression of colorectal cancers correlates with overexpression and loss of polarization of expression of the htid-1 tumor suppressor. Int J Mol Med. 2008 Jan;21(1):19-31

Linnoila J, Wang Y, Yao Y, Wang ZZ. A mammalian homolog of Drosophila tumorous imaginal discs, Tid1, mediates agrin signaling at the neuromuscular junction. Neuron. 2008 Nov 26;60(4):625-41

Song Y, Balice-Gordon R. New dogs in the dogma: Lrp4 and Tid1 in neuromuscular synapse formation. Neuron. 2008 Nov 26;60(4):526-8

Chen CY, Chiou SH, Huang CY, Jan CI, Lin SC, Hu WY, Chou SH, Liu CJ, Lo JF. Tid1 functions as a tumour suppressor in head and neck squamous cell carcinoma. J Pathol. 2009 Nov;219(3):347-55

Ahn BY, Trinh DL, Zajchowski LD, Lee B, Elwi AN, Kim SW. Tid1 is a new regulator of p53 mitochondrial translocation and apoptosis in cancer. Oncogene. 2010 Feb 25;29(8):1155-66

Kurzik-Dumke U, Hörner M, Nicotra MR, Koslowski M, Natali PG. In vivo evidence of htid suppressive activity on ErbB-2 in breast cancers over expressing the receptor. J Transl Med. 2010 Jun 17;8:58

Maselli RA, Arredondo J, Cagney O, Ng JJ, Anderson JA, Williams C, Gerke BJ, Soliven B, Wollmann RL. Mutations in MUSK causing congenital myasthenic syndrome impair MuSK-Dok-7 interaction. Hum Mol Genet. 2010 Jun 15;19(12):2370-9

Qian J, Perchiniak EM, Sun K, Groden J. The mitochondrial protein hTID-1 partners with the caspase-cleaved adenomatous polyposis cell tumor suppressor to facilitate apoptosis. Gastroenterology. 2010 Apr;138(4):1418-28

Trinh DL, Elwi AN, Kim SW. Direct interaction between p53 and Tid1 proteins affects p53 mitochondrial localization and apoptosis. Oncotarget. 2010 Oct;1(6):396-404

Copeland E, Balgobin S, Lee CM, Rozakis-Adcock M. hTID-1 defines a novel regulator of c-Met Receptor signaling in renal cell carcinomas. Oncogene. 2011 May 12;30(19):2252-63

Dhennin-Duthille I, Nyga R, Yahiaoui S, Gouilleux-Gruart V, Régnier A, Lassoued K, Gouilleux F. The tumor suppressor hTid1 inhibits STAT5b activity via functional interaction. J Biol Chem. 2011 Feb 18;286(7):5034-42

Jan CI, Yu CC, Hung MC, Harn HJ, Nieh S, Lee HS, Lou MA, Wu YC, Chen CY, Huang CY, Chen FN, Lo JF. Tid1, CHIP and ErbB2 interactions and their prognostic implications for breast cancer patients. J Pathol. 2011 Nov;225(3):424-37

This article should be referenced as such:

Traicoff JL, Hewitt SM, Chung JY. DNAJA3 (DnaJ (Hsp40) homolog, subfamily A, member 3). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):194-202.

Page 27: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 203

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

MYEOV (myeloma overexpressed (in a subset of t(11;14) positive multiple myelomas)) Jérôme Moreaux

INSERM U1040, institut de recherche en biotherapie, CHRU Saint Eloi, 80 Av Augustain Fliche,

34295 Montpellier CEDEX 5, France (JM)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/MYEOVID395.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI MYEOVID395.txt This article is an update of : Janssen JWG. MYEOV myeloma overexpressed (in a subset of t(11;14) positive multiple myelomas). Atlas Genet Cytogenet Oncol Haematol 2003;7(1) This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: OCIM

HGNC (Hugo): MYEOV

Location: 11q13.3

Local order: 350 kb centromeric of cyclin D1.

Note

Detected by application of the NIH/3T3

tumorigenicity assay. However MYEOV cDNA

was not positive in NIH/3T3 assay.

DNA/RNA

Note

The MYEOV gene was originally isolated by the

application of the NIH/3T3 tumorigenicity assay

with DNA from a gastric carcinoma. The

chromosomal region 11q13 is frequently associated

with genetic rearrangements in a large number of

human malignancies, including B-cell malignancies

and overexpression of MYEOV is frequently

observed in breast tumors and oral, esophageal

squamous cell carcinomas and multiple myeloma.

The presence of functional domains such as RNP-1

(motif typical of RNA binding protein) and the

studies of the short hydrophobic regions and of the

C-terminal leucine/isoleucine tail showed that

MYEOV might be directed to the membrane.

MYEOV small interfering RNA (siRNA) decreased

proliferation of gastric cancer cells, colon cancer

cell lines and multiple myeloma cells in vitro.

Description

2 exons; 3,5 kb transcript represents unspliced

mRNA.

Transcription

Main transcript 2,8 kb (broad band because of

alternative splice products); minor transcript 3,5 kb;

coding sequence 313 or 255 amino acids. In normal

tissues hardly any expression detectable. High

expression in a subset of multiple myeloma cell

lines with a t(11;14)(q13;q32) and in breast tumors

and esophageal squamous cell carcinomas with or

without 11q13 amplification.

Pseudogene

No pseudogenes have been reported for MYEOV.

Page 28: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

MYEOV (myeloma overexpressed (in a subset of t(11;14) positive multiple myelomas))

Moreaux J

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 204

Protein

Description

313 or 255 amino acids; contains one RNP-1 motif

and 6 regions that might function as a

transmembrane domain. Leucine-rich stretch at C-

terminal.

Expression

5' UTR inhibits efficient translation of the protein.

Localisation

In endoplasmic reticulum and mitochondria.

Homology

No known homology.

Implicated in

t(11;14)(q13;q32)

Disease

Subset of multiple myeloma cell lines with

t(11;14)(q13;q32).

Cytogenetics

MYEOV overexpression due to juxtaposition to the

5' enhancer or the most telomeric 3' enhancer of the

immunoglobulin heavy chain (IgH).

11q13 amplification and/or overexpression

Disease

Breast cancer; esophageal squamous cell

carcinomas.

Prognosis

MYEOV DNA amplification correlated with

estrogen and progesterone receptor-positive cancer,

invasive lobular carcinoma type and axillary nodal

involvement. In contrast to Cyclin D1

amplification, no association with disease outcome

could be found.

Multiple myeloma

Prognosis

In a cohort of 171 myeloma patients, patients with

MYEOVabsent

MMC have an increased event-free

survival compared to patients with MYEOVpresent

MMC, after high-dose therapy and stem cell

transplantation and a trend for increased overall

survival. In a Cox proportional hazard model,

MYEOV expression in MMC is predictive for

event-free survival for patients independently of

International Staging System stage, t(4;14)

translocation, albumin, or B2M serum levels. In a

second independent cohort of 208 patients (LR-

TT3, from the University of Arkansas for Medical

Sciences (Little Rock, AR, USA)), MYEOV had a

"present" call in MMCs of 73% of patients. Patients

with MYEOVabsent

MMCs had a significant better

overall survival in the LR-TT3 cohort.

Oncogenesis

In a cohort of 171 patients, MMC of 79% of the

patients with newly diagnosed MM express

MYEOV gene. A treatment with 5-aza-2'-

deoxycytidine of 2 MYEOVabsent

myeloma cell lines

induced MYEOV expression without affecting that

in the MYEOVpresent

myeloma cells. MYEOV

siRNA did not significantly induce apoptosis in

myeloma cell lines, but it blocked the cell cycle

entry into the S-phase.

Colon cancer

Oncogenesis

Knockout of MYEOV RNA (siRNA) has been

shown to decrease proliferation of colon cancer cell

lines in vitro. Furthermore, in colon cancer,

MYEOV stimulates colorectal cancer cell migration

in vitro. MYEOV expression is enhanced by PGE2

treatment in colorectal cancer cells.

Gastric cancer

Oncogenesis

Knockout of MYEOV RNA (siRNA) has been

shown to decrease proliferation and invasion of

gastric cancer cells in vitro.

Neuroblastoma

Oncogenesis

MYEOV is a candidate gene target in

neuroblastoma that was identified by chromosomal

gain 11q13 through SNP analysis. MYEOV

expression was analyzed in 55 neuroblastoma

samples including 25 cell lines. MYEOV was

shown to be upregulated in 11 out of 25

neuroblastoma cell lines and 7 out of 20 fresh

tumors. Knockout of MYEOV RNA (siRNA) has

been shown to decrease proliferation of

neuroblastoma cell line in vitro.

Oral squamous cell carcinoma

Oncogenesis

Gain of 11q13 was significantly associated with

cervical lymph node metastasis in oral squamous

cell carcinoma (54 patients included in the study).

Copy number amplification of MYEOV is

associated with cervical lymph node metastasis in

oral squamous cell carcinoma. Lymph node

metastasis is associated with a significant decrease

of 5-years survival in oral squamous cell

carcinoma.

References Janssen JW, Vaandrager JW, Heuser T, Jauch A, Kluin PM, Geelen E, Bergsagel PL, Kuehl WM, Drexler HG, Otsuki T, Bartram CR, Schuuring E. Concurrent activation of a novel putative transforming gene, myeov, and cyclin D1 in a subset of multiple myeloma cell lines with t(11;14)(q13;q32). Blood. 2000 Apr 15;95(8):2691-8

Janssen JW, Cuny M, Orsetti B, Rodriguez C, Vallés H, Bartram CR, Schuuring E, Theillet C. MYEOV: a candidate gene for DNA amplification events occurring centromeric to

Page 29: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

MYEOV (myeloma overexpressed (in a subset of t(11;14) positive multiple myelomas))

Moreaux J

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 205

CCND1 in breast cancer. Int J Cancer. 2002 Dec 20;102(6):608-14

Janssen JW, Imoto I, Inoue J, Shimada Y, Ueda M, Imamura M, Bartram CR, Inazawa J. MYEOV, a gene at 11q13, is coamplified with CCND1, but epigenetically inactivated in a subset of esophageal squamous cell carcinomas. J Hum Genet. 2002;47(9):460-4

Leyden J, Murray D, Moss A, Arumuguma M, Doyle E, McEntee G, O'Keane C, Doran P, MacMathuna P. Net1 and Myeov: computationally identified mediators of gastric cancer. Br J Cancer. 2006 Apr 24;94(8):1204-12

Moss AC, Lawlor G, Murray D, Tighe D, Madden SF, Mulligan AM, Keane CO, Brady HR, Doran PP, MacMathuna P. ETV4 and Myeov knockdown impairs colon cancer cell line proliferation and invasion. Biochem Biophys Res Commun. 2006 Jun 23;345(1):216-21

Lawlor G, Doran PP, MacMathuna P, Murray DW. MYEOV (myeloma overexpressed gene) drives colon cancer cell migration and is regulated by PGE2. J Exp Clin Cancer Res. 2010 Jun 22;29:81

Moreaux J, Hose D, Bonnefond A, Reme T, Robert N, Goldschmidt H, Klein B. MYEOV is a prognostic factor in multiple myeloma. Exp Hematol. 2010 Dec;38(12):1189-1198.e3

Sugahara K, Michikawa Y, Ishikawa K, Shoji Y, Iwakawa M, Shibahara T, Imai T. Combination effects of distinct cores in 11q13 amplification region on cervical lymph node metastasis of oral squamous cell carcinoma. Int J Oncol. 2011 Oct;39(4):761-9

Takita J, Chen Y, Okubo J, Sanada M, Adachi M, Ohki K, Nishimura R, Hanada R, Igarashi T, Hayashi Y, Ogawa S. Aberrations of NEGR1 on 1p31 and MYEOV on 11q13 in neuroblastoma. Cancer Sci. 2011 Sep;102(9):1645-50

This article should be referenced as such:

Moreaux J. MYEOV (myeloma overexpressed (in a subset of t(11;14) positive multiple myelomas)). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):203-205.

Page 30: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 206

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

PCNA (proliferating cell nuclear antigen) Ivaylo Stoimenov, Thomas Helleday

Department of Genetics Microbiology and Toxicology, Stockholm University, S-106 91 Stockholm,

Sweden (IS), Department of Genetics Microbiology and Toxicology, Stockholm University, S-106 91

Stockholm, Sweden; Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford,

OX3 7DQ, UK (TH)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/PCNAID41670ch20p12.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI PCNAID41670ch20p12.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: MGC8367

HGNC (Hugo): PCNA

Location: 20p12.3

DNA/RNA

Description

The PCNA gene is situated on human chromosome

20 and it spans about 12 kb. It is a single-copy

gene, however, several pseudogenes have been

noted. The precise localization of the PCNA gene is

at the border of two histological G-bands (p12.3

and p13) (Webb et al., 1990), thus it is reported in

both locations depending on the probe used. The

human PCNA gene was first cloned and

characterized in 1989 by Travali and co-workers

(Travali et al., 1989).

Transcription

There are two reported gene transcripts, which

encode the same protein.

PCNA transcript variant 1 is 1355 bp long after the

completion of mRNA splicing. It has NCBI

Reference Sequence code NM_002592.2 (NCBI).

The PCNA transcript variant 1 has seven exons, six

of which are contributing to the protein sequence.

The first intron is relatively large in comparison

with the other PCNA transcript variant. Following

the splicing the length of the transcript is shortened

to about 12% of that of the initial transcript. The

translation starts from the middle of the 2nd

exon

and ends in the beginning of 7th

exon. The product

is a full length protein, designated as NP_002583.1

(NCBI), with 261 amino acids.

PCNA transcript variant 2 is 1319 bp long after the

completion of mRNA splicing.

The localisation of the PCNA gene (in red) at the interface between 20p12.3 and 20p13 histological bands on chromosome 20.

NCBI Reference

Sequence

Length

(unspliced)

Length

(spliced) Exons Protein AA

PCNA transcript variant 1 NM_002592.2 11670 bp 1355 bp 7 NP_002583.1 261

PCNA transcript variant 2 NM_182649.1 5049 bp 1319 bp 6 NP_872590.1 261

Page 31: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

PCNA (proliferating cell nuclear antigen) Stoimenov I, Helleday T

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 207

It has NCBI Reference Sequence code

NM_182649.1 (NCBI). The PCNA transcript

variant 2 has six exons, which are contributing to

the protein sequence. After the splicing the length

of the transcript is shortened to about 26% of that of

the initial transcript. Translation starts from the end

of the 1st exon and ends in the beginning of 7

th

exon. The product is a full length protein,

designated as NP_872590.1 (NCBI), with 261

amino acids.

Pseudogene

PCNAP - one pseudogene on human chromosome

X - p11 (Ku et al., 1989; Webb et al., 1990).

PCNAP1 and PCNAP2 - two pseudogenes in

tandem on human chromosome 4 - q24 (Taniguchi

et al., 1996).

There are several other possible pseudogenes:

LOC390102 on chromosome 11 - p15.1 (Webb et

al., 1990), LOC392454 on chromosome X - p11.3

(Ku et al., 1989; Webb et al., 1990).

Protein

Description

The human PCNA protein is a polypeptide of 261

amino acids and theoretical molecular weight of

about 29 kDa. The functional protein is a

homotrimer, build from three identical units

interacting head-to-tail and forming a doughnut

shaped molecule. There is an evidence for the

existence of a double homotrimer in vivo

(Naryzhny et al., 2005).

Expression

Expressed in nearly all proliferating tissues with

high levels detected in thymus, bone marrow, foetal

liver and certain cells of the small intestine and

colon.

Localisation

PCNA is exclusively localized in the nucleus. It can

be detected by immunofluorescence in all

proliferating nuclei as discrete nuclear foci,

representing sites of ongoing DNA replication

and/or DNA repair.

Function

PCNA was originally discovered as an antigen,

reacting with antibodies derived from sera of

patients with systemic lupus erythematosus

(Miyachi et al., 1978). The first assigned function

of the PCNA protein is as an auxiliary factor of

polymerase delta (Tan et al., 1986; Prelich et al.,

1987). Later it was suggested that PCNA functions

as a cofactor to many other eukaryotic polymerases

such as polymerase epsilon, polymerase beta and

several specialised polymerases known as

translesion synthesis polymerases (eta, kappa,

lambda, theta, etc.), with which PCNA is known to

interact (Naryzhny, 2008). The role of PCNA in

DNA replication is thoroughly investigated and

PCNA is proposed to serve as a switch between the

priming polymerase alpha and replicative

polymerases (delta and epsilon) and functioning as

a cofator of the latter polymerases. Complementary

to enhancing the processivity of DNA replication,

PCNA is known to coordinate the maturation of

Okazaki fragments through interaction with FEN1

and stimulation of the flap endonuclease activity.

PCNA interacts with large number of proteins,

suggesting many functions in vivo (Naryzhny,

2008; Stoimenov and Helleday, 2009). There is

evidence, derived from experiments in yeast, that

PCNA may be involved in the establishment of

sister chromatid cohesion in S phase of the cell

cycle (Moldovan et al., 2006). PCNA is an

indispensable factor for different DNA repair

pathways including mismatch repair, nucleotide

excision repair and sub-pathways of base excision

repair. There is a growing body of evidence for the

function of PCNA in the chromatin remodelling

and organisation. The interaction of PCNA and

CAF1 is in the heart of the nucleosome assembly,

while the chromatin modification is also known to

be regulated by PCNA through the known

interaction with DNMT1 and HDAC1.

PCNA and mapped interactions with several proteins (D-type of cyclins, CDKN1A, FEN1, RFC complex, polymerase epsilon and polymerase delta). Two residues are highlighted, lysine at position 164 (site of ubiquitylation) and tyrosine at position 211 (site of

phosphorylation).

Page 32: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

PCNA (proliferating cell nuclear antigen) Stoimenov I, Helleday T

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 208

One of the most stable interactions of PCNA is that

with the cyclin-kinase inhibitor CDKN1A, which

suggests a role of PCNA in the cell cycle

progression. Another evidence for the involvement

of PCNA in the cell cylcle control is the interaction

with cyclin-D. Several amino-acid residues are

post-translationally modified, suggesting even more

complex functions (Stoimenov and Helleday,

2009). PCNA could be subjected to post-

translational phosphorylation, acetylation,

methylation, ubiquitylation and SUMOylation.

Implicated in

Note

The absence of the proliferating nuclear cell antigen

(PCNA) protein is embryonic lethal in mice (Roa et

al., 2008; Peled et al., 2008). The embryonic

lethality in mice also suggests a critical importance

of the PCNA protein for humans at least in

proliferating tissues (Moldovan et al., 2007). The

knockout mice for PCNA (Pcna-/-

) are dying in

embryonic state, consistent with the role of PCNA

in orchestrating DNA replication (Moldovan et al.,

2007). In addition to this fact, there are no known

mutations of the PCNA protein in humans, which

therefore leads to a speculation that PCNA is so

vital that any alternation of its sequence would have

deleterious consequences. One suggestion for such

essential function is the fact that both sequences of

the PCNA protein and of the respective gene are

highly conserved during evolution (Stoimenov and

Helleday, 2009). Indeed, a human population study

of PCNA polymorphisms shows only 7 intronic

single nucleotide polymorphisms (SNP) and 2

synonymous exonic SNPs (Ma et al., 2000).

According to OMIM and Human Locus Specific

Mutation Databases there is no known disease,

which is caused by mutation or loss of function of

the PCNA protein.

The only implication of PCNA in human disease is

as a prognostic or diagnostic marker, sometimes

used together with other markers. The utilisation of

PCNA as a marker is very much restricted to an

illustration of proliferation potential and therefore

cannot be specific for any disease. However, PCNA

is indeed used as a prognostic and diagnostic

marker in several human diseases in clinical

practice, as shown below. The list is far from

complete since any human disease associated with

proliferation could utilise PCNA as a marker.

Primary breast cancer

Note

A group of patients with high PCNA labeling index

was associated with poor overall survival compared

with the low PCNA labeling index group in several

immunohistochemical studies (Horiguchi et al.,

1998; Chu et al., 1998). PCNA labeling index is

stated to be an independent predictor in primary

breast cancer patients (Horiguchi et al., 1998) with

a prognostic value (Chu et al., 1998).

Chronic lymphoid leukemia (CLL)

Note

There are attempts to correlate the levels of the

PCNA protein in cells derived from patients with

chronic lymphoid leukemia and the prognosis of

survival (del Giglio et al., 1992; Faderl et al., 2002).

The high level of PCNA in the cells of CLL

patients suggests a higher proliferative activity and

potentially shorter survival (del Giglio et al., 1992).

Intracellular levels of PCNA protein can be used as

marker to predict clinical behaviour and overall

survival in patients with CLL (Faderl et al., 2002).

Non-Hodgkin's lymphoma

Note

In studies conducting immunohistochemical

staining of materials from patients with non-

Hodgkin's lymphoma, PCNA labeling index

together with AgNOR score can be used to predict

overall survival (Korkolopoulou et al., 1998).

PCNA is the only independent predictor of the post-

relapse survival and the histologic grade, which is

the most important indicator of disease-free

survival (Korkolopoulou et al., 1998).

Malignant and nonmalignant skin diseases

Note

In one study of comparison between malignant skin

diseases (squamous cell carcinoma, adult T

lymphotrophic leukemia, mycosis fungoides,

malignant melanoma and malignant lymphoma)

and nonmalignant skin diseases (resistant atopic

dermatitis, psoriasis vulgaris, verruca vulgaris) the

anti-PCNA staining was used as a prognostic

marker (Kawahira, 1999). The percentage of

PCNA-positive cells reported in the study was

higher for malignant skin diseases in comparison

with the non-malignant skin deseases (Kawahira,

1999). The localization of PCNA-positive cells was

found to be in the dermis and the basal layer in case

of the malignant skin diseases, whereas in the

nonmalignant skin diseases PCNA-positive cells

were detected only in the basal layer (Kawahira,

1999). The PCNA labeling index and the

distribution of PCNA-positive cells in the skin were

suggested to be helpful in the early diagnosis of

skin malignancies.

Systemic lupus erythematosus (SLE)

Note

The anti-PCNA antibodies were originally found in

patients with systemic lupus erythematosus

(Miyachi et al., 1978), most of whom had diffuse

proliferative glomerulonephritis in a small clinical

study (Fritzler et al., 1983).

Page 33: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

PCNA (proliferating cell nuclear antigen) Stoimenov I, Helleday T

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 209

References Miyachi K, Fritzler MJ, Tan EM. Autoantibody to a nuclear antigen in proliferating cells. J Immunol. 1978 Dec;121(6):2228-34

Fritzler MJ, McCarty GA, Ryan JP, Kinsella TD. Clinical features of patients with antibodies directed against proliferating cell nuclear antigen. Arthritis Rheum. 1983 Feb;26(2):140-5

Tan CK, Castillo C, So AG, Downey KM. An auxiliary protein for DNA polymerase-delta from fetal calf thymus. J Biol Chem. 1986 Sep 15;261(26):12310-6

Prelich G, Tan CK, Kostura M, Mathews MB, So AG, Downey KM, Stillman B. Functional identity of proliferating cell nuclear antigen and a DNA polymerase-delta auxiliary protein. Nature. 1987 Apr 2-8;326(6112):517-20

Ku DH, Travali S, Calabretta B, Huebner K, Baserga R. Human gene for proliferating cell nuclear antigen has pseudogenes and localizes to chromosome 20. Somat Cell Mol Genet. 1989 Jul;15(4):297-307

Travali S, Ku DH, Rizzo MG, Ottavio L, Baserga R, Calabretta B. Structure of the human gene for the proliferating cell nuclear antigen. J Biol Chem. 1989 May 5;264(13):7466-72

Webb G, Parsons P, Chenevix-Trench G. Localization of the gene for human proliferating nuclear antigen/cyclin by in situ hybridization. Hum Genet. 1990 Nov;86(1):84-6

del Giglio A, O'Brien S, Ford R, Saya H, Manning J, Keating M, Johnston D, Khetan R, el-Naggar A, Deisseroth A. Prognostic value of proliferating cell nuclear antigen expression in chronic lymphoid leukemia. Blood. 1992 May 15;79(10):2717-20

Taniguchi Y, Katsumata Y, Koido S, Suemizu H, Yoshimura S, Moriuchi T, Okumura K, Kagotani K, Taguchi H, Imanishi T, Gojobori T, Inoko H. Cloning, sequencing, and chromosomal localization of two tandemly arranged human pseudogenes for the proliferating cell nuclear antigen (PCNA). Mamm Genome. 1996 Dec;7(12):906-8

Chu JS, Huang CS, Chang KJ. Proliferating cell nuclear antigen (PCNA) immunolabeling as a prognostic factor in invasive ductal carcinoma of the breast in Taiwan. Cancer Lett. 1998 Sep 25;131(2):145-52

Horiguchi J, Iino Y, Takei H, Maemura M, Takeyoshi I, Yokoe T, Ohwada S, Oyama T, Nakajima T, Morishita Y. Long-term prognostic value of PCNA labeling index in primary operable breast cancer. Oncol Rep. 1998 May-Jun;5(3):641-4

Korkolopoulou P, Angelopoulou MK, Kontopidou F, Tsengas A, Patsouris E, Kittas C, Pangalis GA. Prognostic implications of proliferating cell nuclear antigen (PCNA), AgNORs and P53 in non-Hodgkin's lymphomas. Leuk Lymphoma. 1998 Aug;30(5-6):625-36

Kawahira K. Immunohistochemical staining of proliferating cell nuclear antigen (PCNA) in malignant and nonmalignant skin diseases. Arch Dermatol Res. 1999 Jul-Aug;291(7-8):413-8

Ma X, Jin Q, Försti A, Hemminki K, Kumar R. Single nucleotide polymorphism analyses of the human proliferating cell nuclear antigen (pCNA) and flap endonuclease (FEN1) genes. Int J Cancer. 2000 Dec 15;88(6):938-42

Faderl S, Keating MJ, Do KA, Liang SY, Kantarjian HM, O'Brien S, Garcia-Manero G, Manshouri T, Albitar M. Expression profile of 11 proteins and their prognostic significance in patients with chronic lymphocytic leukemia (CLL). Leukemia. 2002 Jun;16(6):1045-52

Naryzhny SN, Zhao H, Lee H. Proliferating cell nuclear antigen (PCNA) may function as a double homotrimer complex in the mammalian cell. J Biol Chem. 2005 Apr 8;280(14):13888-94

Moldovan GL, Pfander B, Jentsch S. PCNA controls establishment of sister chromatid cohesion during S phase. Mol Cell. 2006 Sep 1;23(5):723-32

Moldovan GL, Pfander B, Jentsch S. PCNA, the maestro of the replication fork. Cell. 2007 May 18;129(4):665-79

Naryzhny SN. Proliferating cell nuclear antigen: a proteomics view. Cell Mol Life Sci. 2008 Nov;65(23):3789-808

Peled JU, Kuang FL, Iglesias-Ussel MD, Roa S, Kalis SL, Goodman MF, Scharff MD. The biochemistry of somatic hypermutation. Annu Rev Immunol. 2008;26:481-511

Roa S, Avdievich E, Peled JU, Maccarthy T, Werling U, Kuang FL, Kan R, Zhao C, Bergman A, Cohen PE, Edelmann W, Scharff MD. Ubiquitylated PCNA plays a role in somatic hypermutation and class-switch recombination and is required for meiotic progression. Proc Natl Acad Sci U S A. 2008 Oct 21;105(42):16248-53

Stoimenov I, Helleday T. PCNA on the crossroad of cancer. Biochem Soc Trans. 2009 Jun;37(Pt 3):605-13

This article should be referenced as such:

Stoimenov I, Helleday T. PCNA (proliferating cell nuclear antigen). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):206-209.

Page 34: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 210

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

RASSF5 (Ras association (RalGDS/AF-6) domain family member 5) Lee Schmidt, Geoffrey J Clark

University of Louisville, Room 119C, Baxter II Research Building, 580 S Preston Street, Louisville,

KY 40202, USA (LS, GJC)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/RASSF5ID42059ch1q32.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI RASSF5ID42059ch1q32.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: MGC10823, MGC17344, Maxp1,

NORE1, NORE1A, NORE1B, RAPL, RASSF3

HGNC (Hugo): RASSF5

Location: 1q32.1

Note

Murine RASSF5 originally named Nore1a. Nore1B

independently identified and designated RAPL. Rat

RASSF5 also cloned independently and designated

Maxp1.

DNA/RNA

Description

The human gene for RASSF5 is 81 kb in length and

is located on chromosome 1(q32.1). The gene can

produce 4 protein isoforms, two via differential

exon usage, a third via differential promoter usage

and the genesis of the 4th

(which can be found as an

EST clone) is not yet clear. The largest isoform, A,

is 418 amino acids long and has a molecular weight

of about 47 kD. The protein structure of RASSF5A

contains a proline-rich region at the N-terminus

which contains potential SH3 binding sites and a

nuclear localization signal. This is followed by a

cystein rich domain, sometimes referred to as a zinc

finger. Next is the Ras association domain and this

is followed by sequence containing the SARAH

motif required for binding to the pro-apoptotic

kinases MST1 and MST2. A second nuclear

localization sequence has been reported between

amino acids 200-260 and a nuclear export sequence

between amino acids 260-300.

Figure 1. Isoform A is shown as the longest isoform with 6 exons. Isoform B, without an alternate exon, shows that the frameshift gives a shortened and unique C-terminus. Isoform C is shown with a special 5' UTR and lacks an in-frame coding region leading

to a unique N-terminus. The total coding sequence for Isoform A is about 1260 bases with the other isoforms being smaller.

Page 35: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

RASSF5 (Ras association (RalGDS/AF-6) domain family member 5)

Schmidt L, Clark GJ

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 211

Figure 2. A figure showing the processed mRNA as well as the amino acid sequence for isoforms A-D followed by motif

explanation of isoform A (Nore1a).

Page 36: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

RASSF5 (Ras association (RalGDS/AF-6) domain family member 5)

Schmidt L, Clark GJ

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 212

Protein

Description

The full length cDNA (for isoform A) encodes for a

47-kDa protein which contains a proline-rich region

at the N-terminus followed by a putative

diacylglycerol/phorbol ester binding domain. This

is followed by the Ras association (RA) domain and

then the domain containing a SARAH motif. This

later is responsible for binding to the pro-apoptotic

kinases MST1 and MST2.

Expression

Nore1a mRNA is expressed in the lung, kidney,

liver, brain, spleen, thymus and heart.

Localisation

It can be detected on microtubules, in the

centrosome, but appears most obvious in the

nucleus.

Function

RASSF5A is a pro-apoptotic Ras effector that can

bind and relocalize the pro-apoptotic MST kinases

in the presence of activated Ras. It can also promote

cell cycle arrest and modulate the activity of p53 by

regulating its' nuclear localization. Knockdown of

RASSF5A promotes cellular proliferation and soft

agar growth. Thus, RASSF5A appears to function

as a Ras regulated tumor suppressor. Analysis of

human tumors has found little evidence of somatic

mutation but the gene is frequently inactivated by

promoter methylation in a broad range of human

tumors. RASSF5C (also known as Nore1b or

RAPL) has been reported to modulate cellular

adhesion and to be regulated by the Ras related

protein Rap1a. RASSF5C has also been implicated

as serving as an adaptor protein to facilitate the

interaction of Ras and CARMA1.

Mutations

Note

No tumor mutations yet reported.

Implicated in

Clear cell renal carcinoma

Note

RASSF5 is frequently down-regulated by promoter

methylation in a variety of tumors including clear

cell renal carcinomas. Moreover, a rare hereditary

form of kidney cancer has been reported that maps

with a translocation inactivating the RASSF5 gene.

Various cancers

Note

Nore1a is frequently inactivated by promoter

methylation in renal carcinoma, breast cancer, lung

cancer, liver cancer and neurological tumors.

References Vavvas D, Li X, Avruch J, Zhang XF. Identification of Nore1 as a potential Ras effector. J Biol Chem. 1998 Mar 6;273(10):5439-42

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

Chen J, Lui WO, Vos MD, Clark GJ, Takahashi M, Schoumans J, Khoo SK, Petillo D, Lavery T, Sugimura J, Astuti D, Zhang C, Kagawa S, Maher ER, Larsson C, Alberts AS, Kanayama HO, Teh BT. The t(1;3) breakpoint-spanning genes LSAMP and NORE1 are involved in clear cell renal cell carcinomas. Cancer Cell. 2003 Nov;4(5):405-13

Hesson L, Dallol A, Minna JD, Maher ER, Latif F. NORE1A, a homologue of RASSF1A tumour suppressor gene is inactivated in human cancers. Oncogene. 2003 Feb 13;22(6):947-54

Vos MD, Martinez A, Ellis CA, Vallecorsa T, Clark GJ. The pro-apoptotic Ras effector Nore1 may serve as a Ras-regulated tumor suppressor in the lung. J Biol Chem. 2003 Jun 13;278(24):21938-43

Aoyama Y, Avruch J, Zhang XF. Nore1 inhibits tumor cell growth independent of Ras or the MST1/2 kinases. Oncogene. 2004 Apr 22;23(19):3426-33

Praskova M, Khoklatchev A, Ortiz-Vega S, Avruch J. Regulation of the MST1 kinase by autophosphorylation, by the growth inhibitory proteins, RASSF1 and NORE1, and by Ras. Biochem J. 2004 Jul 15;381(Pt 2):453-62

Avruch J, Praskova M, Ortiz-Vega S, Liu M, Zhang XF. Nore1 and RASSF1 regulation of cell proliferation and of the MST1/2 kinases. Methods Enzymol. 2006;407:290-310

Calvisi DF, Ladu S, Gorden A, Farina M, Conner EA, Lee JS, Factor VM, Thorgeirsson SS. Ubiquitous activation of Ras and Jak/Stat pathways in human HCC. Gastroenterology. 2006 Apr;130(4):1117-28

Donninger H, Vos MD, Clark GJ. The RASSF1A tumor suppressor. J Cell Sci. 2007 Sep 15;120(Pt 18):3163-72

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

Calvisi DF, Donninger H, Vos MD, Birrer MJ, Gordon L, Leaner V, Clark GJ. NORE1A tumor suppressor candidate modulates p21CIP1 via p53. Cancer Res. 2009 Jun 1;69(11):4629-37

Richter AM, Pfeifer GP, Dammann RH. The RASSF proteins in cancer; from epigenetic silencing to functional characterization. Biochim Biophys Acta. 2009 Dec;1796(2):114-28

Bee C, Moshnikova A, Mellor CD, Molloy JE, Koryakina Y, Stieglitz B, Khokhlatchev A, Herrmann C. Growth and tumor suppressor NORE1A is a regulatory node between Ras signaling and microtubule nucleation. J Biol Chem. 2010 May 21;285(21):16258-66

Kumari G, Singhal PK, Suryaraja R, Mahalingam S. Functional interaction of the Ras effector RASSF5 with the tyrosine kinase Lck: critical role in nucleocytoplasmic transport and cell cycle regulation. J Mol Biol. 2010 Mar 19;397(1):89-109

Park J, Kang SI, Lee SY, Zhang XF, Kim MS, Beers LF, Lim DS, Avruch J, Kim HS, Lee SB. Tumor suppressor ras

Page 37: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

RASSF5 (Ras association (RalGDS/AF-6) domain family member 5)

Schmidt L, Clark GJ

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 213

association domain family 5 (RASSF5/NORE1) mediates death receptor ligand-induced apoptosis. J Biol Chem. 2010 Nov 5;285(45):35029-38

Overmeyer JH, Maltese WA. Death pathways triggered by activated Ras in cancer cells. Front Biosci. 2011 Jan 1;16:1693-713

This article should be referenced as such:

Schmidt L, Clark GJ. RASSF5 (Ras association (RalGDS/AF-6) domain family member 5). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):210-213.

Page 38: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 214

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

RGS17 (regulator of G-protein signaling 17) Chenguang Li, Lei Wang, Yihua Sun, Haiquan Chen

Department of Thoracic Oncology, Fudan University Shanghai Cancer Center and Department of

Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China (CL, LW, YS, HC)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/RGS17ID47522ch6q25.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI RGS17ID47522ch6q25.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: RGS-17, RGSZ2, hRGS17

HGNC (Hugo): RGS17

Location: 6q25.2

DNA/RNA

Description

The RGS17 gene spans over a region of 120 kbp

DNA including 4 coding exons and 1 non-coding

exon (exon 1).

Transcription

The RGS17 gene mRNA consists of about 1472

nucleotides with an open reading frame (ORF) of

633 bases.

Pseudogene

RGS17P1 regulator of G-protein signaling 17

pseudogene 1.

Protein

Note

210 amino acids; 24 kDa.

Description

The RGS17 protein consists of 210 amino acid

residues. This gene encodes a member of the

regulator of G-protein signaling family. This

protein contains a conserved, 120 amino acid motif

called the RGS domain and a cysteine-rich region.

Expression

Widely expressed in human organs.

Localisation

Its cellular localization has not been formally

monitored to date.

Function

The protein attenuates the signaling activity of G-

proteins by binding to activated, GTP-bound G

alpha subunits and acting as a GTPase activating

protein (GAP), increasing the rate of conversion of

the GTP to GDP. RGS proteins are GTPase-

activating proteins for Gi and Gq class G-alpha

proteins. They accelerate transit through the cycle

of GTP binding and hydrolysis and thereby

accelerate signaling kinetics and termination. This

hydrolysis allows the G alpha subunits to bind G

beta/gamma subunit heterodimers, forming inactive

G-protein heterotrimers, thereby terminating the

signal.

Diagram of the RGS17 protein in scale. The numbers represent specific residues. The regions are RGS_RZ-like (Regulator of G

protein signaling (RGS) domain found in the RZ protein), putative G-alpha interaction site. C: Carboxyl-terminal; N: Amino-terminal.

Page 39: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

RGS17 (regulator of G-protein signaling 17) Li C, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 215

Homology

The RGS17 gene is conserved in chimpanzee, dog,

cow, mouse, rat, chicken, and zebrafish.

Mutations

Germinal

No germline mutations in this gene have been

reported.

Somatic

A synonymous-coding somatic mutations of this

gene is reported in pancreas cancer at codon 166,

P166P (COSMIC).

Implicated in

Various cancer

Note

Lung cancer, prostate cancer.

Disease

RSG17 is overexpressed in lung and prostate cancer

(James et al., 2009). Expression of RGS17 is up-

regulated in 80% of lung tumors, and also up-

regulated in prostate tumors. Overexpression of

RGS17 induce and maintain cell proliferation.

Lung cancer

Disease

hsa-mir-182 is involved in the down regulation of

RGS17 expression through two conserved sites

located in its 3' UTR region (Sun et al., 2010).

Two SNPs in the first intron of RGS17 (rs4083914

and rs9479510) were found associated with familial

lung cancer susceptibility (You et al., 2009).

Ovarian cancer

Disease

RGS2, RGS5, RGS10 and RGS17 transcripts are

expressed at significantly lower levels in cells

resistant to chemotherapy compared with parental,

chemo-sensitive cells in ovarian cancer cells

(Hooks et al., 2010).

Prognosis

RGS17 loss of expression contributes to the

development of chemoresistance in ovarian cancer

cells.

References James MA, Lu Y, Liu Y, Vikis HG, You M. RGS17, an overexpressed gene in human lung and prostate cancer, induces tumor cell proliferation through the cyclic AMP-PKA-CREB pathway. Cancer Res. 2009 Mar 1;69(5):2108-16

You M, Wang D, Liu P, Vikis H, James M, Lu Y, Wang Y, Wang M, Chen Q, Jia D, Liu Y, Wen W, Yang P, et al. Fine mapping of chromosome 6q23-25 region in familial lung cancer families reveals RGS17 as a likely candidate gene. Clin Cancer Res. 2009 Apr 15;15(8):2666-74

Hooks SB, Callihan P, Altman MK, Hurst JH, Ali MW, Murph MM. Regulators of G-Protein signaling RGS10 and RGS17 regulate chemoresistance in ovarian cancer cells. Mol Cancer. 2010 Nov 2;9:289

Sun Y, Fang R, Li C, Li L, Li F, Ye X, Chen H. Hsa-mir-182 suppresses lung tumorigenesis through down regulation of RGS17 expression in vitro. Biochem Biophys Res Commun. 2010 May 28;396(2):501-7

This article should be referenced as such:

Li C, Wang L, Sun Y, Chen H. RGS17 (regulator of G-protein signaling 17). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):214-215.

Page 40: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 216

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

SLC39A1 (solute carrier family 39 (zinc transporter), member 1) Renty B Franklin, Leslie C Costello

Department of Oncology and Diagnostic Sciences, Dental School and The Greenebaum Cancer

Center, University of Maryland, Baltimore, MD, 21201, USA (RBF, LCC)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/SLC39A1ID46571ch1q21.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI SLC39A1ID46571ch1q21.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: ZIP1, ZIRTL

HGNC (Hugo): SLC39A1

Location: 1q21.3

DNA/RNA

Description

The SLC39A1 gene contains 5 exons, three of

which are coding. The length of the gene is 8600

base pairs according to the Entrez Gene database.

Several transcripts have been reported containing

either 3, 4, or 5 exons. However, the coding

sequence for SLC39A1 is the same for all reported

transcripts.

Transcription

Only a single isoform has been reported; mRNA

2445 bases in length; coding region of 975 bases.

Pseudogene

None reported.

Protein

Description

SLC39A1 encodes Zrt/Irt-like protein family

member 1 (ZIP1).

ZIP1 is a 35 kDa molecular weight protein

consisting of 324 amino acids. The protein contains

8 transmembrane spanning domains and shows the

characteristics of a zinc transporter (Gaither and

Eide, 2001).

Expression

ZIP1 is ubiquitously expressed in mammalian cells

(Gaither and Eide, 2001). Expression is down

regulated in prostate malignancy (Franklin et al.,

2005). Its constitutive expression is reported to be

regulated by SP and CREB1 (Makhov et al., 2009).

Down regulation of expression in prostate

malignancy is reported due to transcription

repression by ras response element binding protein-

1 (RREB-1) (Milon et al., 2010).

Localisation

ZIP1 is located at the cell membrane.

Function

ZIP1 is a facilitated zinc uptake transporter

(Franklin et al., 2003). Activity of the transporter

results in the intracellular accumulation of zinc.

ZIP1 is involved in apoptosis induction in prostate

cancer cells.

Homology

Mus musculus Slc39a1; Rattus norvegicus Slc39a1;

Bos taurus Slc39a1; Danio rerio slc39a1.

Mutations

Note

No diseases related to mutation are reported.

Implicated in

Prostate cancer

Note

SLC39A1 is down regulated in prostate cancer

(Franklin et al., 2005). Knockdown of the

Page 41: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

SLC39A1 (solute carrier family 39 (zinc transporter), member 1)

Franklin RB, Costello LC

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 217

SLC39A1 expression decreases cellular zinc and

increases growth of PC-3 cells (Franklin et al.,

2003). Over expression of SLC39A1 inhibits

prostate tumor growth in a xenograft model

(Golovine et al., 2008).

References Gaither LA, Eide DJ. The human ZIP1 transporter mediates zinc uptake in human K562 erythroleukemia cells. J Biol Chem. 2001 Jun 22;276(25):22258-64

Franklin RB, Ma J, Zou J, Guan Z, Kukoyi BI, Feng P, Costello LC. Human ZIP1 is a major zinc uptake transporter for the accumulation of zinc in prostate cells. J Inorg Biochem. 2003 Aug 1;96(2-3):435-42

Franklin RB, Feng P, Milon B, Desouki MM, Singh KK, Kajdacsy-Balla A, Bagasra O, Costello LC. hZIP1 zinc uptake transporter down regulation and zinc depletion in prostate cancer. Mol Cancer. 2005 Sep 9;4:32

Golovine K, Makhov P, Uzzo RG, Shaw T, Kunkle D, Kolenko VM. Overexpression of the zinc uptake transporter hZIP1 inhibits nuclear factor-kappaB and reduces the malignant potential of prostate cancer cells in vitro and in vivo. Clin Cancer Res. 2008 Sep 1;14(17):5376-84

Makhov P, Golovine K, Uzzo RG, Wuestefeld T, Scoll BJ, Kolenko VM. Transcriptional regulation of the major zinc uptake protein hZip1 in prostate cancer cells. Gene. 2009 Feb 15;431(1-2):39-46

Milon BC, Agyapong A, Bautista R, Costello LC, Franklin RB. Ras responsive element binding protein-1 (RREB-1) down-regulates hZIP1 expression in prostate cancer cells. Prostate. 2010 Feb 15;70(3):288-96

This article should be referenced as such:

Franklin RB, Costello LC. SLC39A1 (solute carrier family 39 (zinc transporter), member 1). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):216-217.

Page 42: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 218

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

CBX7 (chromobox homolog 7) Ana O'Loghlen, Jesus Gil

Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith

Campus, London W12 0NN, UK (AO, JG)

Published in Atlas Database: November 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/CBX7ID43845ch22q13.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI CBX7ID43845ch22q13.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

HGNC (Hugo): CBX7

Location: 22q13.1

Note

Orientation: minus strand. Size: 32508 bases.

DNA/RNA

Description

DNA size is 4081 bp with 6 exons. CBX7 is a

highly conserved gene in chimpanzee, dog, cow, rat

and mouse.

Transcription

mRNA size: 3964 bp.

Protein

Note

251 amino acids. Isoelectric point: 10,0228.

Molecular weight of the protein: 28209 Da.

Description

CBX7 has a chromodomain region which is

commonly found in proteins associated with the

remodelling and manipulation of chromatin. In

mammals, chromodomain-containing proteins are

responsible for aspects of gene regulation related to

chromatin remodelling and formation of

heterochromatin regions. Chromodomain-

containing proteins also bind methylated histones

and appear in the RNA-induced transcriptional

silencing complex. Specifically, CBX7 is involved

in maintaining the transcriptionally repressive state

of its target genes. The better characterized target of

CBX7 is the INK4a/ARF locus, which is repressed

by CBX7 in order to overcome the senescent

phenotype in several mouse and human cell lines.

Repression of other targets like E-cadherin has been

also suggested.

Figure 1. Location of Cbx7 within Chromosome 22.

Figure 2. Diagram of Cbx7 transcript. Cbx7 has 6 exons. The black boxes indicate the consensus coding sequences (CCDS).

Page 43: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

CBX7 (chromobox homolog 7) O'Loghlen A, Gil J

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 219

Figure 3. Structure of Cbx7 protein. Cbx7 has a chromodomain motif and a Polycomb (Pc) box which are indicated in grey.

Expression

CBX7 is expressed ubiquitously, but at higher

levels in the nervous system, thyroid gland,

prostate, fallopian tubes and bladder in normal

tissue. CBX7 expression is also high in ES cells.

Localisation

In the nucleus.

Function

CBX7 is a member of the Polycomb group (PcG)

genes, which are transcriptional repressors that play

an essential role in development, cancer progression

and stem cell maintenance. Mainly two different

PcG complexes have been described: Polycomb

Repressive Complex 1 (PRC1) and PRC2. PRC2 is

the complex implicated in initiating the silencing of

its target genes by methylating histone H3 on

lysines 9 and 27. PRC1 is implicated in stabilizing

this repressive state by recognizing the methylation

marks through the Polycomb proteins and by

ubiquitinating the histone H2A on Lys119. CBX7

belongs to the PRC1 complex and has been

described to be a regulator of cellular lifespan by

repressing the INK4a/ARF locus in several mouse

and human cell lines. On the other hand, depletion

of CBX7 from the cell induces a senescent

phenotype by increasing the expression of the cell

cycle regulators p16/ARF.

X chromosome inactivation CBX7 has high affinity for binding H3K9me3 and

H3K27me3. It associates with heterochromatin,

binds RNA and it's enriched in the X chromosome,

giving CBX7 a role in maintaining the repression of

genes in the X chromosome.

Epigenetic regulation CBX7, as part of the PRC1 complex, has a role in

maintaining the repressive state of its target genes.

CBX7 binds to the long non-coding RNA ANRIL

in order to represses the INK4a/ARF locus and this

interaction is essential for CBX7's function. Both

CBX7 and ANRIL have been found to have high

levels in prostate cancer tissues.

Stem cells self-renewal CBX7 has been recently implicated to be essential

for maintaining the pluripotency state of stem cells

(ES cells). Overexpression of CBX7 in ESC

impairs cell differentation. On the other hand,

depletion of CBX7 from ESC induces spontaneous

differentiation. Two different miR families (miR-

125 and miR-181) were identified in a screening for

CBX7 regulators and have been described to have a

role in ESC differentiation by targeting the 3'UTR

of CBX7.

Figure 4. 4a: Summary of Cbx7's mechanism in

embryonic stem cells (ESC). Cbx7 is essential for ESC self-renewal. Loss of Cbx7, either by differentiating ESC or by an exogenous/endogenous induction of the microRNA

(miR) families miR-125 and miR-181, induces ESC differentiation. This is accompanied by an increase in other

Cbxs as they are targets of Cbx7. On the other hand, overexpression of Cbx7 in ESC reinforces pluripotency and

keeps the cells in an ESC-like state when forced to differentiate. 4b and 4c: Summary of Cbx7's mechanism

in human primary fibroblasts (IMR-90). Ectopic expression of the miR families miR-125 and miR-181

induces a degradation of Cbx7 mRNA in IMR-90. Depletion of Cbx7 induces the cells to senesce. Thus, overexpression

of miR-125 and miR-181 induces senescence through downregulation of Cbx7.

Mutations

Note

Expression of CBX7 without the Pc box or with

point mutations in the chromodomain region

(F11A, K31A, W32A, W35A) does not extend the

life span of human or mouse cells. The mutant

R17Q, which affects the binding of CBX7 to RNA,

Page 44: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

CBX7 (chromobox homolog 7) O'Loghlen A, Gil J

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 220

extended the lifespan of cells, but to a lesser extent

than CBX7 wt. Point mutations in the Pc box as

F234D or F244D result in loss or reduced

interaction of CBX7 with RNF2.

Implicated in

Various cancers

Disease

CBX7 has been implicated in several tumors such

as gastric cancer, follicular lymphoma, breast

cancer, colon carcinoma, pancreatic cancer, tyroid

cancer, glioma.

Prognosis

There is a controversy in the role of CBX7 in

cancer, as some papers associate CBX7

overexpression with poor prognosis and advanced

estate of the tumor and aggressiveness, while others

state that depletion of CBX7 from certain cancers

indicates the state of malignancy of the tumor. The

ability of CBX7 to regulate multiple targets and the

relevance of those targets in different tumor types

and stages probably explain those paradoxical

findings.

References Gil J, Bernard D, Martínez D, Beach D. Polycomb CBX7 has a unifying role in cellular lifespan. Nat Cell Biol. 2004 Jan;6(1):67-72

Bernard D, Martinez-Leal JF, Rizzo S, Martinez D, Hudson D, Visakorpi T, Peters G, Carnero A, Beach D, Gil J. CBX7 controls the growth of normal and tumor-derived prostate cells by repressing the Ink4a/Arf locus. Oncogene. 2005 Aug 25;24(36):5543-51

Gil J, Bernard D, Peters G. Role of polycomb group proteins in stem cell self-renewal and cancer. DNA Cell Biol. 2005 Feb;24(2):117-25

Bernstein E, Duncan EM, Masui O, Gil J, Heard E, Allis CD. Mouse polycomb proteins bind differentially to methylated histone H3 and RNA and are enriched in facultative heterochromatin. Mol Cell Biol. 2006 Apr;26(7):2560-9

Scott CL, Gil J, Hernando E, Teruya-Feldstein J, Narita M, Martínez D, Visakorpi T, Mu D, Cordon-Cardo C, Peters G, Beach D, Lowe SW. Role of the chromobox protein CBX7 in lymphomagenesis. Proc Natl Acad Sci U S A. 2007 Mar 27;104(13):5389-94

Pallante P, Federico A, Berlingieri MT, Bianco M, Ferraro A, Forzati F, Iaccarino A, Russo M, Pierantoni GM, Leone V, Sacchetti S, Troncone G, Santoro M, Fusco A. Loss of the CBX7 gene expression correlates with a highly malignant phenotype in thyroid cancer. Cancer Res. 2008 Aug 15;68(16):6770-8

Yap KL, Li S, Muñoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, Gil J, Walsh MJ, Zhou MM. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell. 2010 Jun 11;38(5):662-74

Morey L, Pascual G, Cozzuto L, Roma G, Wutz A, Benitah SA, Di Croce L. Nonoverlapping functions of the polycomb group cbx family of proteins in embryonic stem cells. Cell Stem Cell. 2012 Jan 6;10(1):47-62

O'Loghlen A, Muñoz-Cabello AM, Gaspar-Maia A, Wu HA, Banito A, Kunowska N, Racek T, Pemberton HN, Beolchi P, Lavial F, Masui O, Vermeulen M, Carroll T, Graumann J, Heard E, Dillon N, Azuara V, Snijders AP, Peters G, Bernstein E, Gil J. MicroRNA Regulation of Cbx7 Mediates a Switch of Polycomb Orthologs during ESC Differentiation. Cell Stem Cell. 2012 Jan 6;10(1):33-46

This article should be referenced as such:

O'Loghlen A, Gil J. CBX7 (chromobox homolog 7). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):218-220.

Page 45: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 221

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

RPRM (reprimo, TP53 dependent G2 arrest mediator candidate) Alejandro H Corvalan, Veronica A Torres

Laboratory of Molecular Pathology and Epidemiology, Department of Hemathology - Oncology,

School of Medicine - P Universidad Catolica de Chile, 391 Marcoleta St - Santiago 8330074 Chile

(AHC, TorresVAT)

Published in Atlas Database: November 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/RPRMID42082ch2q23.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI RPRMID42082ch2q23.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: FLJ90327, REPRIMO

HGNC (Hugo): RPRM

Location: 2q23.3

DNA/RNA

Description

Reprimo gene consists of 1 exon. The gene spans

1,47 kb of genomic DNA on the chromosome 2 in

the minus strand.

Transcription

The mRNA is 1496 bp in length.

Protein

Description

The open reading frame encodes a 109 amino acid

protein with an estimated molecular weight of

11774 Da. Reprimo is a highly glycosylated protein

which has two sites in amino acids 7 and 18. The

protein has a potential transmembrane site covering

amino acids 56 to 76.

Expression

The expression of Reprimo is induced by tumor

protein p53 following X-ray irradiation.

Localisation

When Reprimo is ectopically expressed, it is

localized in the cytoplasm.

Function

Reprimo is a candidate tumor suppresor gene

involved in the G2/M phase cell cycle arrest

mediated by tumor protein p53. Reprimo induces

cell cycle arrest by inhibiting the nuclear

translocation of the Cdc2-Cyclin B1 complex.

Implicated in

Various cancers

Note

The aberrant methylation of the promoter region of

Reprimo is a common event that may contribute to

the pathogenesis of some types of human cancer.

Promoter methylation of Reprimo was found in

pancreatic cancer (91%), gastric cancer (90%),

gallbladder cancer (62%), lymphomas (57%),

colorectal cancer (56%) and esophageal

adenocarcinomas (40%). In breast cancer,

leukemias and lung cancer, promoter methylation of

Reprimo was found in less than 40% of tested

cases.

Gastric cancer

Disease

Aberrant hypermethylation of Reprimo is

frequently found in primary gastric cancer as well

as in pair plasma samples. In plasma from

asymptomatic controls, Reprimo is infrequently

methylated. Therefore, plasmatic detection of

Reprimo is a putative biomarker for early detection

of gastric cancer.

Page 46: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

RPRM (reprimo, TP53 dependent G2 arrest mediator candidate)

Corvalan AH, TorresVA

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 222

The above histogram represents the percentage of positive cases for Reprimo and other genes (APC, SHP1, CDH-1, ER, SEMA3B and 3OST2) in 43 prospectively collected gastric cancer cases and 31 asymptomatic age- and gender-matched

controls. Only Reprimo shows a significant difference in plasma between gastric cancer and asymptomatic controls (Bernal et al., Clin Cancer Res. 2008;14:6264-9).

Pancreatic cancer

Disease

Aberrant hypermethylation of Reprimo is also

common in pancreatic cell lines (91%) and in

pancreatic adenocarcinomas (66%). Reprimo

methylation is correlated with poor prognosis in a

large series of resected pancreatic cancers. This fact

raises the possibility that aberrant methylation of

Reprimo is an epigenetic event that may have a

mechanistic role in pancreatic cancer.

References Ohki R, Nemoto J, Murasawa H, Oda E, Inazawa J, Tanaka N, Taniguchi T. Reprimo, a new candidate mediator of the p53-mediated cell cycle arrest at the G2 phase. J Biol Chem. 2000 Jul 28;275(30):22627-30

Sato N, Fukushima N, Maitra A, Matsubayashi H, Yeo CJ, Cameron JL, Hruban RH, Goggins M. Discovery of novel targets for aberrant methylation in pancreatic carcinoma using high-throughput microarrays. Cancer Res. 2003 Jul 1;63(13):3735-42

Suzuki M, Shigematsu H, Takahashi T, Shivapurkar N, Sathyanarayana UG, Iizasa T, Fujisawa T, Gazdar AF. Aberrant methylation of Reprimo in lung cancer. Lung Cancer. 2005 Mar;47(3):309-14

Takahashi T, Suzuki M, Shigematsu H, Shivapurkar N, Echebiri C, Nomura M, Stastny V, Augustus M, Wu CW, Wistuba II, Meltzer SJ, Gazdar AF. Aberrant methylation of Reprimo in human malignancies. Int J Cancer. 2005 Jul 1;115(4):503-10

Hamilton JP, Sato F, Jin Z, Greenwald BD, Ito T, Mori Y, Paun BC, Kan T, Cheng Y, Wang S, Yang J, Abraham JM, Meltzer SJ. Reprimo methylation is a potential biomarker of Barrett's-Associated esophageal neoplastic progression. Clin Cancer Res. 2006 Nov 15;12(22):6637-42

Sato N, Fukushima N, Matsubayashi H, Iacobuzio-Donahue CA, Yeo CJ, Goggins M. Aberrant methylation of Reprimo correlates with genetic instability and predicts poor prognosis in pancreatic ductal adenocarcinoma. Cancer. 2006 Jul 15;107(2):251-7

Bernal C, Aguayo F, Villarroel C, Vargas M, Díaz I, Ossandon FJ, Santibáñez E, Palma M, Aravena E, Barrientos C, Corvalan AH. Reprimo as a potential biomarker for early detection in gastric cancer. Clin Cancer Res. 2008 Oct 1;14(19):6264-9

This article should be referenced as such:

Corvalan AH, TorresVA. RPRM (reprimo, TP53 dependent G2 arrest mediator candidate). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):221-222.

Page 47: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Mini Review

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 223

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

VMP1 (vacuole membrane protein 1) Alejandro Ropolo, Andrea Lo Ré, María Inés Vaccaro

Molecular Pathophysiology Lab, School of Pharmacie and Biochemistry, University of Buenos Aires,

Argentina (AR, AL, MIV)

Published in Atlas Database: November 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/VMP1ID50079ch17q23.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI VMP1ID50079ch17q23.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: DKFZp566I133, EPG3, TMEM49

HGNC (Hugo): VMP1

Location: 17q23.1

DNA/RNA

Description

12 exons, spans approximately 133 kb of genomic

DNA in the centromere-to-telomere orientation.

The translation initiation codon is located to exon 2,

and the stop codon to exon 12.

Transcription

mRNA of 2,17 kb.

Protein

Description

The pancreatitis-associated protein vacuole

membrane protein 1 (VMP1) is a transmembrane

protein of 406 amino-acid length containing 6

putative transmembrane domains and with no

known homologues in yeast.

Expression

VMP1 was characterized because is not

constitutively expressed in pancreatic acinar cells

and it is highly activated early during experimental

acute pancreatitis in acinar cells.

Localisation

Autophagosomal membrane.

Function

VMP1 is an autophagy-related membrane protein.

VMP1 expression triggers autophagy, even under

nutrient-replete conditions. VMP1 is required for

autophagosome development through interaction

with Beclin1. Recently, it has been demonstrated

that participate in a novel selective form of

autophagy, called zymophagy, mediated by VMP1-

USP9x-p62 pathway during acute pancreatitis.

Genomic organization of the VMP1/TMEM49 gene.

Page 48: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

VMP1 (vacuole membrane protein 1) Ropolo A, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 224

Schematic representation of VMP1 protein and localization of transmembrane domains.

Implicated in

Pancreatic cancer

Disease

Pancreatic ductal adenocarcinoma is one of the

most aggressive human malignancies with a 2-3%

5-year survival rate. This is due to both the

aggressive nature of the disease and the lack of

specific symptoms and early-detection tools. It is

relatively refractory to traditional cytotoxic agents

and radiotherapy. Gemcitabine, the standard

chemotherapy agent for the treatment of pancreatic

cancer, induces autophagy of cancer cells and that

this process mediates the cell death-promoting

activity of this compound. Early induction of

autophagy by gemcitabine leads to cancer cell death

and this cellular process is mediated by the

activation of VMP1 expression. In PANC-1 and

MIAPaCa-2 cells the inhibition of autophagy

significantly reduced the percentage of dead cells in

response to gemcitabine. In addition, gemcitabine

promoted early VMP1 expression, and

downregulation of VMP1 expression significantly

reduced cell death.

Acute pancreatitis

Disease

VMP1 was characterized because is not

constitutively expressed in pancreatic acinar cells

and it is highly activated early during experimental

acute pancreatitis in acinar cells. VMP1 is an

autophagy-related membrane protein involved in

the initial steps of the mammalian cell autophagic

process. VMP1 is a transmembrane protein that co-

localizes with LC3, a marker of the

autophagosomes, in pancreas tissue undergoing

pancreatitis-induced autophagy. VMP1 interacts

with with Beclin1, a mammalian autophagy

initiator, to start autophagosome formation. We

developed the ElaI-VMP1 mouse in which acinar

cell-specific constitutive expression of a VMP1-

EGFP chimera induces the formation of

autophagosomes. Upon CCK-R hyperstimulation,

wild type mice developed acute pancreatitis with

high amylase and lipase serum levels.

On the contrary, enzymatic levels in cerulein-

treated ElaI-VMP1 mice were significantly lower

compared to wild type mice. Consistently, ElaI-

VMP1 mouse pancreata showed remarkably less

macroscopic evidence of acute pancreatitis

compared to wild type animals, which showed

marked edema and hemorrhage. Histological

analyses displayed a high degree of necrosis as well

as infiltration in wild type pancreata with acute

pancreatitis. In contrast, neither necrosis nor

significant inflammation was seen in cerulein-

treated ElaI-VMP1 mice. ElaIVMP1 mice showed

secretory granules with normal ultrastructural

characteristics CCK-R hyperstimulation in wild

type animals induced a markedly altered

distribution pattern of the secretory granules.

Acinar cells lose their polarity, which results in the

relocation of zymogen granules to the basolateral

membrane. These alterations in vesicular traffic are

known to occur in acinar cells during acute

pancreatitis and upon hyperstimulation of their

CCK-R with cerulein. ElaI-VMP1 mice subjected

to CCK-R hyperstimulation revealed that acinar

cells preserve their structure and polarity with

negligible or no alteration in vesicular transport.

Surprisingly, in pancreata from cerulein-treated

ElaI-VMP1 mice, we observed autophagosomes

containing zymogen granules displaying a distinct

localization to the apical area of the acinar cell.

VMP1, the ubiquitin-protease USP9x, and the

ubiquitin-binding protein p62 mediate this process.

Moreover, VMP1 interacts with USP9x, indicating

that there is a close cooperation between the

autophagy pathway and the ubiquitin recognition

machinery required for selective autophagosome

formation. We have coined the term "zymophagy"

to refer to this process. Zymophagy is activated by

experimental pancreatitis and by acute pancreatitis

in humans. Furthermore, zymophagy has

pathophysiological relevance by controlling

pancreatitis-induced intracellular zymogen

activation and helping to prevent cell death. This

new selective autophagy is activated in pancreatic

acinar cells during pancreatitis-induced vesicular

transport alteration to sequester and degrade

potentially deleterious activated zymogen granules.

Page 49: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

VMP1 (vacuole membrane protein 1) Ropolo A, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 225

Confocal microscopy of AR42J cell transfected with

pEGFP-VMP1.

Diabetes

Disease

Experimental diabetes activates VMP1 expression

and autophagy in pancreas beta cells as a direct

response to streptozotocin (STZ). VMP1 mRNA

expression is activated after STZ treatment by islet

beta cells. Electron microscopy shows chromatin

aggregation and autophagy morphology that was

confirmed by LC3 expression and LC3-VMP1 co-

localization. Apoptotic cell death and the reduction

of beta cell pool are evident after 24h treatment,

while VMP1 is still expressed in the remaining

cells. VMP1-Beclin1 colocalization in pancreas

tissue from STZ-treated rats suggests that VMP1-

Beclin1 interaction is involved in the autophagic

process activation during experimental diabetes.

Pancreas beta cells trigger VMP1 expression and

autophagy during the early cellular events in

response to experimental diabetes.

References Dusetti NJ, Jiang Y, Vaccaro MI, Tomasini R, Azizi Samir A, Calvo EL, Ropolo A, Fiedler F, Mallo GV, Dagorn JC, Iovanna JL. Cloning and expression of the rat vacuole membrane protein 1 (VMP1), a new gene activated in pancreas with acute pancreatitis, which promotes vacuole formation. Biochem Biophys Res Commun. 2002 Jan 18;290(2):641-9

Vaccaro MI, Grasso D, Ropolo A, Iovanna JL, Cerquetti MC. VMP1 expression correlates with acinar cell cytoplasmic vacuolization in arginine-induced acute pancreatitis. Pancreatology. 2003;3(1):69-74

Jiang PH, Motoo Y, Vaccaro MI, Iovanna JL, Okada G, Sawabu N. Expression of vacuole membrane protein 1 (VMP1) in spontaneous chronic pancreatitis in the WBN/Kob rat. Pancreas. 2004 Oct;29(3):225-30

Ropolo A, Grasso D, Pardo R, Sacchetti ML, Archange C, Lo Re A, Seux M, Nowak J, Gonzalez CD, Iovanna JL, Vaccaro MI. The pancreatitis-induced vacuole membrane protein 1 triggers autophagy in mammalian cells. J Biol Chem. 2007 Dec 21;282(51):37124-33

Vaccaro MI. Autophagy and pancreas disease. Pancreatology. 2008;8(4-5):425-9

Vaccaro MI, Ropolo A, Grasso D, Iovanna JL. A novel mammalian trans-membrane protein reveals an alternative initiation pathway for autophagy. Autophagy. 2008 Apr;4(3):388-90

Grasso D, Sacchetti ML, Bruno L, Lo Ré A, Iovanna JL, Gonzalez CD, Vaccaro MI. Autophagy and VMP1 expression are early cellular events in experimental diabetes. Pancreatology. 2009;9(1-2):81-8

Pardo R, Lo Ré A, Archange C, Ropolo A, Papademetrio DL, Gonzalez CD, Alvarez EM, Iovanna JL, Vaccaro MI. Gemcitabine induces the VMP1-mediated autophagy pathway to promote apoptotic death in human pancreatic cancer cells. Pancreatology. 2010;10(1):19-26

Grasso D, Ropolo A, Lo Ré A, Boggio V, Molejón MI, Iovanna JL, Gonzalez CD, Urrutia R, Vaccaro MI. Zymophagy, a novel selective autophagy pathway mediated by VMP1-USP9x-p62, prevents pancreatic cell death. J Biol Chem. 2011 Mar 11;286(10):8308-24

This article should be referenced as such:

Ropolo A, Lo Ré A, Vaccaro MI. VMP1 (vacuole membrane protein 1). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):223-225.

Page 50: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 226

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

XPO1 (exportin 1 (CRM1 homolog, yeast)) Alessandra Ruggiero, Maria Giubettini, Patrizia Lavia

CNR (National Research Council), Institute of Molecular Biology and Pathology, c/o Sapienza

University of Rome, via degli Apuli 4, 00185 Rome, Italy (AR, MG, PL)

Published in Atlas Database: November 2011

Online updated version : http://AtlasGeneticsOncology.org/Genes/XPO1ID44168ch2p15.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI XPO1ID44168ch2p15.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Other names: CRM1, DKFZp686B1823, emb

HGNC (Hugo): XPO1

Location: 2p15

Note

The human XPO1/hCRM1 gene is localized on the

2p16 region (Fornerod et al., 1997a).

DNA/RNA

Transcription

The human XPO1/hCRM1 gene is transcribed in a

cell cycle-dependent manner, with the onset of

mRNA transcription taking place in late G1 phase

and peaking in the G2/M phases of the cell cycle

(Kudo et al., 1997). NFY/CBP, Sp1 and p53

transcription factors are reported to interact with the

XPO1/hCRM1 gene promoter and play an

important role in XPO1/hCRM1 promoter activity

in transformed and cancer cells (van der Watt and

Leaner, 2011).

Protein

Note

A human protein, originally named CC112 based

on its apparent molecular weight, was identified in

a search for interacting partners of CAN/NUP214, a

nucleoporin regarded as a proto-oncogenic factor.

CAN was implicated in acute myeloid leukemia

and in myelodysplastic syndrome (von Lindern et

al., 1992) as part of the DEK-CAN fusion gene

generated in the translocation t(6;9)(p23;q34).

Another potentially oncogenic fusion protein

involving CAN was identified in a patient with

acute undifferentiated leukemia, in which case the

t(6;9) yielded a SET-CAN fusion. Wild-type CAN

is identical to the nucleoporin NUP214. CC112 was

capable of interacting with both wild-type

CAN/NUP214 and with both its fusion proteins,

DEK-CAN and SET-CAN, suggesting potential

roles in proliferation of cancer cells (Fornerod et

al., 1996).

Description

The human XPO1/CRM1 protein is composed of

1071 aminoacidic residues with a molecular weight

of 112 kDa (Fornerod et al., 1997b). It is a modular

protein composed of several fuctional domains:

- The N-terminal region shares sequence similarity

with importin β in a region called the CRIME

domain (acronym for CRM1, importin beta etc.).

This domain interacts with the GTPase RAN. In the

GTP-bound form, RAN stabilizes export complex

formed by CRM1 and NES-containing proteins.

- Most of the XPO1/CRM1 protein is composed of

19 HEAT repeat motifs. HEAT repeat 8 contains an

acidic loop which cooperates with the CRIME

domain in RANGTP binding.

- The central region of XPO1/CRM1 is involved in

NES binding. Cys528, lying in this region, is

specifically blocked by the inhibitor leptomycin B

(LMB), which therefore blocks the export activity

of XPO1/CRM1 (Wolff et al., 1997).

- The C-terminal region is thought to modulate the

affinity of XPO1/CRM1 for its cargoes.

Structures The structure of the region corresponding to

residues 707-1034 (C-terminal region) was

elucidated by X-ray crystallography (Petosa et al.,

2004).

The structure of XPO1/CRM1 complexed to

various NESs and to RANGTP has been solved

(Güttler et al., 2010).

Page 51: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

XPO1 (exportin 1 (CRM1 homolog, yeast)) Ruggiero A, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 227

The plates show the subcellular localisation of CRM1 (detected by indirect immunofluorescence) in interphase and mitotic human

HeLa cells. Upper row: an interphase cell showing CRM1 (in red in the left panel) within the nucleus and especially around the nuclear envelope, where it concentrates with a regular, punctuated pattern typical of the association with nuclear pore

complexes. The nuclear shape is depicted in the upper right panel by staining the DNA with the fluorochrome 4',6-diamidino-2-phenylindole (DAPI, in blue). Lower row: a metaphase cell showing CRM1 (in red in the left panel) concentrating at the

kinetochores (compare with the middle panel, where kinetochore proteins are stained using CREST antiserum and a blue-emitting secondary antibody). A CRM1 fraction is also visible at spindle poles (compare with the staining of the mitotic spindle

microtubules using an antibody against alpha-tubulin, in green). The merged picture shows a 3.5x magnification of the overlay of all three images: CRM1 (red) lies at the interface between the kinetochores (blue) and the microtubules (green) projecting from

opposite spindle poles.

Expression

XPO1/CRM1 protein levels remain constant

throughout the cell cycle (Kudo et al., 1997).

Localisation

Due to its function as a shuttling nuclear transport

receptor between the nucleus and cytoplasm, the

human XPO1/CRM1 protein is preferentially

localized at the nuclear envelope in interphase cells

(Kudo et al., 1997; Fornerod et al., 1997b). In the

nucleus it can be detected in specific bodies called

CRM1 nucleolar bodies (CNoBs). CNoBs depend

on RNA polymerase I activity, suggesting a role in

ribosome biogenesis (Ernoult-Lange et al., 2009).

In mitotic cells, a fraction of XPO1 is found at

centrosomes (Forgues et al., 2003; Wang et al.,

2005) and a substantial fraction localizes to the

kinetochores (Arnaoutov et al., 2005).

Function

hCRM1 was found to interact stably in complexes

containing not only NUP214/CAN (or its

derivatives), but also another component of nuclear

pores, the nucleoporin NUP88 (Fornerod et al.,

1997b). These interactions hinted at a possible role

of hCRM1 in nucleocytoplasmic transport. Further

studies indeed demonstrated that hCRM1 acts as a

nuclear export factor (reviewed by Fried and Kutaj,

2003; Hutten and Kehlenbach, 2007): it interacts

with various classes of RNAs and with proteins

carrying nuclear export signals (NES) (Fornerod et

al., 1997c; Fukuda et al., 1997; Ossareh-Nazari et

al., 1997), short aminoacidic stretches harbouring

hydrophobic residues (general consensus LX(2-

3)ΦX(2-3)LXΦ, where can be L, I, M or F), present

in many shuttling proteins of cellular or viral origin,

and transports these molecules out of the nucleus

through nuclear pore complexes in a manner

dependent on the GTPase RAN. The protein is

therefore alternatively called either exportin-1 or

XPO1, based on its function, or hCRM1, based on

evolutionary conservation.

Regulated export of some shuttling proteins (e.g.,

p53, p27, STAT, NF-kB and many viral proteins)

out of the nucleus is essential for regulated cell

cycle and cell proliferation (reviewed by Fabbro

and Henderson, 2003; Rensen et al., 2008). This has

lead some authors to view nuclear export as a

promising target process in cancer therapy

(reviewed by Yashiroda and Yoshida, 2003; Turner

and Sullivan, 2008).

Recent findings have revealed additional roles of

XPO1/CRM1 in mitosis: first, an XPO1/CRM1

Page 52: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

XPO1 (exportin 1 (CRM1 homolog, yeast)) Ruggiero A, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 228

fraction regulates the localisation of nucleophosmin

(NPM/B23), a regulator of centrosome duplication.

XPO1/CRM1 is required to prevent centrosome

overduplication and the formation of multipolar

spindles (reviewed by Budhu and Wang, 2005;

Ciciarello and Lavia, 2005). Second, a kinetochore-

associated fraction of XPO1/CRM1 regulates the

assembly of the so-called k-fibers, bundles of

microtubules that stably connect the spindle poles

to the kinetochores of mitotic chromosomes to

ensure proper chromosome segregation (reviewed

by Arnaoutov and Dasso, 2005; Ciciarello and

Lavia, 2005; Dasso, 2006). Third, XPO1/CRM1

regulates survivin, a member of the chromosomal

passenger complex with roles in chromosome

segregation and apoptosis (reviewed by Knauer et

al., 2007).

In synthesis, XPO1/CRM1 acts in control of cell

proliferation, and affects loss of proliferation

control in cancer cells, through several pathways: 1.

as a nuclear export factor, it directly regulates the

subcellular localisation, and hence the activity, of

oncogenes and tumour suppressor proteins that

contain nuclear export sequences; 2. it acts in

control of the mitotic apparatus and chromosome

segregation; 3. it influences the maintenance of

nuclear and chromosome structure.

Homology

The human protein originally named CC112

showed homology to the Schizosaccharomyces

pombe CRM1 protein, first identified for being

implicated in the control of higher order

chromosome structure: mutation of the coding gene

was associated with the appearance of "deformed

nuclear chromosome domains" in fission yeast

conditional mutant strains. The gene product was

therefore named CRM1 (chromosome region

maintenance 1; Adachi and Yanagida, 1989). Based

on this homology, the human protein name of

CC112 was abandoned and the name hCRM1 was

used.

Implicated in

Ovarian cancer (Noske et al., 2008)

Prognosis

Increased nuclear (52.7%) and cytoplasmic (56.8%)

expression of CRM1 were reported observed in

carcinomas compared with borderline tumors and

benign lesions. Cytoplasmic CRM1 expression

significantly correlated with advanced tumor stage

(P= 0.043), poorly differentiated carcinomas (P=

0.011) and high mitotic rate (P= 0.008). Nuclear

CRM1 was significantly associated with high

cyclooxygenase-2 (COX-2) expression (P= 0.002)

and poor overall survival (P= 0.01). CRM1 was

previously directly implicated in nuclear export of

COX-2 (Jang et al., 2003). The study by Noske et

al. (2008) suggests that elevated expression of

CRM1 may be causal to COX-2 up-regulation, with

direct clinical relevance.

Oncogenesis

CRM1 is highly expressed in ovarian carcinomas

tissues and regulates export of COX-2.

Osteosarcoma (Yao et al., 2009)

Prognosis

The CRM1 protein is reported to be expressed with

increased abundance in osteosarcoma compared to

non-tumour tissues (P= 0.037, 57 patients). High

levels of CRM1 were significantly associated with

increased serum levels of alkaline phosphatase

(ALP, P= 0.001). In univariate analysis, a

significant association between CRM1 expression

and tumor size (P= 0.014), as well as histological

grade (P= 0.003) was observed. In Kaplan-Meier

survival analysis, high CRM1 expression was a

significant prognostic indicator for poor

progression-free survival (P= 0.016) as well as

overall survival (P= 0.008). Multivariate analysis

demonstrated that high expression of CRM1 was

significantly related to shorter survival (95% CI,

1.27-5.39).

Oncogenesis

CRM1 is significantly increased in osteosarcoma

compared with normal tissue.

Cervical cancer (van der Watt et al., 2009)

Oncogenesis

CRM1 protein abundance is significantly increased

in cervical cancer cells compared with normal

tissue (P< 0.05). Inhibition of CRM1 by RNA

interference resulted in increased cell death,

associated with nuclear retention of p53, likely

protecting p53 from degradation as the latter

predominantly occurs in the cytoplasm.

Pancreas cancer (Huang et al., 2009)

Prognosis

Increased expression abundance of CRM1 protein

was detected in pancreatic cancer tissues (P=

0.0013, 69 patients at stages I and II). CRM1

expression correlates with increased levels of serum

CEA (P= 0.002) and CA19.9 (P= 0.005), tumour

size (P= 0.011), lymphadenopathy (P= 0.004) and

metastasis (P= 0.0041). High CRM1 expression

was a prognostic indicator for progression-free

survival (PFS) (P= 0.0011) as well as overall

survival (OS) (P= 0.004). The authors proposed that

CRM1 be used as a prognostic parameter for poor

PFS and OS (95% CI, 1.27-5.39).

Glioma (Shen et al., 2009)

Prognosis

CRM1 overexpression is significantly associated

with the pathological state (P= 0.001, 56 patients),

with glioma tumour grade, with high expression of

phospho-ser10p27 and with reduced overall

abundance of p27.

Page 53: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

XPO1 (exportin 1 (CRM1 homolog, yeast)) Ruggiero A, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 229

Oncogenesis

Given the direct implication of CRM1 in nuclear

export of p27, the data in this study suggest that

increased CRM1 abundance yields increased

cytoplasmic localisation of p27, which is probably

targeted to degradation, leading to uncontrolled

tumour growth. Phospho-ser10p27 may be resistant

to CRM1-mediated nuclear export. High CRM1

and low p27 expression are associated with high

grade glioma and high CRM1 protein expression is

proposed as a prognostic factor of overall survival

and poor outcome.

To be noted

Note

CRM1 protein levels are abnormally high in several

cancers, with high levels of CRM1 being associated

with poor patient survival (van der Watt and

Leaner, 2011).

References Adachi Y, Yanagida M. Higher order chromosome structure is affected by cold-sensitive mutations in a Schizosaccharomyces pombe gene crm1+ which encodes a 115-kD protein preferentially localized in the nucleus and its periphery. J Cell Biol. 1989 Apr;108(4):1195-207

von Lindern M, Fornerod M, van Baal S, Jaegle M, de Wit T, Buijs A, Grosveld G. The translocation (6;9), associated with a specific subtype of acute myeloid leukemia, results in the fusion of two genes, dek and can, and the expression of a chimeric, leukemia-specific dek-can mRNA. Mol Cell Biol. 1992 Apr;12(4):1687-97

Fornerod M, Boer J, van Baal S, Morreau H, Grosveld G. Interaction of cellular proteins with the leukemia specific fusion proteins DEK-CAN and SET-CAN and their normal counterpart, the nucleoporin CAN. Oncogene. 1996 Oct 17;13(8):1801-8

Fornerod M, van Baal S, Valentine V, Shapiro DN, Grosveld G. Chromosomal localization of genes encoding CAN/Nup214-interacting proteins--human CRM1 localizes to 2p16, whereas Nup88 localizes to 17p13 and is physically linked to SF2p32. Genomics. 1997a Jun 15;42(3):538-40

Fornerod M, van Deursen J, van Baal S, Reynolds A, Davis D, Murti KG, Fransen J, Grosveld G. The human homologue of yeast CRM1 is in a dynamic subcomplex with CAN/Nup214 and a novel nuclear pore component Nup88. EMBO J. 1997b Feb 17;16(4):807-16

Fornerod M, Ohno M, Yoshida M, Mattaj IW. CRM1 is an export receptor for leucine-rich nuclear export signals. Cell. 1997c Sep 19;90(6):1051-60

Fukuda M, Asano S, Nakamura T, Adachi M, Yoshida M, Yanagida M, Nishida E. CRM1 is responsible for intracellular transport mediated by the nuclear export signal. Nature. 1997 Nov 20;390(6657):308-11

Kudo N, Khochbin S, Nishi K, Kitano K, Yanagida M, Yoshida M, Horinouchi S. Molecular cloning and cell cycle-dependent expression of mammalian CRM1, a protein involved in nuclear export of proteins. J Biol Chem. 1997 Nov 21;272(47):29742-51

Ossareh-Nazari B, Bachelerie F, Dargemont C. Evidence for a role of CRM1 in signal-mediated nuclear protein export. Science. 1997 Oct 3;278(5335):141-4

Wolff B, Sanglier JJ, Wang Y. Leptomycin B is an inhibitor of nuclear export: inhibition of nucleo-cytoplasmic translocation of the human immunodeficiency virus type 1 (HIV-1) Rev protein and Rev-dependent mRNA. Chem Biol. 1997 Feb;4(2):139-47

Fabbro M, Henderson BR. Regulation of tumor suppressors by nuclear-cytoplasmic shuttling. Exp Cell Res. 2003 Jan 15;282(2):59-69

Forgues M, Difilippantonio MJ, Linke SP, Ried T, Nagashima K, Feden J, Valerie K, Fukasawa K, Wang XW. Involvement of Crm1 in hepatitis B virus X protein-induced aberrant centriole replication and abnormal mitotic spindles. Mol Cell Biol. 2003 Aug;23(15):5282-92

Fried H, Kutay U. Nucleocytoplasmic transport: taking an inventory. Cell Mol Life Sci. 2003 Aug;60(8):1659-88

Jang BC, Muñoz-Najar U, Paik JH, Claffey K, Yoshida M, Hla T. Leptomycin B, an inhibitor of the nuclear export receptor CRM1, inhibits COX-2 expression. J Biol Chem. 2003 Jan 31;278(5):2773-6

Yashiroda Y, Yoshida M. Nucleo-cytoplasmic transport of proteins as a target for therapeutic drugs. Curr Med Chem. 2003 May;10(9):741-8

Petosa C, Schoehn G, Askjaer P, Bauer U, Moulin M, Steuerwald U, Soler-López M, Baudin F, Mattaj IW, Müller CW. Architecture of CRM1/Exportin1 suggests how cooperativity is achieved during formation of a nuclear export complex. Mol Cell. 2004 Dec 3;16(5):761-75

Arnaoutov A, Azuma Y, Ribbeck K, Joseph J, Boyarchuk Y, Karpova T, McNally J, Dasso M. Crm1 is a mitotic effector of Ran-GTP in somatic cells. Nat Cell Biol. 2005 Jun;7(6):626-32

Arnaoutov A, Dasso M. Ran-GTP regulates kinetochore attachment in somatic cells. Cell Cycle. 2005 Sep;4(9):1161-5

Budhu AS, Wang XW. Loading and unloading: orchestrating centrosome duplication and spindle assembly by Ran/Crm1. Cell Cycle. 2005 Nov;4(11):1510-4

Ciciarello M, Lavia P. New CRIME plots. Ran and transport factors regulate mitosis. EMBO Rep. 2005 Aug;6(8):714-6

Wang W, Budhu A, Forgues M, Wang XW. Temporal and spatial control of nucleophosmin by the Ran-Crm1 complex in centrosome duplication. Nat Cell Biol. 2005 Aug;7(8):823-30

Dasso M. Ran at kinetochores. Biochem Soc Trans. 2006 Nov;34(Pt 5):711-5

Hutten S, Kehlenbach RH. CRM1-mediated nuclear export: to the pore and beyond. Trends Cell Biol. 2007 Apr;17(4):193-201

Knauer SK, Mann W, Stauber RH. Survivin's dual role: an export's view. Cell Cycle. 2007 Mar 1;6(5):518-21

Noske A, Weichert W, Niesporek S, Röske A, Buckendahl AC, Koch I, Sehouli J, Dietel M, Denkert C. Expression of the nuclear export protein chromosomal region maintenance/exportin 1/Xpo1 is a prognostic factor in human ovarian cancer. Cancer. 2008 Apr 15;112(8):1733-43

Rensen WM, Mangiacasale R, Ciciarello M, Lavia P. The GTPase Ran: regulation of cell life and potential roles in cell transformation. Front Biosci. 2008 May 1;13:4097-121

Turner JG, Sullivan DM. CRM1-mediated nuclear export of proteins and drug resistance in cancer. Curr Med Chem. 2008;15(26):2648-55

Page 54: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

XPO1 (exportin 1 (CRM1 homolog, yeast)) Ruggiero A, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 230

Ernoult-Lange M, Wilczynska A, Harper M, Aigueperse C, Dautry F, Kress M, Weil D. Nucleocytoplasmic traffic of CPEB1 and accumulation in Crm1 nucleolar bodies. Mol Biol Cell. 2009 Jan;20(1):176-87

Huang WY, Yue L, Qiu WS, Wang LW, Zhou XH, Sun YJ. Prognostic value of CRM1 in pancreas cancer. Clin Invest Med. 2009 Dec 1;32(6):E315

Shen A, Wang Y, Zhao Y, Zou L, Sun L, Cheng C. Expression of CRM1 in human gliomas and its significance in p27 expression and clinical prognosis. Neurosurgery. 2009 Jul;65(1):153-9; discussion 159-60

van der Watt PJ, Maske CP, Hendricks DT, Parker MI, Denny L, Govender D, Birrer MJ, Leaner VD. The Karyopherin proteins, Crm1 and Karyopherin beta1, are overexpressed in cervical cancer and are critical for cancer cell survival and proliferation. Int J Cancer. 2009 Apr 15;124(8):1829-40

Yao Y, Dong Y, Lin F, Zhao H, Shen Z, Chen P, Sun YJ, Tang LN, Zheng SE. The expression of CRM1 is associated with prognosis in human osteosarcoma. Oncol Rep. 2009 Jan;21(1):229-35

Güttler T, Madl T, Neumann P, Deichsel D, Corsini L, Monecke T, Ficner R, Sattler M, Görlich D. NES consensus redefined by structures of PKI-type and Rev-type nuclear export signals bound to CRM1. Nat Struct Mol Biol. 2010 Nov;17(11):1367-76

van der Watt PJ, Leaner VD. The nuclear exporter, Crm1, is regulated by NFY and Sp1 in cancer cells and repressed by p53 in response to DNA damage. Biochim Biophys Acta. 2011 Jul;1809(7):316-26

This article should be referenced as such:

Ruggiero A, Giubettini M, Lavia P. XPO1 (exportin 1 (CRM1 homolog, yeast)). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):226-230.

Page 55: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 231

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(11;18)(p15;q12) Jean-Loup Huret

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

France (JLH)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t1118p15q12ID1466.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI t1118p15q12ID1466.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Clinics and pathology

Disease

T-cell acute lymphoid leukemia (T-ALL)

Epidemiology

One case to date, a 9-year-old boy.

Evolution

Remission was obtained and the patient remains in

complete remission 28 months after diagnosis.

Cytogenetics

Cytogenetics morphological

The translocation was accompanied with a del(12p).

Genes involved and proteins

NUP98

Location

11p15.4

Protein

Nucleoporin: associated with the nuclear pore

complex. Role in nucleocytoplasmic transport

processes.

SETBP1

Location

18q12.3

Protein

Contains 3 DNA binding domains (A.T hooks).

SETBP1 protects SET from protease cleavage.

SETBP1 forms a complex with SET and PP2A

(protein phosphatase 2A). SETBP1 impairs PP2A

activity via SET and promotes proliferation of acute

myeloid leukemia cells (Cristóbal et al., 2010).

Germinal mutations

In Schinzel-Giedion midface retraction syndrome.

Result of the chromosomal anomaly

Hybrid gene

Description

5' NUP98 - 3' SETBP1

Transcript

Exon 12 of NUP98 (nucleotide (nt) 1552) fused in-

frame with exon 5 of SETBP1 (nt 4015).

References Panagopoulos I, Kerndrup G, Carlsen N, Strömbeck B, Isaksson M, Johansson B. Fusion of NUP98 and the SET binding protein 1 (SETBP1) gene in a paediatric acute T cell lymphoblastic leukaemia with t(11;18)(p15;q12). Br J Haematol. 2007 Jan;136(2):294-6

Cristóbal I, Blanco FJ, Garcia-Orti L, Marcotegui N, Vicente C, Rifon J, Novo FJ, Bandres E, Calasanz MJ, Bernabeu C, Odero MD. SETBP1 overexpression is a novel leukemogenic mechanism that predicts adverse outcome in elderly patients with acute myeloid leukemia. Blood. 2010 Jan 21;115(3):615-25

This article should be referenced as such:

Huret JL. t(11;18)(p15;q12). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3): 231.

Page 56: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 232

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(11;21)(q21;q22) Jean-Loup Huret

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

France (JLH)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t1121q21q22ID1592.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI t1121q21q22ID1592.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Note

This translocation is different from the

t(11;21)(q12;q22) with MACROD1/RUNX1

involvement.

Clinics and pathology

Disease

Acute myeloid leukemia (AML)

Epidemiology

One case to date, a 65-year-old male patient with

M2-AML (Dai et al., 2007).

Evolution

The patient died 10 months after diagnosis.

Genes involved and proteins

LPXN

Protein

LPXN contains two types of protein-protein

interaction domains: leucine-aspartate (LD) repeats

in N-term, and LIM (Lin-11 Isl-1 Mec-3) domains

at the C-term. Belongs to the paxillin family (PXN,

LPXN, TGFB1I1). Protein involved in focal

adhesion. LPXN and paxillin had opposite roles in

adhesion to collagen LPXN siRNA stimulated

whereas paxillin siRNA inhibited cell adhesion.

Strongly expressed in hematopoietic cells. LPXN is

involved in bone resorption and stimulates prostate

cancer cell migration (Chen and Kroog, 2010).

RUNX1

Location

21q22.3

Protein

Transcription factor (activator) for various

hematopoietic-specific genes.

Result of the chromosomal anomaly

Hybrid gene

Description

5' RUNX1 - 3' LPXN

Transcript

Two in frame fusion transcripts -fusion of exon 5 or

6 of RUNX1 to LPXN exon 8.

Fusion protein

Description

The two variant fusion proteins RUNX1-LPXN

localized in the nucleus and inhibited RUNX1

transactivation (Dai et al., 2009). It is hypothesized

that the reciprocal LPXN-RUNX1 may also play a

role in leukemogenesis.

Page 57: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

t(11;21)(q21;q22) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 233

References Dai H, Xue Y, Pan J, Wu Y, Wang Y, Shen J, Zhang J.. Two novel translocations disrupt the RUNX1 gene in acute myeloid leukemia. Cancer Genet Cytogenet. 2007 Sep;177(2):120-4.

Dai HP, Xue YQ, Zhou JW, Li AP, Wu YF, Pan JL, Wang Y, Zhang J.. LPXN, a member of the paxillin superfamily, is fused to RUNX1 in an acute myeloid leukemia patient with a t(11;21)(q12;q22) translocation. Genes Chromosomes Cancer. 2009 Dec;48(12):1027-36.

Chen PW, Kroog GS.. Leupaxin is similar to paxillin in focal adhesion targeting and tyrosine phosphorylation but has distinct roles in cell adhesion and spreading. Cell Adh Migr. 2010 Oct-Dec;4(4):527-40.

This article should be referenced as such:

Huret JL. t(11;21)(q21;q22). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):232-233.

Page 58: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Leukaemia Section Short Communication

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 234

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

t(8;17)(q24;q22) ???BCL3/MYC Jean-Loup Huret

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

France (JLH)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Anomalies/t0817q24q22ID1494.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI t0817q24q22ID1494.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity

Note

It is unlikely that the BCL3 gene (HGNC official

name) is involved in this translocation with a

breakpoint in 17q22, since BCL3 sits in 19q13.32

(coordonates: starts at 45251978 and ends at

45263301 bp from 19pter); the alternative would be

a cryptic translocation, involving a cryptic inserted

fragment of 19q13.32, including BCL3, within

17q22.

Clinics and pathology

Disease

Aggresive prolymphocytic leukemia

Epidemiology

Only one case to date, with no clinical data.

Cytogenetics

Cytogenetics morphological

The karyotype also showed the classical

t(14;18)(q32;q21), usually found in follicular

lymphoma, a 12q+ and a Xp+, not otherwise

described.

Genes involved and proteins

Note

As said above, it is unprobable that the MYC

partner is BCL3.

MYC

Protein

MYC regulates the transcription of genes required

to coordinate a range of cellular processes,

including those essential for proliferation, growth,

differentiation, apoptosis and self-renewal, and

protein synthesis through ribosome biogenesis (van

Riggelen et al., 2010).

BCL3

Location

19q13.32

Protein

BCL3 is mainly found in the nucleus. Protein which

contains seven ankyrin repeats. Ankyrin repeats are

found in IkB family members, including IkBa,

IkBb, and IkBe. BCL3 is a member of the IkappaB

family, whose proteins regulate the NFkappaB

family of transcription factors. Component of a

complex with a NF-kB p52-p52 homodimer Down-

regulates inflammatory responses through limiting

the transcription of NF-kB-dependent genes. Binds

to NF-kB p50 and p52, Jab1, Pirin, Tip60 and

Bard1. Bcl-3 is an adaptor protein (Dechend et al.,

1999; Kreisel et al., 2011). Regulates genes

involved in cell proliferation and apoptosis.

NFkappaB plays a major role in B-cell

development.

Result of the chromosomal anomaly

Hybrid gene

Description

Disruption of MYC close to the first intron, with

the decapitation of the first intron, replaced by a

sequence of 1.7 kb, that the authors have called

"BCL3".

Page 59: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

t(8;17)(q24;q22) ???BCL3/MYC Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 235

References Gauwerky CE, Huebner K, Isobe M, Nowell PC, Croce CM. Activation of MYC in a masked t(8;17) translocation results in an aggressive B-cell leukemia. Proc Natl Acad Sci U S A. 1989 Nov;86(22):8867-71

Dechend R, Hirano F, Lehmann K, Heissmeyer V, Ansieau S, Wulczyn FG, Scheidereit C, Leutz A. The Bcl-3 oncoprotein acts as a bridging factor between NF-kappaB/Rel and nuclear co-regulators. Oncogene. 1999 Jun 3;18(22):3316-23

van Riggelen J, Yetil A, Felsher DW. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat Rev Cancer. 2010 Apr;10(4):301-9

Kreisel D, Sugimoto S, Tietjens J, Zhu J, Yamamoto S, Krupnick AS, Carmody RJ, Gelman AE. Bcl3 prevents acute inflammatory lung injury in mice by restraining emergency granulopoiesis. J Clin Invest. 2011 Jan 4;121(1):265-76

This article should be referenced as such:

Huret JL. t(8;17)(q24;q22) ???BCL3/MYC. Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):234-235.

Page 60: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Deep Insight Section

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 236

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Plasticity and Tumorigenicity Elena Campos-Sanchez, Isidro Sanchez-Garcia, Cesar Cobaleda

Centro de Biologia Molecular "Severo Ochoa", CSIC/Universidad Autonoma de Madrid, C/Nicolas

Cabrera 1, Universidad Autonoma, Cantoblanco, 28049 Madrid, Spain (ECS, CC), Experimental

Therapeutics and Translational Oncology Program, Instituto de Biologia Molecular y Celular del

Cancer, CSIC/ Universidad de Salamanca, Campus M de Unamuno s/n, 37007-Salamanca, Spain

(ISG)

Published in Atlas Database: September 2011

Online updated version : http://AtlasGeneticsOncology.org/Deep/PluripotencyID20103.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI PluripotencyID20103.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Summary The research fields of developmental biology and oncology have always been tightly linked, since the times of

Rudolf Virchow's cellular theory ("omnis cellula e cellula") and embryonal rest hypothesis. On the other side,

for many years, contemporary cancer research has been mainly focused on the altered controls of proliferation in

tumoral cells. This has been reflected in the therapeutic approaches employed in the clinic to treat the patients:

with very few exceptions, anti-cancer treatments are targeted at the mechanisms of abnormal tumoral growth.

Such therapies, however, are very unspecific, highly toxic and, ultimately, inefficient in most cases. In the last

years, a new recognition of the role of aberrant differentiation at the root of cancer has arisen, mainly driven by

the coming of age of the "cancer stem cell" (CSC) theory. From this point of view, the comprehensive

knowledge of the developmental mechanisms by which normal cells acquire their identity is essential to

understand how these controls are deregulated in tumours. New insights into the mechanisms that maintain the

molecular boundaries of cell identity have been gained from the study of induced pluripotency, showing that cell

fate can be much more susceptible to change than previously thought. Applied to cancer, these findings imply

that the oncogenic events that take place in an otherwise healthy cell lead to a reprogramming of the normal

cellular fate and establish a new pathologic developmental program. Therefore, cancer reprogramming and

cellular plasticity are closely related, since only some cells possess the plasticity required to allow

reprogramming to occur, and only some oncogenic events can, in the right plastic cell, induce this change. Here

we discuss the latest findings in the fields of cellular plasticity and reprogramming and we consider their

consequences for our understanding of cancer development and treatment.

Historical perspective The search for the capacity of regenerating disease-

affected organs is probably as old as mankind

(Odelberg, 2004). The examples are abundant in

ancient religions, from the Egyptian god Osiris,

who resurrected and recomposed his maimed body

from the pieces that had been thrown into the Nile,

to the legendary Hydra that could regenerate its

severed heads. Or the mythological Prometheus,

who had his viscera eaten by an eagle every day,

only to regenerate them again. But also from a more

scientific point of view, it was already noticed by

Aristotle (384-322 BC) that lizards can regenerate

their tails after amputation. But until the 18th

century this knowledge was mainly anecdotic, and

only with the arrival of the Age of Enlightenment,

regeneration and plasticity will become the matter

of scientific research. In 1712, Réaumur describes

the regeneration of the limbs and claws of crayfish

(Réaumur, 1712); in 1744, Trembley discovers that

a part of the Hydra polyp can regenerate the

complete organism (Trembley, 1744); in 1769,

Spallanzani reports that tadpoles can regenerate

their tails and salamanders their amputated jaws,

limbs and tails (Spallanzani, 1769). The research

performed during most of the 19th

and first half of

Page 61: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 237

the 20th

centuries showed that, for regeneration to

occur, the cells that are normally forming part of

the organs are not sufficient, and a special type of

cells are required: the progenitor cells (Odelberg,

2004; Birnbaum and Sánchez Alvarado, 2008). The

origin of these cells was not very clear (and it is

still a matter of debate and intense research, in fact,

see (Sánchez Alvarado, 2000; Kragl et al., 2009;

Rinkevich et al., 2011)); for some tissues, like skin,

blood, muscles or bones, progenitors were shown to

exist in the tissues in small numbers, and to become

activated as a consequence of the lesions. In other

cases, the progenitors seemed to arise from mature

cells that become dedifferentiated. The best

example supporting this possibility has been

described in primitive vertebrates like the urodeles

(e.g. salamanders and axolotls). In these animals,

after a wound harms the organism, the cells from

the normal tissues form a group of cells known as

the regenerative blastema, which will generate all

the tissues in the new limb/tail (Chalkey, 1954;

Bodemer and Everett, 1959; Hay and Fischman,

1961). It has long been held that the blastema was

the result of cellular dedifferentiation to

progenitors. However, the most recent findings

seem to indicate that there is no cellular

dedifferentiation to progenitors involved in this

process, and the regeneration is always due to the

action of resident tissue-specific stem cells and

progenitors, thus questioning the role of mature

cellular plasticity in tissue regeneration (Kragl et

al., 2009; Rinkevich et al., 2011). We have

therefore seen how the study of "naturally"

occurring regeneration opened the way to a new

understanding of the stem cell-based architecture of

the organs and tissues, especially with the study of

primitive vertebrates. In 1952, amphibians also

provided the first animal model of experimentally-

induced reprogramming when Briggs and King

generated Xenopus tadpoles by transplanting the

nucleus of cells from the blastula into oocytes,

therefore reverting the cellular differentiation

program (Briggs and King, 1952). Afterwards, it

was shown that more differentiated cells, like those

from the intestinal epithelia, could also be

reprogrammed by nuclear transfer (Gurdon, 1962).

These landmark findings undoubtedly showed that

the genetic potential of cells was not lost during

differentiation, and that development did not imply

genetic changes. This principle was extended to

mammals with the cloning of Dolly the sheep in

1997 (Wilmut et al., 1997). This was the ultimate

proof showing that the changes that occur during

differentiation are totally reversible, and

demonstrated that the fate restrictions that take

place during development are the result of

epigenetic modifications. These studies also

showed that there were factors in the oocyte

cytoplasm capable of inducing a reprogramming

that led to the appearance of a totipotent phenotype.

In a parallel way, the search for the molecular

regulators responsible for establishing and

controlling cellular identity led finally to the

identification of the factors capable of

reprogramming cellular fate. In 1987, it was shown

that ectopic expression of the Antennapedia

homeotic gene lead to changes in the body plan of

Drosophila, that got extra legs instead of antennae

(Schneuwly et al., 1987). Also, Gehring et al.

showed that the ectopic expression of eyeless

controlled eye development and led to the

development of ectopic eyes in the fly's legs

(Gehring, 1996). In mammals, the first master

regulator factor to be identified was MyoD, which

was shown to be capable of transdifferentiating

fibroblasts into the myogenic lineage (Davis et al.,

1987). Other examples of factors with fate-

reprogramming capacity are C/EBPα, capable of

mediating the transdifferentiation of mouse B cells

into macrophages (Xie et al., 2004) or Pax5, whose

loss leads to the dedifferentiation of committed B

cells (Nutt et al., 1999; Cobaleda et al., 2007a;

Cobaleda and Busslinger, 2008). All these data

proved that the lack or excess of just one factor

could lead to a radical alteration of the

transcriptional profile and could cause stable fate

changes. This evidence, together with the one

coming from reprogramming by nuclear

transplantation, paved the way to the search for the

factors capable of reprogramming to full

pluripotency that led, in 2006, to the identification

of the four transcription factors capable of inducing

pluripotency in virtually every kind of terminally

differentiated cells (Takahashi and Yamanaka,

2006). We will discuss this aspect with more detail

afterwards.

On the other side, cancer has also been recognized

as a distinct pathological entity since the origins of

mankind. The first references are the Edwin Smith

and Ebers papyri from the 1600 BC and 1500 BC,

approximately (Hajdu, 2004). The Edwin Smith

papyrus contains the first mention and description

of breast cancer, and it concludes that there is no

treatment for the disease. Cancer was not so

common in ancient times, mainly because life span

was much shorter, but it was already clearly

recognized. Hippocrates (460-375 BC) realized that

growing tumors occurred typically in adults and

they reminded him of a moving crab, which led to

the terms carcinos and cancer. Celsus (25 BC-AD

50) also compared cancer with a crab, because it

penetrates the surrounding organs like if it had

claws; Celsus introduced the first classification for

breast cancer and advocated for surgical therapy.

Furthermore, he already realized that tumors could

only be cured if they were removed in their early

stages and that, even after removal and wound

healing, breast carcinomas tended to recur causing

swelling in the armpit and, finally, death by

spreading throughout the body. Galen (131-AD

Page 62: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 238

200) already recommended surgery by cutting a

wide margin of healthy tissue around the edges of

the tumor (Hajdu, 2004). If we jump now to our

days, it seems disappointing to see how little those

old critical findings have been overcome by modern

medicine, 2000 years later. Indeed, still today, clean

surgical margins and lack of lymph node invasion

are the most important prognostic markers for the

successful eradication of solid tumors, and only if

tumors are completely resected before they

metastasize (something that it is anyhow impossible

to determine with current technologies) can

curation be guaranteed. However, in the last thirty

years we have gained an enormous knowledge

about the molecular biology of the disease. In 1979,

it was shown that the phenotype of transformed

cells could be transferred to normal fibroblasts by

DNA transfection (Shih et al., 1979), a finding that

lead to the rapid molecular cloning of the first

human oncogene (the RAS gene), simultaneously by

several groups (Goldfarb et al., 1982; Lane et al.,

1982; Parada et al., 1982; Santos et al., 1982). Since

then, many genes have been described as being

either oncogenes or tumor suppressors, and the

molecular mechanisms of their transforming

capabilities have been analyzed to great detail, in

close relationship with their functions in "normal"

conditions. This is a field that has expanded

tremendously in the last decades, and a

comprehensive study of the topic falls out of the

scope of this revision. However, there are some

aspects that must be taken into account for posterior

debate. A very important one is the fact that, for

many types of tumors, specific genetic mutations

have been shown to correlate closely with the

phenotype of the tumors, suggesting that the

oncogenic alterations might be acting as new

specification factors that determine the tumor

appearance and/or phenotype. This association is

especially evident in the case of mesenchymal

tumors caused by chromosomal aberrations

(Sánchez-García, 1997; Cobaleda et al., 1998). In

2000, Hanahan and Weinberg summarized the main

features that had to be disrupted in normal cellular

behavior in order for allow a tumor to appear and

progress (Hanahan and Weinberg, 2000), and this

list has expanded with the years (Hanahan and

Weinberg, 2011). These main aspects are related

with the survival and proliferation of cancer cells,

but it must be noted that most of them are equally

shared by non-malignant tumors (Lazebnik, 2010).

However, all the aspects related to the alterations of

the normal developmental regulatory mechanisms

in tumorigenesis have received much less attention.

But in fact, if cellular fate was carved into stone,

cancer would be impossible, since no new lineages

could be generated other than the normal,

physiologic ones. Here is where the normal

mechanisms regulating cellular identity and

plasticity play an essential role in allowing cancers

to arise and hopefully, as we will discuss, they

might be the key to its eradication.

The specification of cellular identity during

development and differentiation is a dynamic

process that starts with stem and progenitor cells

and ends with terminal differentiation into each

specialized cellular type. In this progression there

can be many cellular intermediates; some of them

are transient, and some can be long-lasting, but the

maintenance of cellular identity at each stage is

determined by the signals from the environment

and, in an intrinsic manner, by specific transcription

factors and epigenetic modifiers that establish a

defined chromatin architecture and a specific gene

expression profile.

As we have seen, evidences about cellular plasticity

had being accumulating for decades (Hochedlinger

and Jaenisch, 2006; Gurdon and Melton, 2008; Graf

and Enver, 2009; Vicente-Dueñas et al., 2009a), but

the latest findings in the field of reprogramming

have definitively shown how switching to a

different phenotype can be a lot easier than

previously expected, and can have real

physiological relevance, beyond basic research.

Cancer is a perfect example of pathological

reprogramming in which, from a normal tissue, a

whole new differentiation lineage is opened with its

own hierarchy and structure (Reya et al., 2001;

Sánchez-García et al., 2007). So, without forgetting

the so well-studied aberrant proliferation,

reprogramming is an essential part of the

tumorigenesis process, and it is closely dependent

on the cellular plasticity of the cancer-initiating

cells. The term plasticity, as we will use it here,

refers to the ability of cells (stem or differentiated)

to adopt the biological properties (gene expression

profile, phenotype, etc.) of other differentiated

types of cells (belonging to the same or different

lineages). This definition comprises also the

property of competence, i.e. the ability of stem cells

and progenitors to give rise to their different

descendant lineages during normal development.

We use such an ample definition of the term

precisely to reflect the fact that the molecular

mechanisms that are important for progenitors'

competence during normal development are the

same ones responsible for the plasticity changes of

more differentiated types of cells, both in

pathological processes and in experimentally-

induced reprogramming. Here we will discuss the

vital role of cellular plasticity in the origin and

maintenance of tumoral cells. We will first revise

the latest research discoveries in the fields of

normal developmental and experimentally-induced

plasticity, and afterwards will link these findings

with what we know about cancer biology.

Page 63: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 239

Lineage commitment and cellular identity Adult stem cells are the responsible of generating

all the different specialized cellular types forming

the organism. The majority of them perform this

job throughout the whole life of the organism,

thanks to their self-renewal capacity. This property

allows them to divide asymmetrically, therefore

given rise to a new identical daughter stem cell and

to a multipotential progenitor, lacking self-renewal

capacity, which will give rise to all the

differentiated tissue cells. Although it is known that

there are some specific factors that are essential for

the specification and maintenance of stem cell

identity (Boyer et al., 2005), the molecular bases of

the choice that stem cells have to make between

maintaining competence (i.e. plasticity) or entering

into the differentiation programs are not yet

completely understood (Niakan et al., 2010). In this

context, a first important aspect to consider is the

fact that the stem cell population itself is

intrinsically heterogeneous. This means that the

"stemness" is not a static condition defined by

stable, constant levels of expression of intrinsic

stem factors and surface stem markers, but it is

more of a continuum that moves within certain

margins. For example, in a clonal population of

haematopoietic progenitor cells, there is a Gaussian

distribution of the levels of expression of Sca-1,

one of the most classical stem cell markers (Chang

et al., 2008). Furthermore, cells at both the low- or

high-end levels of expression can, when isolated,

regenerate the whole population with all the range

of expression levels. However, every one of these

sub-populations, defined by their levels of a surface

marker, also expresses different transcriptomes, and

has therefore distinct intrinsic differentiation

tendencies towards different lineages. Therefore,

each individual cell in the stem population

represents a metastable transitional point in a

continuum of constantly changing transcriptomes.

In fact, this is most probably the mechanism at the

basis of the stochastic choice of lineage, when some

cells approach too much to the "edges" of the

normal distribution and the transcriptome changes

become irreversible (Chang et al., 2008).

In 1957 Waddington conceptualized the irreversible

process of cellular differentiation as marbles falling

down a slope (Waddington, 1957). This

metaphorical concept has regained new momentum

with the mathematical interpretation of

transcriptional cellular states as Gene Regulatory

Networks (GRNs). In this type of analysis,

pluripotency is represented as a mathematical

attractor (a condition towards which a dynamical

system tends to progress over time), in such a way

that the points (cells) that get close enough to the

attractor remain close even if slightly disturbed.

This attractor is surrounded by a "differentiation

landscape" where other stable cellular fates are

represented by stable "valleys" and differentiation

routes towards them are "channels" through which

the cells move (Enver et al., 2009; Huang, 2009).

Under this light, pluripotency can be considered as

a dynamic state of controlled heterogeneity within a

population, where small individual fluctuations in

the levels of expression of transcription factors and

epigenetic regulators maintain a global status of

apparent stability. The cells that approach the limits

of the attractor (those who, in their random

fluctuations, go too far from the middle point of the

Gaussian curve) are therefore more prone to

differentiate, suggesting that commitment, although

rare, is an spontaneous phenomenon (unless it is

specifically triggered by an external signal that

unbalances the dynamic equilibrium) (Huang,

2009).

Maintenance of cellular identity throughout the differenciation process Although in some rare cases they are unipotent (e.g.

spermatogonial stem cells), adult stem cells are

usually multipotent, and they can give rise to a wide

range of differentiated cell types. In the first

instance, stem cells lose their self-renewal potential

(their stemness) and start the differentiation process

by becoming multipotential progenitors. We have

seen that the differentiation program can be pre-set

already by the oscillatory patterns of gene

expression at the stem cell population level, and

that cells lying at the different ends of specific

gradients of gene expression can have opposite

differentiation preferences (Chang et al., 2008). So,

once they leave the stem cell state, the cells start

making lineage choices that are usually mutually

excluding and are normally conceptualized in a

branching pattern. These alternative options are

usually controlled by the cross-antagonism between

transcription factors with competing, opposing

functions (Swiers et al., 2006; Loose et al., 2007).

A very well characterized developmental system is

hematopoietic differentiation where several models

of lineage-specification have been identified which

seem to be based on the aforementioned

mechanism. For example, the choice between

erythroid/megakaryocyte or myeloid-monocytic

fates at the level of erythromyeloid progenitors is

controlled by the reciprocal inhibition between the

transcription factors GATA-1 and PU.1, therefore

creating a binary decision for the progenitor (Laiosa

et al., 2006; Enver et al., 2009). The bipotent

progenitor itself would therefore be this

intermediate state created and maintained by the

equilibrium between the both factors. This fact

helps understanding the phenomenon of

multilineage gene priming, in which uncommitted

progenitors present low levels of simultaneous

expression of multiple transcription factors

corresponding to different mature cell types and

Page 64: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 240

possessing antagonistic functions (Hu et al., 1997;

Enver et al., 2009). In general, there seems to be a

progressive loss of developmental potential in a

hierarchical process that moves through sequential

differentiation options and in which, at any given

point, a progenitor would only have to choose

between two mutually exclusive options (Brown et

al., 2007; Ceredig et al., 2009). Additionally, in the

process of maturation into a given lineage, the

progenitors will receive (and react to) the necessary

extrinsic signals (for example, cytokines) that,

according to this model, would be more permissive

than instructive.

Maintenance of the cellular identity of mature differentiated cells Plasticity, in normal development, is a property that

is "intended" to be restricted to stem cells and

progenitors. In general, the final differentiated

cellular types of any given organ or tissue possesses

stable identities, in consequence with the fact that

they usually are highly specialized cells with very

specific physiological functions. Therefore, it

would not make sense, from the biological point of

view, that a specialized cell would be the source of

other differentiated cell types. This, as we have

mentioned, is the role of stem cells, with their

physiological plasticity (i.e., normal competence)

that we have previously discussed. However, the

concept of the stability of differentiated cell types

has been shaken by the discovery of the fact that the

4 Yamanaka transcription factors (4Y TFs) Oct4,

Sox2, c-Myc and Klf4 (Takahashi and Yamanaka,

2006) are enough for the reprogramming of most

differentiated cells types into induced pluripotent

stem cells (iPSCs). This finding has altered our

notion of the latent developmental potential hidden

in differentiated cells, showing how it can be

"awakened" by experimental manipulations in the

laboratory. This, as we have described, was already

known to a certain extent from the nuclear

reprogramming experiments performed in

amphibians more than 50 years ago (Briggs and

King, 1952; Gurdon, 1962). Nevertheless, although

those experiments already proved that the cell

nucleus could be reprogrammed from a

differentiated cell type into a pluripotent progenitor,

Yamanaka's experiments showed that only 4 factors

were actually enough to make the whole process

possible. We have seen that, a more modest level, it

had already been proven that the overexpression or

loss of individual transcription factors could induce

fate changes in differentiated cells (MyoD, C/EBPa,

Pax5, etc). Although these were examples of

transdifferentiation taking place between closely

related cell types, they already pointed the way for

the search of the factors capable of reprogramming

to full pluripotency. Since the differentiated state is

the more stable one (indicating that the GRNs are

less subject to fluctuation), where the cells have

reached after "rolling down" the differentiation

pathway in the normal process of development,

therefore an "activation energy" is required to move

the cells "uphill" to become again pluripotent.

Conceptually, there are at least two main possible

scenarios to explain the population dynamics in the

process of reprogramming to pluripotency

(Yamanaka, 2009): one possibility (the so-called

elite model) is that only some cells can be

reprogrammed, and these are the ones that are

selected among the entire population, since they are

the only ones that are receptive to the action of the

reprogramming factors. Alternatively, it might

happen that all the differentiated cells are equally

capable of undergoing reprogramming, and it is

only due to technical or methodological reasons

that we are not able to reveal this potential in all of

them (stochastic model). According to the

accumulating evidences, it would seem that the

stochastic model is the one that is closer to reality

and that, given the right combination of factors; any

cell could be reprogrammed to pluripotency

(Yamanaka, 2009). However, as we have

mentioned, this is a developmentally and

energetically unfavourable process, a fact that is

evidenced by several details. The most obvious one

is the very low efficiency of the reprogramming

process, even in the most favourable conditions.

This fact clearly indicates that, independently on

how many cells of the population are initially

responsive to the reprogramming factors, very few

of them can complete the path towards full

reprogramming (Yamanaka, 2009). Also, this is a

gradual process in which several non-physiological

cellular intermediates can be isolated (Mikkelsen et

al., 2008; Stadtfeld et al., 2008). The study of these

incompletely reprogrammed intermediates has

revealed that they have re-activated the self-renewal

and maintenance stem cell genes, but not yet those

of pluripotency; also, these stages of aborted

reprogramming have not been able to completely

repress the expression of lineage-specific

transcription factors and retain persistent DNA

hypermethylation marks as a proof of their failure

in achieving complete epigenetic remodelling

(Mikkelsen et al., 2008). But perhaps the most

patent proof of the difficulty of the process of full

reprogramming to pluripotency is the persistence of

an epigenetic memory in the iPCs that makes them

more prone to re-differentiate into the lineages from

which they were initially derived, indicating that a

complete elimination of the initial epigenetic

program cannot yet be achieved (Kim et al., 2010;

Bar-Nur et al., 2011).

Page 65: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 241

Tumoral reprogramming and induction of pluripotency: similarities The role of transcription factors in the control of

tumoral reprogramming and induction of

pluripotency

We have seen in the initial section of this review

that both cancer research and developmental

biology have been the focus of intense attention

since ancient times. What's more, they have always

been closely related from the conceptual point of

view. The cellular theory of Rudolf Virchow is

clearly essential for the understanding of both

development and tumorigenenesis. But he went

further, since he already proposed the embryonal

rest hypothesis of tumour origin, after realising the

histological similarities between tumours and

embryonic tissues (Virchow, 1855). This concept

was afterwards expanded by Julius Conheim, who

suggested that tumours arise from residual

embryonic remnants "lost" during normal

development (Cohnheim, 1867). This hypothesis

actually connects with the current theory of the

cancer stem cells (CSCs) in which progenitors are

situated at the root of cancer maintenance (see

below). Another example of the influence of cancer

research in the progress of the fields of stem cell

biology and developmental biology is the fact that

embryonic stem (ES) cells were identified in a

search that has been initiated in the study of

teratocarcinomas (Solter, 2006; Morange, 2007;

Hochedlinger and Plath, 2009).

In the field of cancer research it has traditionally

been postulated that more than one molecular hit is

required to generate a tumour cell, because several

aspects of cellular biology must be altered in the

progress towards a full-blown tumour (Hanahan

and Weinberg, 2000). Therefore, in order to achieve

tumoral reprogramming (although this was not the

terminology traditionally used), more than one

single molecular alteration had to happen. We have

mentioned before that for a "simple"

transformation, like a lineage switch, the change in

the levels of expression of a single transcription

factor could be enough (Davis et al., 1987; Nutt et

al., 1999; Xie et al., 2004; Cobaleda et al., 2007a).

Similarly, a single initial oncogenic lesion may

contribute to just a part of the tumoral phenotype,

by causing a block in differentiation, or an

alteration in the control of cell cycle. In

oncogenesis, many factors and routes have been

shown to be altered, and their individual

contributions to the tumoral phenotype are clear,

although their synergy and interactions are less

known. In the case of reprogramming to

pluripotency, the discovery of Takahashi and

Yamanaka (Takahashi and Yamanaka, 2006)

revealed the nature of these factors. Before,

reprogramming to pluripotency was only possible

by the use of nuclear transplantation, but it was not

known which of the factors present in the zygote

possessed the required reprogramming capacity.

Interestingly enough, the 4 Yamanaka factors are

known to be involved in tumorigenesis in different

contexts, and both c-Myc and Klf4 are well-known

oncogenes (Rowland et al., 2005; Okita et al., 2007;

Tanaka et al., 2007; Chen et al., 2008), thus further

linking reprogramming to tumorigenesis.

In summary, the experimental results show that the

maintenance of cellular identity is essential for

differentiated cells, and that only strong

transcriptional or epigenetic regulators can subvert

it. In this way, the multistep nature of tumorigenesis

is paralleled by reprogramming to pluripotency in

the series of "uphill" steps required and in the need

for the sum of the effects of several factors to

overcome the built-in safety mechanisms designed

to protect cells from transformation or, in other

words, to prevent cells from changing their identity.

In the case of the reprogramming factors, the

precise role of each of them is not yet clear, but

their experimental introduction at different times

during the process of reprogramming is shedding

some light on this issue (Sridharan et al., 2009), by

identifying distinct contributions of the different

factors along the reprogramming progression. In the

early stages of reprogramming, the most important

process happening is the silencing of the gene

expression programs of the differentiated cells. This

aspect is previous to the induction of the ES-like

expression program, and the main molecular

responsible for this function seems to be c-Myc.

However, it has also been shown that treatment

with histone deacetylase inhibitors like valproic

acid (VPA) can substitute for c-Myc, because of

their capacity for repressing the gene expression

programs of differentiated cells (Huangfu et al.,

2008). Therefore, it would seem that the action of c-

Myc takes place mainly before the activation of the

regulators of the pluripotent state and,

consequently, ectopic expression of c-Myc is

required only during the first few days of the

reprogramming process (Sridharan et al., 2009). In

fact, c-Myc is dispensable for reprogramming, but

in its absence there is an enormous drop in the

efficiency of the procedure (Nakagawa et al., 2008;

Wernig et al., 2008). The other three factors, Oct4,

Sox2, and Klf4, need to act together to achieve the

entry into the pluripotent condition, as evidenced by

the fact that, when they are used individually, they

cannot bind their pluripotent target genes in cells

that are sill incompletely reprogrammed, most

likely because the pattern of epigenetic

modifications at these loci is not permissive for

their binding (Sridharan et al., 2009). Indeed, Oct4,

Sox2, and also Nanog co-bind to a plethora of

genes in overlapping genomic sites (Boyer et al.,

2005; Loh et al., 2006), in such a way that the

transcriptional program required for pluripotency is

Page 66: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 242

maintained by the coordinated action of these key

genes.

In general, for the reprogramming of almost every

cell type to pluripotency, the 4 Yamanaka

transcription factors are enough. However, there are

some exceptional cases in which additional

alterations are required. For example, in the case of

mature B cells it is necessary to interfere with the

activity of the transcription factor Pax5, which is

the master regulator of B cell identity (Cobaleda et

al., 2007a; Hanna et al., 2008). Previous

experiments had revealed that the elimination of

Pax5, in the absence of any other genetic

manipulation, allowed mature B cells to

dedifferentiate to early haematopoietic

multipotential progenitors (Cobaleda et al., 2007b).

These findings again correlate reprogramming with

cancer development, since it has also been shown

that the elimination of Pax5 function in mature B

cells induces a process of pathological

dedifferentiation that gives rise to progenitor cell

lymphomas (Cobaleda et al., 2007a). Therefore, the

loss of a transcription factor that is required for the

maintenance of cellular identity can be a tumour-

inducing lesion. However, and contrary to mature B

cells, earlier stages of B cell development can be

reprogrammed to pluripotency in the presence of

functional Pax5, just with the 4 Yamanaka

transcription factors (Hanna et al., 2008), thus

supporting the intuitive idea that the degree of

differentiation of the target cell has an effect on the

final efficiency of reprogramming (see below).

In the genetic landscape, the oncogenic mutations

alter the architecture of the whole gene regulatory

network, since it modifies one of the nodes. This

leads to an alteration in the landscape that gives rise

to new abnormal attractors (new "valleys") where

cancer cells reside (Huang et al., 2009).

Furthermore, this alteration in the landscape gives

the cell a new momentum to move towards new

directions, and this effect can persist even when the

initial stimulus has disappeared. From the point of

view of tumoral reprogramming, this implies that

the expression of a tumour-promoting gene, even if

it is transient, can by itself trigger a durable

malignant phenotype that does not require anymore

of the initial mutation for its maintenance (Huang et

al., 2009).

The role of epigenetic factors in the control of

tumoral reprogramming and induction of

pluripotency

In the previous section we have seen that either the

gain or the loss of function of transcription factors

plays an essential role in reprogramming to

pluripotency, in the same way as how oncogene

overexpression or loss of tumour suppressors

promote tumorigenesis. Also, similarly to tumour

progression, large-scale epigenetic changes are

required for full reprogramming to happen. Today,

it is clearly established that not only genetic

alterations are responsible for cancer development,

but there is also an important role of epigenetic

alterations (Esteller and Herman, 2002; Esteller,

2007; Esteller, 2008) that lead to the specification

of an heritable, abnormal pattern of gene expression

that plays an essential role in cancer initiation and

progression (Ting et al., 2006). All the relevant

epigenetic marks, from DNA methylation to histone

modifications, are perturbed in tumour progression.

The subsequent changes in gene expression patterns

are especially relevant when they affect the levels

of expression of specific oncogenes or tumour

suppressors, but they affect in fact the whole

epigenome, and therefore condition all cellular

identity. All these epigenetic alterations are usually

secondary, and they can be just due to tumour

progression and therefore independent from (i.e.,

not directly caused by) the initiating oncogenic

mutation, but they can also be directly induced by

the first oncogenic event, like it happens when

chromosomal aberrations deregulate histone

modification genes (Esteller, 2008). In the process

of reprogramming to pluripotency, epigenetic

modifications are intrinsically required for the

process to take place, and they have to occur all

throughout the genome, not being just restricted to

the activation or repression of individual genes,

something that is already achieved by the

transcription factors. This explains why the

efficiency of reprogramming is significantly

superior in the presence of chemicals that can

globally interfere with epigenetic marks. For

example, the DNA methyltransferase inhibitor 5-

aza-cytidine (AZA) causes a rapid and stable

transition to a fully reprogrammed iPS state

(Huangfu et al., 2008; Mikkelsen et al., 2008).

Similarly, treatment with valproic acid (VPA), a

histone deacetylase (HDAC) inhibitor, considerably

improves the induction to pluripotency (Huangfu et

al., 2008). Other example is provided by the use of

the compound BIX-01294, an inhibitor of G9a

methyltransferase that makes it possible to achieve

reprogramming to pluripotency using only Oct4 and

Klf4 transcription factors, with an efficiency

comparable to the one obtained when using the four

factors (Shi et al., 2008). In normal development,

the biological role of G9a is to terminate the

pluripotencial state as the progenitors exit to the

differentiation process (Feldman et al., 2006;

Epsztejn-Litman et al., 2008). This is achieved by

its histone methylation activity, that prevents the

reactivation of its target genes (for example

embryonic genes like Oct4) when their

transcriptional repressors are no longer present

(Feldman et al., 2006). Also, at the same time, G9a

promotes DNA methylation, that stops reversion

towards the undifferentiated state (Feldman et al.,

2006; Epsztejn-Litman et al., 2008). Therefore,

genome-wide epigenetic changes affecting many

still unknown loci, are essential in the late stages of

Page 67: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 243

direct reprogramming, and inhibition of the proteins

responsible for generating or maintaining these

marks lowers the "activation energy" required for

the transition to pluripotency. Therefore, it makes

sense that several of the chemical inhibitors that we

have just mentioned are in fact already in use, or in

clinical trials to be used as therapeutic agents

against cancer. AZA was approved by the FDA in

2004 for the treatment of myelodysplastic

syndromes, being the first drug into the new class

of demethylating agents (Kaminskas et al., 2005).

Its mechanism of action is very unspecific, aimed at

the restoration of the normal levels of expression of

genes whose expression has been lost due to

promoter hypermethylation during tumoral

progression, and that might be necessary for the

control of proliferation and differentiation. Like in

the case of most antitumoral drugs, AZA is

expected to affect primarily the tumoral cells and

leave non-proliferative cells unaffected (Sacchi et

al., 1999; Kaminskas et al., 2005). Something

similar happens for HDAC inhibitors (Dey, 2006;

Lane and Chabner, 2009). All these findings

underscore once more the concept of cancer as a

reprogramming disease and a case of wrong

differentiation.

Instructive and permissive factors in the

progression and selection of the processes of

tumoral reprogramming and induction of

pluripotency

We have seen how both genome-wide changes in

epigenetic marks and the loss and/or gain of

transcriptional regulators are essential components

of the processes of tumour generation and

reprogramming to pluripotency. However, it is clear

that these changes are clearly unwanted from the

points of view of normal development and cellular

function. Therefore, cells have developed many

built-in protection mechanisms to maintain their

identity against these transcriptional, genetic and

epigenetic changes. Nevertheless, all these

mechanisms are bypassed, in one way or another

(Hanahan and Weinberg, 2000; Hanahan and

Weinberg, 2011), and cancer appears. How this

happens in "progression to pluripotency" (in

analogy to tumoral progression) is still to be

discovered. However, it has recently been shown by

several groups (Zhao et al., 2008; Banito et al.,

2009; Hong et al., 2009; Kawamura et al., 2009;

Krizhanovsky and Lowe, 2009; Li et al., 2009;

Marión et al., 2009; Utikal et al., 2009) that, exactly

as it happens in cancer progression, the elimination

of the DNA damage control checkpoint (p53-p21)

greatly improves the efficiency of the

reprogramming process, making it possible that

many of the starting cells become successfully

reprogrammed. This is done at the expense of an

increased level of genetic instability, and most of

the iPSCs obtained in the absence of a functional

p53-p21 axis carry genetic aberrations of different

kinds. This is in connection with what we have

mentioned before about reprogramming being an

"uphill", unfavourable process, which most of the

cells fail to complete (Mikkelsen et al., 2008).

Therefore, eliminating the DNA damage checkpoint

diminishes the selection and allows a larger number

of cells to survive until pluripotency. These results

support the idea of cancer as a disease of cellular

differentiation and, furthermore, reinforce the idea

that suggests that the driving forces behind the

tumoral process are aberrantly expressed

transcription factors, epigenetic regulators and

signalling molecules, while the role of many of the

other alterations found in tumours (for example, the

loss of p53) is mainly permissive.

Role of the cell of origin in tumoral reprogramming

and induction of pluripotency

In the study of oncogenesis, it has traditionally been

assumed that the phenotype of the tumour cells was

a reflection of that of the normal cell that gave rise

to the tumour in the first place. There were some

classical examples in which this what not the case

like, for example, chronic myelogenous leukaemia

(CML), where the t(9;22) chromosomal

translocation could be found in most types of

differentiated haematopoietic cells, therefore

indicating that a common, earlier progenitor, should

be the cell of origin (Melo and Barnes, 2007). But,

in general, since most cancerous cells are

reminiscent of some differentiated cell type, for

every type of tumour, the cell of origin was

postulated to be the corresponding normal

differentiated cell. However, the cancer stem cell

(CSC) theory has led to a change in our perspective

(Cobaleda and Sánchez-García, 2009; Vicente-

Dueñas et al., 2009a; Vicente-Dueñas et al., 2009b).

The CSC theory proposes that tumours are stem

cell-based tissues just like any other, and this has

several radical consequences for our understanding

of cancer. The most important one is the fact that

not all the tumoral cells are equally capable of

regenerating the tumour. This means that, when

tumoral cells are experimentally transplanted into a

new host, or when some tumour cells remain in the

patient after incomplete tumour excision, the

reappearance of the tumour is caused by just a

certain tumoral cellular subpopulation. Only those

cells, possessing stem cell characteristics, can give

rise to the whole tumour with all its cellular

heterogeneity. Although there can be a big range of

variability in the percentage of CSCs within a

tumour, from very few to 25% (Quintana et al.,

2008; Cobaleda and Sánchez-García, 2009;

Vicente-Dueñas et al., 2009a; Vicente-Dueñas et

al., 2009b), the fact is that, like in any other stem-

cell based tissue, the majority of cells composing

the tumour mass lack this capacity. Hence, if

tumours are maintained by aberrant cells possessing

stem cell characteristics, then what is the origin of

these cells? This cancer cell-of-origin (not to be

Page 68: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 244

confused with the CSC, which would be the cancer-

maintaining cell of the already developed tumour)

is initially a normal cell (not necessarily a stem cell)

that will be reprogrammed by the oncogenic events

in order to finally originate (or convert into) a

tumoral cell with stem properties. There are two

main mechanisms that could be invoked in this

scenario. One option is that the cell-of-origin

suffering the oncogenic mutation(s) is already a

stem cell, which therefore becomes reprogrammed

to give rise to a new pathological tissue instead of

the normal one. In the case of CML, it has recently

been demonstrated, using genetically modified

mice, that the restricted expression of the oncogenic

alteration in the stem cell/progenitor compartment

is enough to generate a human-like tumour with all

the variety of differentiated tumour cells (Pérez-

Caro et al., 2009; Vicente-Dueñas et al., 2009b). In

mouse models of intestinal cancer it has also been

found that tumours originate in the crypt stem cell,

since when the oncogenic stimulus (activation of

the Wnt signalling pathway) is targeted to the stem

cell compartment, intestinal adenomas develop in

which a developmental hierarchy is maintained. On

the contrary, when the oncogenic lesions are

targeted at the non-stem intestinal epithelial cells,

they only generate short-lived, small

microadenomas (Barker et al., 2008; Zhu et al.,

2008). In the nervous system, targeting

astrocytoma-associated oncogenic lesions to

progenitors (in this case in the subventricular zone)

results in tumour development, while targeting

them to the differentiated cells of the adult

parenchyma does not result in tumours, only in

local astrogliosis (Alcantara Llaguno et al., 2009).

Therefore, there are many examples (Dirks, 2008;

Joseph et al., 2008; Zheng et al., 2008) where it has

been proven that the initiating event takes place in a

normal stem cell, even if the mature tumour is

composed by differentiated cells, indicating a true

tumoral reprogramming mediated by the oncogenic

lesions (Vicente-Dueñas et al., 2009b).

The other alternative is that the cancer cell-of-origin

can be a differentiated cell that regains stem cell

characteristics in the process of tumoral

reprogramming. This option relies on two

requirements: first, the oncogenic alteration must be

capable of conferring or programming these

characteristics in the target cell and, second, the cell

must be plastic enough so as to be reprogrammed

by this precise oncogenic alteration. It has been

shown that some oncogenes, like MOZ-TIF2

(Huntly et al., 2004), MLL-AF9 (Krivtsov et al.,

2006; Somervaille and Cleary, 2006), MLL-ENL

(Cozzio et al., 2003), MLL-GAS (So et al., 2003) or

PML-RARα (Guibal et al., 2009; Wojiski et al.,

2009) can generate CSCs when they are introduced

into committed target cells. Gene expression arrays

have revealed that MLL-AF9 can activate a stem

cell-like program in committed granulocyte-

macrophage progenitors, therefore conferring them

the property of self-renewal (Krivtsov et al., 2006).

Also c-Myc can induce a transcriptional program

reminiscent of that of embryonic stem cells in

differentiated epithelial cells, and originate

epithelial CSCs (Wong et al., 2008). However,

other oncogenes are unable of conferring self-

renewal properties, like for example BCR-

ABLp190 (Huntly et al., 2004). In these cases the

oncogene, since it cannot immediately confer stem

cell properties, could give rise to a precancerous

cell that can afterwards, with the presence of

additional alterations conferring "stemness", give

rise to the cancer stem cell (Chen et al., 2007). In

any case, the cellular origin where the cancer-

initiating lesions take place is difficult to determine

since, in many cases, the functional impact of the

oncogenic lesion (i.e. the tumour clonal expansion)

can present with phenotypes mimicking

differentiation stages that can be either upstream or

downstream of the initiating cell. For example, the

translocations that are the initiating lesions of many

childhood B acute lymphoblastic leukaemias (ALL)

originate in utero during embryonic haematopoiesis

and promote the conversion of partially committed

cells into preleukaemic cells with altered self-

renewal and survival properties, that will require a

second postnatal hit to develop into full leukemias

(Hong et al., 2008). Also, in leukemias carrying the

AML1-ETO translocation, this aberration can be

detected in stem cells in patients in remission.

These stem cells behave apparently normal during

the remission phase, indicating that they can remain

dormant and, with time, some of their descendants

can become tumorigenic and originate the relapse

(Miyamoto et al., 2000). We have described

previously that, in mice, the loss of Pax5 in mature

B cells leads to the dedifferentiation to multipotent

progenitors and the appearance of progenitor B cell

lymphomas (Cobaleda et al., 2007a). In human

Hodking lymphomas, the overexpression of specific

antagonists leads to the functional inactivation of

the B cell factor E2A, which in turn causes the loss

of B cell markers and induces the expression of

lineage-inappropriate genes characteristic of the

Reed-Sternberg Hodking lymphoma cells (Mathas

et al., 2006). Also in children's B-ALLs, the CSCs

can present with the phenotypes of different stages

of early B cell development that, on top of that, can

apparently interconvert among them, therefore

complicating even more the task of identifying the

cancer-cell of origin (le Viseur et al., 2008). A

genomic analysis of samples from relapsed ALL

patients, when compared with the samples at

diagnosis, has shown that the same ancestral clone

can be found at both stages of the disease

(Mullighan et al., 2008). So, clearly in many cases

the cancer-maintaining cell evolves over time and

adapts to treatment to finally lead to relapse, and

therefore the characteristics of the CSC population

Page 69: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 245

in a certain moment may not relate at all any more

to those of the initial cancer cell-of-origin (Barabé

et al., 2007).

As we already mentioned when we described the

view of reprogramming to pluripotency from the

perspective of the GRNs, the inducing factors are

not required anymore once the cells have reached

the pluripotent condition and the new identity

(however plastic this is) has been established. If

cancer stem cells are generated by a tumoral

reprogramming process, then maybe the oncogenes

that initiate tumour formation might be not be

required for tumour progression (Krizhanovsky and

Lowe, 2009). If this were the case, it would explain

the aforementioned examples in which a pre-

cancerous lesion exists stably in an aberrant cell

population that does only evolve to an open tumour

when secondary mutations occur. In this scenario,

the initiating lesion would be the driving force in

the reprogramming process, but once this has been

completed, it would only be a passenger mutation,

or could even perform a different role that would be

independent from its reprogramming capacity, like

for example in tumour expansion/proliferation. A

mechanism of this kind would explain why some

targeted therapies fail in spite of their initial

apparent efficacy: for example, imatinib, a drug

targeted against the deregulated kinase activity of

BCR-ABL, successfully eliminates differentiated

tumour cells, but it fails to kill the BCR-ABL+

CSCs, since it does not seem to interfere with the

function of the chimeric oncogene in this cellular

context (Graham et al., 2002; Barnes and Melo,

2006).

The fact that CSCs can originate from differentiated

cells represents the last and most patent similarity

between tumorigenesis and reprogramming to

pluripotency. Also in iPSCs generation, the nature

of the cell of origin is key in determining the global

success. In this way, it has been described that, in

the haematopoietic system, the capacity of

reprogramming cells decreases as they differentiate,

since HSC are 300 times more likely to be

reprogrammed than B or T cells (Eminli et al.,

2009). In the case of the nervous system, when the

starting cells are adult neural stem cells (NSCs),

then pluripotency can be achieved using only Oct4

(Kim et al., 2009), probably because of the high

similarity of NSCs transcriptional profile to that of

ES cells. Similarly, in a liver model of

transdetermination it has been demonstrated that

Neurogenin3 can convert hepatic progenitor cells

into neo-islets but it cannot transdifferentiate

mature hepatocytes (Yechoor et al., 2009).

Outlook The knowledge obtained in the research of the

molecular and cellular mechanisms that control

cellular plasticity, pluripotency and reprogramming

will also have a profound impact in our

understanding of tumorigenesis and, in a more

distant future, in the treatment of cancer. It is clear

that the two fields of research will continue being

mutually interdependent. By way of example, the

main obstacle for the future use of iPSCs in the

clinic is precisely the generation of tumours as a

result of uncontrolled growth or differentiation of

the cells, once they are in the patient. Therefore, the

knowledge and control of the narrow limits of gene

expression that mark the difference between normal

and tumoral differentiation and reprogramming will

be required before this problem can be overcome.

Assuming the role that reprogramming plays in

cancer generation makes it possible to initiate the

development of new therapeutic strategies aimed at

re-directing the wrong differentiation program

towards a new outcome (ideally, in most cases,

terminal non-tumoral differentiation and cellular

death). Differentiation therapies are already in use

in some cases, like the administration of retinoic

acid to differentiate tumoral cells in PML-RARα+

positive acute promyelocytic leukemias. We have

described how reprogramming to pluripotency, due

to its inefficiency, can get caught up at several

points before reaching the iPSC state (Mikkelsen et

al., 2008). Tumoral cells are probably very close to

these incompletely reprogrammed intermediates,

and the study of the latter should help us in

understanding how to get the former ones out of

their pathologic block. In fact, epigenetic therapies

are most probably going to be on the rise in the

coming years for the treatment of many types of

tumours, since our knowledge about the molecular

mechanisms controlling the epigenetic marks and

their role in self-renewal, differentiation and

maintenance is increasing very quickly, and this

should help us to obtain more and better (more

specific) epigenetic drugs (Jones, 2007; Shen et al.,

2009).

The discovery of reprogramming to pluripotency

has transfigured the research in the field of cellular

plasticity. It is nowadays possible, using just three

ectopic factors, to reprogram fibroblasts into

functional neurons (Vierbuchen et al., 2010), to

convert in vivo pancreatic exocrine cells to β cells

(Zhou et al., 2008) or to directly transdifferentiate

mouse mesoderm into heart tissue (Takeuchi and

Bruneau, 2009). One of the most remarkable

examples in this context is the phenotype caused by

the deletion of a single gene, Foxl2, in adult ovarian

follicles. This inactivation immediately upregulates

testis-specific genes and leads to a full organ

reprogramming (Uhlenhaut et al., 2009) that shows

that the maintenance of the identity of the ovarian

cells requires the active and constant presence of a

specific gene. This is therefore an active process

that resembles very much what we have described

for Pax5 and B cells, but affecting a whole organ

with all its cellular diversity.

Our increasing knowledge and technical control

over cellular identity should help us in the

Page 70: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 246

development of strategies for the reprogramming of

tumoral cells. In fact, several experimental

evidences seem to suggest that this is perfectly

possible. For example, melanoma cells can be

reprogrammed by nuclear transplantation

(Hochedlinger et al., 2004). Also, embryonal

carcinoma cells or mouse brain tumours have been

used as a valid starting material for nuclear cloning

experiments (Li et al., 2003; Blelloch et al., 2004).

Therefore, maybe in a not so distant future we

might have the knowledge and tools to manipulate

tumoral cell identity to force cancer cells to

differentiate, or to make them vulnerable to therapy.

Acknowledgments Research in C.C. lab was partially supported by

FEDER, Fondo de Investigaciones Sanitarias

(PI080164), CSIC P.I.E. 200920I055 and

201120E060, from the ARIMMORA project (FP7-

ENV-2011, European Union Seventh Framework

Program) and from an institutional grant from the

"Fundación Ramón Areces". Research in ISG group

was partially supported by FEDER and by MICINN

(SAF2009-08803 to ISG), by Junta de Castilla y

León (REF. CSI007A11-2 and Proyecto

Biomedicina 2009-2010), by MEC OncoBIO

Consolider-Ingenio 2010 (Ref. CSD2007-0017), by

NIH grant (R01 CA109335-04A1), by Sandra

Ibarra Foundation, by Group of Excellence Grant

(GR15) from Junta de Castilla y Leon, and the

ARIMMORA project (FP7-ENV-2011, European

Union Seventh Framework Program) and by

"Proyecto en Red de Investigación en Celulas

Madre Tumorales en Cancer de Mama", supported

by Obra Social Kutxa y Conserjería de Sanidad de

la Junta de Castilla y León. All Spanish funding is

co-sponsored by the European Union FEDER

program. ISG is an API lab of the EuroSyStem

project. ECS is the recipient of a JAE-predoc

Fellowship from CSIC and a "Residencia de

Estudiantes" Fellowship. The authors declare no

conflict of interest.

References Reaumur RAF.. Sur les diverses reproductions qui se font dans les ecrevisses, les omars, les crabes, etc. et entr'autres sur celles de leurs jambes et de leurs ecailles. Mem Acad Roy. 1712;Sci 223-245.

Trembley A.. Memoires Pour Servir A L'Histoire D'Un Genre de Polypes D'Eau Douce, a Bras En Forme de Cornes. Leiden, Jean and Herman Verbeek. 1744.

Spallanzani L.. Prodromo di un opera da imprimersi sopra la riproduzioni animali (An Essay on Animal Reproduction). London: T. Becket and de Hondt. 1769.

Virchow R.. Editorial. Virchows arch. Pathol Anat Physiol Klin Med. 1855.

Cohnheim J.. Ueber entzundung und eiterung. Path Anat Physiol Klin Med.1867; 40:1-79.

Briggs R, King TJ.. Transplantation of Living Nuclei From Blastula Cells into Enucleated Frogs' Eggs. Proc Natl Acad Sci U S A. 1952 May;38(5):455-63.

Chalkey DT.. A quantitative hystological analysis of forelimb regeneration in Triturus viridescens. J Morphol. 1954;94:21-70.

Waddington CH.. The Strategy of the Genes. London: Allen and Unwin. 1957.

Bodemer CW, Everett NB.. Localization of newly synthesized proteins in regenerating newt limbs as determined by radioautographic localization of injected methionine-S35. Dev Biol. 1959;1:327-342.

Hay ED, Fischman DA.. Origin of the blastema in regenerating limbs of the newt Triturus viridescens. An autoradiographic study using tritiated thymidine to follow cell proliferation and migration. Dev Biol. 1961 Feb;3:26-59.

Gurdon JB.. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J Embryol Exp Morphol. 1962 Dec;10:622-40.

Shih C, Shilo BZ, Goldfarb MP, Dannenberg A, Weinberg RA.. Passage of phenotypes of chemically transformed cells via transfection of DNA and chromatin. Proc Natl Acad Sci U S A. 1979 Nov;76(11):5714-8.

Goldfarb M, Shimizu K, Perucho M, Wigler M.. Isolation and preliminary characterization of a human transforming gene from T24 bladder carcinoma cells. Nature. 1982 Apr 1;296(5856):404-9.

Lane MA, Sainten A, Cooper GM.. Stage-specific transforming genes of human and mouse B- and T-lymphocyte neoplasms. Cell. 1982 Apr;28(4):873-80.

Parada LF, Tabin CJ, Shih C, Weinberg RA.. Human EJ bladder carcinoma oncogene is homologue of Harvey sarcoma virus ras gene. Nature. 1982 Jun 10;297(5866):474-8.

Santos E, Tronick SR, Aaronson SA, Pulciani S, Barbacid M.. T24 human bladder carcinoma oncogene is an activated form of the normal human homologue of BALB- and Harvey-MSV transforming genes. Nature. 1982 Jul 22;298(5872):343-7.

Davis RL, Weintraub H, Lassar AB.. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 1987 Dec 24;51(6):987-1000.

Schneuwly S, Klemenz R, Gehring WJ.. Redesigning the body plan of Drosophila by ectopic expression of the homoeotic gene Antennapedia. Nature. 1987 Feb 26-Mar 4;325(6107):816-8.

Gehring WJ.. The master control gene for morphogenesis and evolution of the eye. Genes Cells. 1996 Jan;1(1):11-5. (REVIEW)

Hu M, Krause D, Greaves M, Sharkis S, Dexter M, Heyworth C, Enver T.. Multilineage gene expression precedes commitment in the hemopoietic system. Genes Dev. 1997 Mar 15;11(6):774-85.

Sanchez-Garcia I.. Consequences of chromosomal abnormalities in tumor development. Annu Rev Genet. 1997;31:429-53. (REVIEW)

Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH.. Viable offspring derived from fetal and adult mammalian cells. Nature. 1997 Feb 27;385(6619):810-3.

Cobaleda C, Perez-Losada J, Sanchez-Garcia I.. Chromosomal abnormalities and tumor development: from genes to therapeutic mechanisms. Bioessays. 1998 Nov;20(11):922-30. (REVIEW)

Nutt SL, Heavey B, Rolink AG, Busslinger M.. Commitment to the B-lymphoid lineage depends on the transcription factor Pax5. Nature. 1999 Oct 7;401(6753):556-62.

Page 71: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 247

Sacchi S, Kantarjian HM, O'Brien S, Cortes J, Rios MB, Giles FJ, Beran M, Koller CA, Keating MJ, Talpaz M.. Chronic myelogenous leukemia in nonlymphoid blastic phase: analysis of the results of first salvage therapy with three different treatment approaches for 162 patients. Cancer. 1999 Dec 15;86(12):2632-41.

Hanahan D, Weinberg RA.. The hallmarks of cancer. Cell. 2000 Jan 7;100(1):57-70. (REVIEW)

Miyamoto T, Weissman IL, Akashi K.. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci U S A. 2000 Jun 20;97(13):7521-6.

Sanchez Alvarado A.. Regeneration in the metazoans: why does it happen? Bioessays. 2000 Jun;22(6):578-90. (REVIEW)

Reya T, Morrison SJ, Clarke MF, Weissman IL.. Stem cells, cancer, and cancer stem cells. Nature. 2001 Nov 1;414(6859):105-11. (REVIEW)

Esteller M, Herman JG.. Cancer as an epigenetic disease: DNA methylation and chromatin alterations in human tumours. J Pathol. 2002 Jan;196(1):1-7. (REVIEW)

Graham SM, Jorgensen HG, Allan E, Pearson C, Alcorn MJ, Richmond L, Holyoake TL.. Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood. 2002 Jan 1;99(1):319-25.

Cozzio A, Passegue E, Ayton PM, Karsunky H, Cleary ML, Weissman IL.. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 2003 Dec 15;17(24):3029-35.

Li L, Connelly MC, Wetmore C, Curran T, Morgan JI.. Mouse embryos cloned from brain tumors. Cancer Res. 2003 Jun 1;63(11):2733-6.

So CW, Karsunky H, Passegue E, Cozzio A, Weissman IL, Cleary ML.. MLL-GAS7 transforms multipotent hematopoietic progenitors and induces mixed lineage leukemias in mice. Cancer Cell. 2003 Feb;3(2):161-71.

Blelloch RH, Hochedlinger K, Yamada Y, Brennan C, Kim M, Mintz B, Chin L, Jaenisch R.. Nuclear cloning of embryonal carcinoma cells. Proc Natl Acad Sci U S A. 2004 Sep 28;101(39):13985-90. Epub 2004 Aug 11.

Hajdu SI.. Greco-Roman thought about cancer. Cancer. 2004 May 15;100(10):2048-51.

Hochedlinger K, Blelloch R, Brennan C, Yamada Y, Kim M, Chin L, Jaenisch R.. Reprogramming of a melanoma genome by nuclear transplantation. Genes Dev. 2004 Aug 1;18(15):1875-85.

Huntly BJ, Shigematsu H, Deguchi K, Lee BH, Mizuno S, Duclos N, Rowan R, Amaral S, Curley D, Williams IR, Akashi K, Gilliland DG.. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cell. 2004 Dec;6(6):587-96.

Odelberg SJ.. Unraveling the molecular basis for regenerative cellular plasticity. PLoS Biol. 2004 Aug;2(8):E232. Epub 2004 Aug 17. (REVIEW)

Xie H, Ye M, Feng R, Graf T.. Stepwise reprogramming of B cells into macrophages. Cell. 2004 May 28;117(5):663-76.

Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, Murray HL, Jenner RG, Gifford DK, Melton DA, Jaenisch R, Young RA.. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005 Sep 23;122(6):947-56.

Kaminskas E, Farrell A, Abraham S, Baird A, Hsieh LS, Lee SL, Leighton JK, Patel H, Rahman A, Sridhara R, Wang YC, Pazdur R; FDA.. Approval summary: azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res. 2005 May 15;11(10):3604-8.

Rowland BD, Bernards R, Peeper DS.. The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol. 2005 Nov;7(11):1074-82.

Barnes DJ, Melo JV.. Primitive, quiescent and difficult to kill: the role of non-proliferating stem cells in chronic myeloid leukemia. Cell Cycle. 2006 Dec;5(24):2862-6. Epub 2006 Dec 15. (REVIEW)

Dey P.. Chromatin remodeling, cancer and chemotherapy. Curr Med Chem. 2006;13(24):2909-19. (REVIEW)

Feldman N, Gerson A, Fang J, Li E, Zhang Y, Shinkai Y, Cedar H, Bergman Y.. G9a-mediated irreversible epigenetic inactivation of Oct-3/4 during early embryogenesis. Nat Cell Biol. 2006 Feb;8(2):188-94. Epub 2006 Jan 15.

Hochedlinger K, Jaenisch R.. Nuclear reprogramming and pluripotency. Nature. 2006 Jun 29;441(7097):1061-7. (REVIEW)

Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, Levine JE, Wang J, Hahn WC, Gilliland DG, Golub TR, Armstrong SA.. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006 Aug 17;442(7104):818-22. Epub 2006 Jul 16.

Laiosa CV, Stadtfeld M, Graf T.. Determinants of lymphoid-myeloid lineage diversification. Annu Rev Immunol. 2006;24:705-38. (REVIEW)

Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, Bourque G, George J, Leong B, Liu J, Wong KY, Sung KW, Lee CW, Zhao XD, Chiu KP, Lipovich L, Kuznetsov VA, Robson P, Stanton LW, Wei CL, Ruan Y, Lim B, Ng HH.. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet. 2006 Apr;38(4):431-40. Epub 2006 Mar 5.

Mathas S, Janz M, Hummel F, Hummel M, Wollert-Wulf B, Lusatis S, Anagnostopoulos I, Lietz A, Sigvardsson M, Jundt F, Johrens K, Bommert K, Stein H, Dorken B.. Intrinsic inhibition of transcription factor E2A by HLH proteins ABF-1 and Id2 mediates reprogramming of neoplastic B cells in Hodgkin lymphoma. Nat Immunol. 2006 Feb;7(2):207-15. Epub 2005 Dec 20.

Solter D.. From teratocarcinomas to embryonic stem cells and beyond: a history of embryonic stem cell research. Nat Rev Genet. 2006 Apr;7(4):319-27.

Somervaille TC, Cleary ML.. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell. 2006 Oct;10(4):257-68.

Swiers G, Patient R, Loose M.. Genetic regulatory networks programming hematopoietic stem cells and erythroid lineage specification. Dev Biol. 2006 Jun 15;294(2):525-40. Epub 2006 Apr 19.

Takahashi K, Yamanaka S.. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006 Aug 25;126(4):663-76. Epub 2006 Aug 10.

Ting AH, McGarvey KM, Baylin SB.. The cancer epigenome--components and functional correlates. Genes Dev. 2006 Dec 1;20(23):3215-31. (REVIEW)

Barabe F, Kennedy JA, Hope KJ, Dick JE.. Modeling the initiation and progression of human acute leukemia in mice. Science. 2007 Apr 27;316(5824):600-4.

Page 72: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 248

Brown G, Hughes PJ, Michell RH, Rolink AG, Ceredig R.. The sequential determination model of hematopoiesis. Trends Immunol. 2007 Oct;28(10):442-8. Epub 2007 Sep 7.

Cobaleda C, Jochum W, Busslinger M.. Conversion of mature B cells into T cells by dedifferentiation to uncommitted progenitors. Nature. 2007a Sep 27;449(7161):473-7. Epub 2007 Sep 12.

Cobaleda C, Schebesta A, Delogu A, Busslinger M.. Pax5: the guardian of B cell identity and function. Nat Immunol. 2007b May;8(5):463-70. (REVIEW)

Chen L, Shen R, Ye Y, Pu XA, Liu X, Duan W, Wen J, Zimmerer J, Wang Y, Liu Y, Lasky LC, Heerema NA, Perrotti D, Ozato K, Kuramochi-Miyagawa S, Nakano T, Yates AJ, Carson WE 3rd, Lin H, Barsky SH, Gao JX.. Precancerous stem cells have the potential for both benign and malignant differentiation. PLoS One. 2007 Mar 14;2(3):e293.

Esteller M.. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet. 2007 Apr;8(4):286-98. Epub 2007 Mar 6. (REVIEW)

Jones RS.. Epigenetics: reversing the 'irreversible'. Nature. 2007 Nov 15;450(7168):357-9.

Loose M, Swiers G, Patient R.. Transcriptional networks regulating hematopoietic cell fate decisions. Curr Opin Hematol. 2007 Jul;14(4):307-14. (REVIEW)

Melo JV, Barnes DJ.. Chronic myeloid leukaemia as a model of disease evolution in human cancer. Nat Rev Cancer. 2007 Jun;7(6):441-53. (REVIEW)

Morange M.. The field of cancer research: an indicator of present transformations in biology. Oncogene. 2007 Dec 6;26(55):7607-10. Epub 2007 Jul 23. (REVIEW)

Okita K, Ichisaka T, Yamanaka S.. Generation of germline-competent induced pluripotent stem cells. Nature. 2007 Jul 19;448(7151):313-7. Epub 2007 Jun 6.

Sanchez-Garcia I, Vicente-Duenas C, Cobaleda C.. The theoretical basis of cancer-stem-cell-based therapeutics of cancer: can it be put into practice? Bioessays. 2007 Dec;29(12):1269-80. (REVIEW)

Tanaka Y, Era T, Nishikawa S, Kawamata S.. Forced expression of Nanog in hematopoietic stem cells results in a gammadeltaT-cell disorder. Blood. 2007 Jul 1;110(1):107-15. Epub 2007 Mar 14.

Birnbaum KD, Sanchez Alvarado A.. Slicing across kingdoms: regeneration in plants and animals. Cell. 2008 Feb 22;132(4):697-710. (REVIEW)

Cobaleda C, Busslinger M.. Developmental plasticity of lymphocytes. Curr Opin Immunol. 2008 Apr;20(2):139-48. Epub 2008 May 9. (REVIEW)

Chang HH, Hemberg M, Barahona M, Ingber DE, Huang S.. Transcriptome-wide noise controls lineage choice in mammalian progenitor cells. Nature. 2008 May 22;453(7194):544-7.

Chen Y, Shi L, Zhang L, Li R, Liang J, Yu W, Sun L, Yang X, Wang Y, Zhang Y, Shang Y.. The molecular mechanism governing the oncogenic potential of SOX2 in breast cancer. J Biol Chem. 2008 Jun 27;283(26):17969-78. Epub 2008 May 2.

Dirks PB.. Cancer's source in the peripheral nervous system. Nat Med. 2008 Apr;14(4):373-5.

Epsztejn-Litman S, Feldman N, Abu-Remaileh M, Shufaro Y, Gerson A, Ueda J, Deplus R, Fuks F, Shinkai Y, Cedar H, Bergman Y.. De novo DNA methylation promoted by G9a prevents reprogramming of embryonically silenced

genes. Nat Struct Mol Biol. 2008 Nov;15(11):1176-83. Epub 2008 Oct 26.

Esteller M.. Epigenetics in cancer. N Engl J Med. 2008 Mar 13;358(11):1148-59. (REVIEW)

Gurdon JB, Melton DA.. Nuclear reprogramming in cells. Science. 2008 Dec 19;322(5909):1811-5. (REVIEW)

Hanna J, Markoulaki S, Schorderet P, Carey BW, Beard C, Wernig M, Creyghton MP, Steine EJ, Cassady JP, Foreman R, Lengner CJ, Dausman JA, Jaenisch R.. Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency. Cell. 2008 Apr 18;133(2):250-64.

Hong D, Gupta R, Ancliff P, Atzberger A, Brown J, Soneji S, Green J, Colman S, Piacibello W, Buckle V, Tsuzuki S, Greaves M, Enver T.. Initiating and cancer-propagating cells in TEL-AML1-associated childhood leukemia. Science. 2008 Jan 18;319(5861):336-9.

Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE, Melton DA.. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol. 2008 Jul;26(7):795-7. Epub 2008 Jun 22.

Joseph NM, Mosher JT, Buchstaller J, Snider P, McKeever PE, Lim M, Conway SJ, Parada LF, Zhu Y, Morrison SJ.. The loss of Nf1 transiently promotes self-renewal but not tumorigenesis by neural crest stem cells. Cancer Cell. 2008 Feb;13(2):129-40.

le Viseur C, Hotfilder M, Bomken S, Wilson K, Rottgers S, Schrauder A, Rosemann A, Irving J, Stam RW, Shultz LD, Harbott J, Jurgens H, Schrappe M, Pieters R, Vormoor J.. In childhood acute lymphoblastic leukemia, blasts at different stages of immunophenotypic maturation have stem cell properties. Cancer Cell. 2008 Jul 8;14(1):47-58.

Mikkelsen TS, Hanna J, Zhang X, Ku M, Wernig M, Schorderet P, Bernstein BE, Jaenisch R, Lander ES, Meissner A.. Dissecting direct reprogramming through integrative genomic analysis. Nature. 2008 Jul 3;454(7200):49-55. Epub 2008 May 28.

Mullighan CG, Phillips LA, Su X, Ma J, Miller CB, Shurtleff SA, Downing JR.. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science. 2008 Nov 28;322(5906):1377-80.

Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi T, Okita K, Mochiduki Y, Takizawa N, Yamanaka S.. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol. 2008 Jan;26(1):101-6. Epub 2007 Nov 30.

Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ.. Efficient tumour formation by single human melanoma cells. Nature. 2008 Dec 4;456(7222):593-8.

Shi Y, Do JT, Desponts C, Hahm HS, Scholer HR, Ding S.. A combined chemical and genetic approach for the generation of induced pluripotent stem cells. Cell Stem Cell. 2008 Jun 5;2(6):525-8.

Stadtfeld M, Maherali N, Breault DT, Hochedlinger K.. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell. 2008 Mar 6;2(3):230-40. Epub 2008 Feb 14.

Wernig M, Meissner A, Cassady JP, Jaenisch R.. c-Myc is dispensable for direct reprogramming of mouse fibroblasts. Cell Stem Cell. 2008 Jan 10;2(1):10-2. Epub 2007 Dec 13.

Wong DJ, Liu H, Ridky TW, Cassarino D, Segal E, Chang HY.. Module map of stem cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell. 2008 Apr 10;2(4):333-44.

Page 73: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 249

Zhao Y, Yin X, Qin H, Zhu F, Liu H, Yang W, Zhang Q, Xiang C, Hou P, Song Z, Liu Y, Yong J, Zhang P, Cai J, Liu M, Li H, Li Y, Qu X, Cui K, Zhang W, Xiang T, Wu Y, Zhao Y, Liu C, Yu C, Yuan K, Lou J, Ding M, Deng H.. Two supporting factors greatly improve the efficiency of human iPSC generation. Cell Stem Cell. 2008 Nov 6;3(5):475-9.

Zheng H, Chang L, Patel N, Yang J, Lowe L, Burns DK, Zhu Y.. Induction of abnormal proliferation by nonmyelinating schwann cells triggers neurofibroma formation. Cancer Cell. 2008 Feb;13(2):117-28.

Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA.. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008 Oct 2;455(7213):627-32. Epub 2008 Aug 27.

Alcantara Llaguno S, Chen J, Kwon CH, Jackson EL, Li Y, Burns DK, Alvarez-Buylla A, Parada LF.. Malignant astrocytomas originate from neural stem/progenitor cells in a somatic tumor suppressor mouse model. Cancer Cell. 2009 Jan 6;15(1):45-56.

Banito A, Rashid ST, Acosta JC, Li S, Pereira CF, Geti I, Pinho S, Silva JC, Azuara V, Walsh M, Vallier L, Gil J.. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 2009 Sep 15;23(18):2134-9. Epub 2009 Aug 20.

Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M, Danenberg E, Clarke AR, Sansom OJ, Clevers H.. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 2009 Jan 29;457(7229):608-11. Epub 2008 Dec 17.

Ceredig R, Rolink AG, Brown G.. Models of haematopoiesis: seeing the wood for the trees. Nat Rev Immunol. 2009 Apr;9(4):293-300.

Cobaleda C, Sanchez-Garcia I.. B-cell acute lymphoblastic leukaemia: towards understanding its cellular origin. Bioessays. 2009 Jun;31(6):600-9. (REVIEW)

Eminli S, Foudi A, Stadtfeld M, Maherali N, Ahfeldt T, Mostoslavsky G, Hock H, Hochedlinger K.. Differentiation stage determines potential of hematopoietic cells for reprogramming into induced pluripotent stem cells. Nat Genet. 2009 Sep;41(9):968-76. Epub 2009 Aug 9.

Enver T, Pera M, Peterson C, Andrews PW.. Stem cell states, fates, and the rules of attraction. Cell Stem Cell. 2009 May 8;4(5):387-97. (REVIEW)

Graf T, Enver T.. Forcing cells to change lineages. Nature. 2009 Dec 3;462(7273):587-94. (REVIEW)

Guibal FC, Alberich-Jorda M, Hirai H, Ebralidze A, Levantini E, Di Ruscio A, Zhang P, Santana-Lemos BA, Neuberg D, Wagers AJ, Rego EM, Tenen DG.. Identification of a myeloid committed progenitor as the cancer-initiating cell in acute promyelocytic leukemia. Blood. 2009 Dec 24;114(27):5415-25. Epub 2009 Oct 1.

Hochedlinger K, Plath K.. Epigenetic reprogramming and induced pluripotency. Development. 2009 Feb;136(4):509-23. (REVIEW)

Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa M, Okita K, Yamanaka S.. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature. 2009 Aug 27;460(7259):1132-5. Epub 2009 Aug 9.

Huang S.. Reprogramming cell fates: reconciling rarity with robustness. Bioessays. 2009 May;31(5):546-60.

Huang S, Ernberg I, Kauffman S.. Cancer attractors: a systems view of tumors from a gene network dynamics and developmental perspective. Semin Cell Dev Biol. 2009 Sep;20(7):869-76. Epub 2009 Jul 10. (REVIEW)

Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, Raya A, Wahl GM, Belmonte JC.. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature. 2009 Aug 27;460(7259):1140-4. Epub 2009 Aug 9.

Kim JB, Sebastiano V, Wu G, Arauzo-Bravo MJ, Sasse P, Gentile L, Ko K, Ruau D, Ehrich M, van den Boom D, Meyer J, Hubner K, Bernemann C, Ortmeier C, Zenke M, Fleischmann BK, Zaehres H, Scholer HR.. Oct4-induced pluripotency in adult neural stem cells. Cell. 2009 Feb 6;136(3):411-9.

Kragl M, Knapp D, Nacu E, Khattak S, Maden M, Epperlein HH, Tanaka EM.. Cells keep a memory of their tissue origin during axolotl limb regeneration. Nature. 2009 Jul 2;460(7251):60-5.

Krizhanovsky V, Lowe SW.. Stem cells: The promises and perils of p53. Nature. 2009 Aug 27;460(7259):1085-6.

Lane AA, Chabner BA.. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol. 2009 Nov 10;27(32):5459-68. Epub 2009 Oct 13. (REVIEW)

Li H, Collado M, Villasante A, Strati K, Ortega S, Canamero M, Blasco MA, Serrano M.. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature. 2009 Aug 27;460(7259):1136-9. Epub 2009 Aug 9.

Marion RM, Strati K, Li H, Murga M, Blanco R, Ortega S, Fernandez-Capetillo O, Serrano M, Blasco MA.. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature. 2009 Aug 27;460(7259):1149-53. Epub 2009 Aug 9.

Perez-Caro M, Cobaleda C, Gonzalez-Herrero I, Vicente-Duenas C, Bermejo-Rodriguez C, Sanchez-Beato M, Orfao A, Pintado B, Flores T, Sanchez-Martin M, Jimenez R, Piris MA, Sanchez-Garcia I.. Cancer induction by restriction of oncogene expression to the stem cell compartment. EMBO J. 2009 Jan 7;28(1):8-20. Epub 2008 Nov 27.

Shen X, Kim W, Fujiwara Y, Simon MD, Liu Y, Mysliwiec MR, Yuan GC, Lee Y, Orkin SH.. Jumonji modulates polycomb activity and self-renewal versus differentiation of stem cells. Cell. 2009 Dec 24;139(7):1303-14.

Sridharan R, Tchieu J, Mason MJ, Yachechko R, Kuoy E, Horvath S, Zhou Q, Plath K.. Role of the murine reprogramming factors in the induction of pluripotency. Cell. 2009 Jan 23;136(2):364-77.

Takeuchi JK, Bruneau BG.. Directed transdifferentiation of mouse mesoderm to heart tissue by defined factors. Nature. 2009 Jun 4;459(7247):708-11. Epub 2009 Apr 26.

Uhlenhaut NH, Jakob S, Anlag K, Eisenberger T, Sekido R, Kress J, Treier AC, Klugmann C, Klasen C, Holter NI, Riethmacher D, Schutz G, Cooney AJ, Lovell-Badge R, Treier M.. Somatic sex reprogramming of adult ovaries to testes by FOXL2 ablation. Cell. 2009 Dec 11;139(6):1130-42.

Utikal J, Polo JM, Stadtfeld M, Maherali N, Kulalert W, Walsh RM, Khalil A, Rheinwald JG, Hochedlinger K.. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature. 2009 Aug 27;460(7259):1145-8. Epub 2009 Aug 9.

Vicente-Duenas C, Gutierrez de Diego J, Rodriguez FD, Jimenez R, Cobaleda C.. The role of cellular plasticity in cancer development. Curr Med Chem. 2009a;16(28):3676-85. (REVIEW)

Vicente-Duenas C, Perez-Caro M, Abollo-Jimenez F, Cobaleda C, Sanchez-Garcia I.. Stem-cell driven cancer: "hands-off" regulation of cancer development. Cell Cycle. 2009b May 1;8(9):1314-8. Epub 2009 May 18.

Page 74: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Plasticity and Tumorigenicity Campos-Sanchez E, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 250

Wojiski S, Guibal FC, Kindler T, Lee BH, Jesneck JL, Fabian A, Tenen DG, Gilliland DG.. PML-RARalpha initiates leukemia by conferring properties of self-renewal to committed promyelocytic progenitors. Leukemia. 2009 Aug;23(8):1462-71. Epub 2009 Mar 26.

Yamanaka S.. Elite and stochastic models for induced pluripotent stem cell generation. Nature. 2009 Jul 2;460(7251):49-52.

Yechoor V, Liu V, Espiritu C, Paul A, Oka K, Kojima H, Chan L.. Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes. Dev Cell. 2009 Mar;16(3):358-73.

Zhu L, Gibson P, Currle DS, Tong Y, Richardson RJ, Bayazitov IT, Poppleton H, Zakharenko S, Ellison DW, Gilbertson RJ.. Prominin 1 marks intestinal stem cells that are susceptible to neoplastic transformation. Nature. 2009 Jan 29;457(7229):603-7. Epub 2008 Dec 17.

Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, Kim J, Aryee MJ, Ji H, Ehrlich LI, Yabuuchi A, Takeuchi A, Cunniff KC, Hongguang H, McKinney-Freeman S, Naveiras O, Yoon TJ, Irizarry RA, Jung N, Seita J, Hanna J, Murakami P, Jaenisch R, Weissleder R, Orkin SH, Weissman IL, Feinberg AP, Daley GQ.. Epigenetic memory in induced pluripotent stem cells. Nature. 2010 Sep 16;467(7313):285-90.

Lazebnik Y.. What are the hallmarks of cancer? Nat Rev Cancer. 2010 Apr;10(4):232-3.

Niakan KK, Ji H, Maehr R, Vokes SA, Rodolfa KT, Sherwood RI, Yamaki M, Dimos JT, Chen AE, Melton DA, McMahon AP, Eggan K.. Sox17 promotes differentiation in mouse embryonic stem cells by directly regulating extraembryonic gene expression and indirectly antagonizing self-renewal. Genes Dev. 2010 Feb 1;24(3):312-26.

Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Sudhof TC, Wernig M.. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010 Feb 25;463(7284):1035-41. Epub 2010 Jan 27.

Bar-Nur O, Russ HA, Efrat S, Benvenisty N.. Epigenetic memory and preferential lineage-specific differentiation in induced pluripotent stem cells derived from human pancreatic islet Beta cells. Cell Stem Cell. 2011 Jul 8;9(1):17-23. doi: 10.1016/j.stem.2011.06.007.

Hanahan D, Weinberg RA.. Hallmarks of cancer: the next generation. Cell. 2011 Mar 4;144(5):646-74. (REVIEW)

Rinkevich Y, Lindau P, Ueno H, Longaker MT, Weissman IL.. Germ-layer and lineage-restricted stem/progenitors regenerate the mouse digit tip. Nature. 2011 Aug 24;476(7361):409-13. doi: 10.1038/nature10346.

This article should be referenced as such:

Campos-Sanchez E, Sanchez-Garcia I, Cobaleda C. Plasticity and Tumorigenicity. Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):236-250.

Page 75: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Deep Insight Section

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 251

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function Xiaodong Lu, Wenxin Qin

School of Medical Science and Laboratory Medicine, Jiangsu University, China (XL), State Key

Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai

Jiao Tong University School of Medicine, China (WQ)

Published in Atlas Database: September 2011

Online updated version : http://AtlasGeneticsOncology.org/Deep/V-ATPaseInCancerID20104.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI V-ATPaseInCancerID20104.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Vacuolar H+-ATPase (V-ATPase) is a highly

evolutionarily conserved enzyme, which is

distributed within the plasma membranes and the

membranes of some organelles such as endosome,

lysosome and secretory vesicle. The mayor function

of V-ATPase is to pump protons across the cell

membrane to extracellular milieu or across the

organelle membrane to intracellular compartments.

V-ATPases located in cell surface act as important

proton transporters that regulate the cytosolic pH to

~7.0 which is essential for most physiological

processes, whereas V-ATPases within intracellular

membrane are involved in cellular processes as

receptor-mediated endocytosis, membrane

trafficking, protein processing or degradation, and

nutrients uptake (Nishi et al., 2002; Forgac et al.,

2007; Toei et al., 2010; Cruciat et al., 2010).

Malfunctioned V-ATPase is closely related to

several diseases including tumor. More and more

evidences indicate that V-ATPase is an enhancer

for carcinogenesis and cancer progression, such as

malignant transformation, growth and proliferation,

invasion and metastasis, acquirement of multi-drug

resistance, etc., which strongly supports that V-

ATPase should be an effective target of anticancer

strategy (Fais et al., 2007).

The structure of V-ATPases and its expression in tumor cells The molecular structure of normal V-ATPase of

yeast and mammalian cells has been well studied.

V-ATPase is a delicate complex which is composed

of a cytosolic catalytic domain V1 and an integral

domain V0, the former responsible for ATP

hydrolysis and the latter providing

transmembraneous proton channel (Nishi et al.,

2002; Yokoyama et al., 2005; Wang al., 2007). The

core of the V1 section is composed of a hexameric

arrangement of alternating A and B subunits, which

participate in ATP binding and hydrolysis. Other

subunits of V1 include three copies of E and G

subunits which are the stator, one copy of the

regulatory C and H subunits, one copy of subunits

D and F which form a central rotor axle. The V0

section includes a ring of proteolipid subunits (c, c'

and c") that are adjacent to subunits a and e.

Subunits D and F of V1 and subunit a of V0 form

the central stalk, whereas the multiple peripheral

stalks are composed of subunits C, E, G, H and the

N-terminal domain of subunit a. V1 and V0 is

connected by both stalks. Several subunits like a, d,

e, C, G, H, D and F contain slice variants as to

spatial and temporal expression pattern in different

cell types (Forgac et al., 2007; Miranda et al.,

2010). As for tumor cells, especially those with

high metastatic potential, the V-ATPases are

usually excessively agitated. The altered structures

of V-ATPase of tumor cells may include the

increased level of subunit expressions and unique

spliced variants of some subunits.

The level of the subunit c expression was found to

be related to the metastasis potentials in tumors.

One of the studies is the comparison of subunit c

expression between normal and pancreatic

carcinoma tissues and between invasive and non-

invasive pancreatic cancers, which

immunohistochemical data showed the notable

difference - 92% invasive ductal cancers (42/46)

Page 76: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function

Lu X, Qin W

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 252

were mild to marked subunit c positive in the

cytoplasm, whereas neither non-invasive ductal

cancers nor benign cystic neoplasms expressed

detectable immunoreactive proteins (Ohta et al.,

1996). Subunit c seems to be one of the V-ATPase

subunit which significantly influence the

proliferation and metastasis of tumor cells. The

inhibition of the V-ATPase subunit c via siRNA

resulted in the suppression of growth and metastasis

of a hepatocellular carcinoma cell line in vitro and

in mice model (Lu et al., 2005), which is according

to another result of the suppression of subunit c in

Hela cell via antisense oligonucleotides (Zhan et al.,

2003). But in oral squamous cell carcinoma cells,

subunit C1 was the most strongly over-expressed

gene at the mRNA level compared to other genes of

the V-ATPase complex (Otero-Rey et al., 2008).

Specific spliced variants of subunit have been

observed in tumors. A study of expression of

subunit a of V-ATPase in breast cancer cell lines

displayed the metastasis-specific subunit a isoform

expression profile. In highly metastatic breast

cancer cell line compared with its lowly metastatic

parallel, levels of a3 and a4 were much higher

although all the four a isoforms - a1-4 can be

detectable. They distribute differently, and

especially, a4-containing v-ATPases were located

mainly in the plasma membrane of higher

metastatic breast cancer cell, seeming to be

involved in the formation of the leading surface of

the cells due to the combination with F-actin and

closely correlated to the potency of invasion. a3-

containing V-ATPases were located in intracellular

compartment membrane, which regulated the pH of

the cytosol and intracellular compartments and also

involved in invasion (Hinton et al., 2009). In

accordance with this data, the strongly expressed a3

isoform were observed in high-metastatic

melanoma cells and in bone metastases (Nishisho et

al., 2011). Other tumor-relevant spliced variants are

yet to be found.

The roles of the v-ATPase in the growth, proliferation or apoptosis in tumor cells One of cancer hallmarks is the shift in energy

production from oxidative phosphorylation to

aerobic glycolysis, ie "Warburg effect", which

produces excess intracellular acidosis (Gillies et al.,

2008). However, cancer cells usually have neutral

to alkaline intracellular pH in the acidized

extracellular microenvironment. The V-ATPase is

among the four major types of pH regulators (the

other three are: Na+/H+ exchangers, bicarbonate

transporters, proton/lactate symporters). Much data

implies proton pump is essential in tumors and cells

seem to render V-ATPases more than any other

three transporters to regulate pH in cytosol (Torigoe

et al., 2002). The ability to extrude intracellular

protons and maintain the cytosol pH is critical for

cancer cell survival from a cascade of self-digestion

triggered by acidosis.

The inhibition of v-ATPase may induce apoptotic

cell death in several human cancer cell lines

including pancreatic cancer (Ohta et al., 1998;

Hayash et al., 2006), liver cancer (Morimura et al.,

2008), gastric cancer (Nakashima et al., 2003), B-

cell hybridoma cells (Nishihara et al., 1995; De

Milito et al., 2007) and breast cancer (McHenry et

al., 2010).The deficiency of V-ATPase will

decrease cytosol pH and increased lysosome pH,

both of which might influence lysosome function.

The apoptosis induced by V-ATPase inhibitors

were in either lysosome-mediated or non-lysosome-

mediated manner. In the first case, when lysosomal

V-ATPase was defected, lysosomal pH and

permeability will be increased, resulted in the

release of cathepsin D and activation of caspase,

with no significant impact on mitochondrial

transmembrane potential (Nakashima et al., 2003).

In the other case, mitochondria and lysosome might

be together involved in V-ATPase-inhibitor-

induced apoptosis via capsase pathway or ROS-

dependant manner (Ishisaki et al., 1999; De Milito

et al., 2007). The inhibition of V-ATPase could also

induce apoptosis by suppressing anti-apoptotic Bcl-

2 or Bcl-xL and facilitate the caspase-independent

apoptotic pathway (Sasazawa et al., 2009). In order

to survive from the apoptosis induced by acidosis

resulted from glycolysis, tumor cells needs to

extrude excessive acid, in which processes V-

ATPase plays a crucial role. It is reasonable to

postulate that the inhibition of proton extrusion may

be more susceptible or vulnerable to cell death of

cancer cells than normal cells.

Moreover, the slightly alkalized cytosolic pH favors

the growth and proliferation of the cells. Some

glycolysis-related enzymes or oncogenes are

sensitive the narrow range of pH alteration.

Alkalization of cytosol, which mainly regulated by

V-ATPase in tumor cells, could activate glycolysis

whereas repress oxidative phosphorylation,

meanwhile also promote the transcription of

oncogenes like HIF-1, akt, myc, ras, etc (Gillies et

al., 2008; López-Lázaro, 2008). The cytosol pH of

tumor cells was found to be higher than in

untransformed controls (Busa et al.,1984; Casey et

al., 2010) and increasing cytosol pH was sufficient

to confer tumourigenicity to cultured fibroblasts

(Perona et al., 1988). On the contrast, p53, the

important tumor suppressor could be inactivated in

the condition of alkalization (Xiao et al., 2003). It is

much likely that the glucose metabolism shift and

mutant V-ATPase may be the co-selectors in

selecting those "adaptive phenotype", which may

take the advantages for survival and proliferation

during the initial stage of carcinogenesis.

Page 77: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function

Lu X, Qin W

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 253

The functions of the v-ATPase in cellular signals processing V-ATPase is the important factor that regulates the

process of internization and activation of cellular

signals. It is mainly due that the V-ATPase is the

main contributor of low intracellular vesicles pH,

which is essential for various membrane traffic

processes. V-ATPase activity influence endocytosis

and degradation of molecule-receptor complex,

recycling of the released receptor, recruitment of

signal molecules, and their proper spatial

intracellular distributions (Hurtado-Lorenzo et al.,

2006; Marshansky et al., 2008), therefore exerts a

profound effect on cell behavior such as growth,

proliferation or metastasis via the modulated signals

and their pathways. It has been reported that tumor-

associated m-TOR (mammalian target of

rapamycin) (O'Callaghan et al., 2009), Notch

(Fortini and Bilder, 2009; Vaccari et al., 2010) or

Wnt (Cruciat et al., 2010; Buechling et al., 2010)

could be regulated by V-ATPase.

Early endosomes are important sites for signal

molecules internalization and activation in

mammalian cells. Studies of the effects of V-

ATPases inhibitors on isolated rat hepatocytes and

rat sinusoidal endothelial cells suggested that the

pH gradient between the endocytic compartments

and the cytoplasm was necessary for the receptor-

mediated endocytosis (Harada et al., 1996; Harada

et al., 1997). Inhibition of V-ATPases can retard

recycling of transferrin receptor (Presley et al.,

1997), impair the formation of endosomal carrier

vesicle (Clague et al., 1994), and inhibit late

endosome-lysosome fusion (van Weert et al.,

1995). Although the significance of active V-

ATPase in signal molecules endocytosis and

processing on the behavior of tumor cells is not yet

full elucidated for most data was gained from yeast

or normal mammalian cells, it could be

hypothesized that V-ATPase might regulate some

signal pathways via modulating the recycling rate

of receptor, which would be responsible for the

sensitivity of tumor cells to some signal molecules,

ie, the faster rate at which the receptor cycling in a

V-ATPase-regulated membrane trafficking, the

more efficiently the cells render the receptors, the

more signal molecules could be recruited, and the

stronger or more lasting response to the stimulation

by the signal molecules could be expected.

For example, the activation of Notch, a common

hallmark of an increasing number of cancers (Miele

et al., 2006; Roy et al., 2007), is involved in V-

ATPase-associated endosomal system (Yan et al.,

2009; Vaccari et al., 2010). V-ATPase activity is

required for Notch signaling. In V-ATPase mutant

cells, Notch and its receptors are trapped in an

expanded lysosome-like compartment, where they

accumulate rather than being degraded and a

substantial reduction expression in downstream

gene of notch. V-ATPase regulates Notch via: i)

endocytosis of Notch, for acidification of earlier

endosomal compartments is required in this process

and a reduced rate of Notch endocytosis was found

in V-ATPase mutant cells ii) endosomal cleavage

patterns of the protease that degrade the Notch in

the accordingly forms, each of which process

exerting its own activating potency (Vaccari et al.,

2010) iii) regulating endosome-lysosome fusion

and Notch intracellular re-distribution or the

targeting to cell surface.

The V-ATPase-associated signal molecules

processing itself may also be regulated by

endosomal protein, for example, HRG-1(heme-

regulated genes), a downstream gene of IGF-I

(insulin-like growth factor) and having an

interaction with subunit c. HRG-1 could promote

endosomal acidification and receptor trafficking,

enhance the proliferative and invasive phenotype of

cancer cells. It was implied that the increased active

V-ATPase by HRG-1 not only regulate the

endocytosis and degradation of receptors that

promote signaling for survival, growth, and

migration of cancer tumor, but also facilitate

micronutrient uptake necessary for tumor cellular

metabolism (O'Callaghan et al., 2009).

The contributions of the V-ATPase in cancer metastasis Invasion and metastasis is the relatively late event

of development of malignant cells, which is the

continuous process of breaking through the

basement membrane, degrading extracellular

matrix, angiogenesis, invading vascular system and

redistributing in the distinct host sites. The

activation of the proteases which break down

extracellular matrix is required during the

procedure. The invasive phenotype is closely

related to its highly active V-ATPase. It has been

reported that the improper activated V-ATPases

correlates with an invasive phenotype of several

types of tumors, including breast cancer (Sennoune

et al., 2004; Hinton et al., 2009), pancreatic cancer

(Chung et al., 2011) and melanoma (Nishisho et al.,

2011). The tumor metastasis can be suppressed in

vitro or in animal model by the inhibition of V-

ATPase inhibitors or siRNA (Lu et al., 2005;

Hinton et al., 2009; Supino et al., 2008). Subunit a

isoform and c seem to be important factors in

regulating the metastasis of cancer.

The main mechanisms by which overly active V-

ATPases enhance the tumor invasion and metastasis

may be that the extracellular milieu is acidized and

it is suitable for optimal pH of proteases that

degenerate extracellular matrix (ECM). The plasma

membrane V-ATPases is responsible for pumping

cytosol protons to the extracellular space resulting

in a low extracelluar pH, which is required for the

activation of several types of proteases including

cathepsins, metalloproteases, and gelatinases. V-

ATPase may influence the expression of proteases

Page 78: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function

Lu X, Qin W

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 254

directly independent of the whole enzyme V-

ATPase function. For example, transfectants which

over express V-ATPase subunit c at the mRNA

level showed an enhance invasiveness in vitro with

a concomitant increases in secretion of matrix

metalloproteinase-2 (Kubota et al., 2000). V-

ATPase may also regulate metastasis by enhancing

proteases activation. Cathepsin is an example,

which is secreted by several types of tumor cells

and related to invasion. Once the extracellular

cathepsin is activated, it can both degrade

extracellular matrix proteins and activate other

secreted proteases involved in invasion, such as

matrix metalloprotease (Joyce et al., 2004; Gocheva

et al., 2007) and gelatinases (Martínez-Zaguilá et

al., 1996). The plasma membrane V-ATPase

appeared to be recruited at the proceeding edge of

the cancer cell by the interaction with F-actin so as

to give rise an acidic microenvironment by the edge

(Hinton et al., 2009). Moreover, intracellular V-

ATPases, the major contributor of acidity of

intracellular compartment and membrane

trafficking regulator, also facilitate in the invasion

and metastasis, which is due to possible modulating

proteolytic activation of cathepsins or matrix

metalloproteases within lysosomes or secretory

vesicles and targeting the proteases-containing

secretory vesicles to the cell surface to be

extracytosed (Hinton et al., 2009). The

accumulation of acidity, concentration of plasma

membrane V-ATPase and activated protease crown

the proceeding surface of a metastatic cell,

conferring the tumor cell a "cutting edge".

Mobility is crucial for spread of tumor cells to the

distant sites. NiK-12192, one of V-ATPase

inhibitor was shown able to reduce the

migration/invasion of human lung cancer cells in

vitro and significantly reduce the number of

spontaneous metastases in the lung of nude mice

implanted with a human lung carcinoma. After the

treatment of NiK-12192, the lung cancer cells in

vitro showed that actin fibers were broken, spots of

aggregation were evident and no pseudopodia and

regular structure for actin filaments could be seen,

comparing to the control cells with long and regular

fibers of tubulin in the cell cytoplasm and filaments

of actin forming pseudopodia. NiK-12192-treated

cells also demonstrate a reduction in the experiment

of wound healing assay due to the retard of

migration (Supino et al., 2008). V-ATPase subunit

B and C appear to contain the binding sites to the

actin cytoskeleton (Vitavska et al., 2003; Vitavska

et al., 2005; Zuo et al., 2006). The interactions

between V-ATPase and cytoskeleton implicate their

involvement and regulation of cell mobility and

membrane trafficking (Sun-Wada et al., 2009).

Angiogenesis, a consequence of the mutual

interaction between cancer cells and the stoma cells

of extracellular microenvironments, is another

important step during metastasis, during which

process, endothelial cells is mainly involved. It was

documented that V- ATPases play a crucial role in

growth and phenotypic modulation of

myofibroblasts that contribute to neointimal

formation in cultured human saphenous vein (Otani

et al., 2000) The microvascular endothelial cells in

tumor tissue also incline to render plasma

membrane V-ATPase to cope with the acidic

extracellular environment. The ability of migration

of endothelial cell toward the adjacent tissue is

required during angiogenesis, in which process V-

ATPase plays a role, shown in the result that the

penetration of basement membrane of endothelial

cell was suppressed by bafilomycin treatment

(Rojas et al., 2006).

The relations of V-ATPase and drug resistance in cancer Acquired multidrug resistance (MDR) can limit

therapeutic potential and one of the reasons of

relapse. It is well known that MDR is correlate to

the evolutionarily conserved family of the ATP

binding cassette (ABC) proteins pg, yet it is

documented that V-ATPase plays a role in MDR in

a pg-independent manner, and the inhibition of V-

ATPase could not only suppress tumor cells

directly, but also sensitize the tumor cells to the

chemical therapy (De Milito et al., 2005). It was

documented that proton pump inhibitor (PPI)

pretreatment sensitized tumor cell lines to the

effects of cisplatin, 5-fluorouracil, and vinblastine

significantly. PPI treatment will increases both

extracellular pH and the pH of lysosomal

organelles, which induced a marked increase in the

cytoplasmic retention of the cytotoxic drugs, with

clear targeting to the nucleus in the case of

doxorubicin. In vivo experiments, oral pretreatment

with omeprazole was able to induce sensitivity of

human solid tumors to cisplatin (Lucian et al.,

2004).

V-ATPase renders several mechanisms of

multidrug resistance including: neutralized drug

extracellularly or intracellularly, decreased drug

internalization, altered DNA repair and inhibition of

apoptosis. The pH of the tumor microenvironment

may influence the uptake of anticancer drugs.

Molecules diffuse passively across the cell

membrane most efficiently in the uncharged form.

Because the extracellular pH in tumors is low and

the intracellular pH of tumor cells is neutral to

alkaline, weakly basic drugs that have an acid

dissociation constant of 7.5-9.5, such as

doxorubicin, mitoxantrone, vincristine, and

vinblastine, are protonated and display decreased

cellular uptake (Raghunand et al., 1999; Gerweck et

al., 2006; McCarty and Whitaker, 2010). The data

in vitro or in animal models indicates that

extracellular alkalinization leads to substantial

improvement in the therapeutic effectiveness of

antitumor drugs via enhanced the cellular drug

Page 79: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function

Lu X, Qin W

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 255

The roles of V-ATPase in cancer cells. 1) Protons produced by glycolysis are pumped by plasma membrane V-ATPase (green

circle: V0; blue circle: V1) which prevents the cell from acidosis-induced apoptosis and the slightly basic of cytosolic pH enhanced cell growth and proliferation; 2) Acidification of secretary vesicle, which is maintained by intracellular V-ATPase, is essential for protease secretion and activation (orange bars: active form; orange-red bars: inactive forms of protease). The

interaction between V-ATPase and actin (green wave line) may contribute the recruitment of V-ATPase on plasma membrane. The accumulation of V-ATPase on the plasma membrane, the extracellular acidic-microenvironment and activated-protease

appear to crown the tumor cell, conferring it a "cutting edge" at the proceeding surface which facilitates invasion and metastasis. Moreover, in acidic microenvironment, angiogenesis is enhanced; 3) V-ATPases might regulate signal pathway via controlling

international of signal molecules (red circle), releasing and recycling the receptors, and processing signal molecules. Therefore, V-ATPases may exert effects on cell behavior via signal pathway; 4) V-ATPases contributes to acquirement of resistance of

anticancer drug (green square) supported by the data that inhibition of V-ATPase sensitize the tumor cells to chemical therapy, which is partly due to the increased influx of anticancer drug when in a basic extracellular condition.

uptake and cytotoxicity (Gerweck et al., 2006;

Trédan et al., 2007).The reduced intracellular

accumulation of anticancer drugs may also be due

that V-ATPase has a role as cooperating factor of

ATP-dependent membrane proteins that function as

drug efflux pumps (Raghunand et al., 1999).

Interestingly, the levels of V-ATPase subunit

expressions can be up-regulated by anticancer drug.

The treatment of cisplatin on human epidermoid

cancer KB cells increased the protein levels of the

majority of the subunits such as c, c", D, a, A, C

and E, which indicates it may stimulate the

expression of the V-ATPase complex as a whole. It

is suggested that the V-ATPase expression may be

a defensive response to the anticancer drug

(Murakami et al., 2001; Torigoe et al., 2002). Still,

there are also some controversial results on the

relationship between the cationic drugs uptake and

V-ATPase - the inhibition of V-ATPase decreased

the uptake of the cationic drugs (Morissette et al.,

2009; Marceau et al., 2009), which might be

explained that the influence of V-ATPase on the

drug uptake may also be depend upon the

characteristics of the drugs and its relation to

membrane trafficking.

That the defects of V-ATPase increase the

sensitivity to drugs may be partly due to the

decreased cytosolic pH, which were observed in the

influence of cisplatin on the V-ATPase mutant

yeast Saccharomyces cerevisiae (Liao et al., 2006)

or increased toxicity of combined treatment of V-

ATPase inhibition and anticancer drug on lung

cancer cell, breast cancer or liver cancer cell lines

(Wong et al., 2005; Farina et al., 2006; You et al.,

2009). At low cytosolic pH, sensitivity to DNA

damaging drugs or UV irradiation in V-ATPase

mutants may be associated with altered DNA

conformation or defective DNA damage repair

mechanisms, rendering DNA more prone to

damage (Robinson et al., 1992; Petrangolini et al.,

2006; Liao et al., 2006).

Conclusions According to the roles V-ATPase in tumor cells, we

conclude that alteration of V-ATPase is much likely

the necessary initial step of transformation of the

malignant cells and the malfunctional V-ATPase

acts as a continual enhancer of carcinogenesis and

tumor progression. Tumor cells take the advantages

of disfunctioned plasma and intracellular V-ATPase

in these aspects: enhanced proliferation and growth,

Page 80: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function

Lu X, Qin W

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 256

evading apoptosis, facilitating metastasis and

angiogenesis, and acquirement of the drug

resistance. V-ATPase will be a prospective

candidate for cancer diagnosis and treatment.

References Busa WB, Nuccitelli R. Metabolic regulation via intracellular pH. Am J Physiol. 1984 Apr;246(4 Pt 2):R409-38

Perona R, Serrano R. Increased pH and tumorigenicity of fibroblasts expressing a yeast proton pump. Nature. 1988 Aug 4;334(6181):438-40

Robinson H, van der Marel GA, van Boom JH, Wang AH. Unusual DNA conformation at low pH revealed by NMR: parallel-stranded DNA duplex with homo base pairs. Biochemistry. 1992 Nov 3;31(43):10510-7

Clague MJ, Urbé S, Aniento F, Gruenberg J. Vacuolar ATPase activity is required for endosomal carrier vesicle formation. J Biol Chem. 1994 Jan 7;269(1):21-4

Nishihara T, Akifusa S, Koseki T, Kato S, Muro M, Hanada N. Specific inhibitors of vacuolar type H(+)-ATPases induce apoptotic cell death. Biochem Biophys Res Commun. 1995 Jul 6;212(1):255-62

van Weert AW, Dunn KW, Gueze HJ, Maxfield FR, Stoorvogel W. Transport from late endosomes to lysosomes, but not sorting of integral membrane proteins in endosomes, depends on the vacuolar proton pump. J Cell Biol. 1995 Aug;130(4):821-34

Harada M, Sakisaka S, Yoshitake M, Kin M, Ohishi M, Shakado S, Mimura Y, Noguchi K, Sata M, Tanikawa K. Bafilomycin A1, a specific inhibitor of vacuolar-type H(+)-ATPases, inhibits the receptor-mediated endocytosis of asialoglycoproteins in isolated rat hepatocytes. J Hepatol. 1996 May;24(5):594-603

Martínez-Zaguilán R, Seftor EA, Seftor RE, Chu YW, Gillies RJ, Hendrix MJ. Acidic pH enhances the invasive behavior of human melanoma cells. Clin Exp Metastasis. 1996 Mar;14(2):176-86

Ohta T, Numata M, Yagishita H, Futagami F, Tsukioka Y, Kitagawa H, Kayahara M, Nagakawa T, Miyazaki I, Yamamoto M, Iseki S, Ohkuma S. Expression of 16 kDa proteolipid of vacuolar-type H(+)-ATPase in human pancreatic cancer. Br J Cancer. 1996 Jun;73(12):1511-7

Harada M, Shakado S, Sakisaka S, Tamaki S, Ohishi M, Sasatomi K, Koga H, Sata M, Tanikawa K. Bafilomycin A1, a specific inhibitor of V-type H+-ATPases, inhibits the acidification of endocytic structures and inhibits horseradish peroxidase uptake in isolated rat sinusoidal endothelial cells. Liver. 1997 Oct;17(5):244-50

Presley JF, Mayor S, McGraw TE, Dunn KW, Maxfield FR. Bafilomycin A1 treatment retards transferrin receptor recycling more than bulk membrane recycling. J Biol Chem. 1997 May 23;272(21):13929-36

Ohta T, Arakawa H, Futagami F, Fushida S, Kitagawa H, Kayahara M, Nagakawa T, Miwa K, Kurashima K, Numata M, Kitamura Y, Terada T, Ohkuma S. Bafilomycin A1 induces apoptosis in the human pancreatic cancer cell line Capan-1. J Pathol. 1998 Jul;185(3):324-30

Ishisaki A, Hashimoto S, Amagasa T, Nishihara T. Caspase-3 activation during the process of apoptosis induced by a vacuolar type H(+)-ATPase inhibitor. Biol Cell. 1999 Sep;91(7):507-13

Raghunand N, He X, van Sluis R, Mahoney B, Baggett B, Taylor CW, Paine-Murrieta G, Roe D, Bhujwalla ZM, Gillies

RJ. Enhancement of chemotherapy by manipulation of tumour pH. Br J Cancer. 1999 Jun;80(7):1005-11

Kubota S, Seyama Y. Overexpression of vacuolar ATPase 16-kDa subunit in 10T1/2 fibroblasts enhances invasion with concomitant induction of matrix metalloproteinase-2. Biochem Biophys Res Commun. 2000 Nov 19;278(2):390-4

Otani H, Yamamura T, Nakao Y, Hattori R, Fujii H, Ninomiya H, Kido M, Kawaguchi H, Osako M, Imamura H, Ohta T, Ohkuma S. Vacuolar H(+)-ATPase plays a crucial role in growth and phenotypic modulation of myofibroblasts in cultured human saphenous vein. Circulation. 2000 Nov 7;102(19 Suppl 3):III269-74

Murakami T, Shibuya I, Ise T, Chen ZS, Akiyama S, Nakagawa M, Izumi H, Nakamura T, Matsuo K, Yamada Y, Kohno K. Elevated expression of vacuolar proton pump genes and cellular PH in cisplatin resistance. Int J Cancer. 2001 Sep;93(6):869-74

Nishi T, Forgac M. The vacuolar (H+)-ATPases--nature's most versatile proton pumps. Nat Rev Mol Cell Biol. 2002 Feb;3(2):94-103

Torigoe T, Izumi H, Ise T, Murakami T, Uramoto H, Ishiguchi H, Yoshida Y, Tanabe M, Nomoto M, Kohno K. Vacuolar H(+)-ATPase: functional mechanisms and potential as a target for cancer chemotherapy. Anticancer Drugs. 2002 Mar;13(3):237-43

Torigoe T, Izumi H, Ishiguchi H, Uramoto H, Murakami T, Ise T, Yoshida Y, Tanabe M, Nomoto M, Itoh H, Kohno K. Enhanced expression of the human vacuolar H+-ATPase c subunit gene (ATP6L) in response to anticancer agents. J Biol Chem. 2002 Sep 27;277(39):36534-43

Nakashima S, Hiraku Y, Tada-Oikawa S, Hishita T, Gabazza EC, Tamaki S, Imoto I, Adachi Y, Kawanishi S. Vacuolar H+-ATPase inhibitor induces apoptosis via lysosomal dysfunction in the human gastric cancer cell line MKN-1. J Biochem. 2003 Sep;134(3):359-64

Vitavska O, Wieczorek H, Merzendorfer H. A novel role for subunit C in mediating binding of the H+-V-ATPase to the actin cytoskeleton. J Biol Chem. 2003 May 16;278(20):18499-505

Xiao H, Li TK, Yang JM, Liu LF. Acidic pH induces topoisomerase II-mediated DNA damage. Proc Natl Acad Sci U S A. 2003 Apr 29;100(9):5205-10

Zhan H, Yokoyama K, Otani H, Tanigaki K, Shirota N, Takano S, Ohkuma S. Different roles of proteolipids and 70-kDa subunits of V-ATPase in growth and death of cultured human cells. Genes Cells. 2003 Jun;8(6):501-13

Joyce JA, Hanahan D. Multiple roles for cysteine cathepsins in cancer. Cell Cycle. 2004 Dec;3(12):1516-619

Luciani F, Spada M, De Milito A, Molinari A, Rivoltini L, Montinaro A, Marra M, Lugini L, Logozzi M, Lozupone F, Federici C, Iessi E, Parmiani G, Arancia G, Belardelli F, Fais S. Effect of proton pump inhibitor pretreatment on resistance of solid tumors to cytotoxic drugs. J Natl Cancer Inst. 2004 Nov 17;96(22):1702-13

Sennoune SR, Bakunts K, Martínez GM, Chua-Tuan JL, Kebir Y, Attaya MN, Martínez-Zaguilán R. Vacuolar H+-ATPase in human breast cancer cells with distinct metastatic potential: distribution and functional activity. Am J Physiol Cell Physiol. 2004 Jun;286(6):C1443-52

De Milito A, Fais S. Tumor acidity, chemoresistance and proton pump inhibitors. Future Oncol. 2005 Dec;1(6):779-86

Lu X, Qin W, Li J, Tan N, Pan D, Zhang H, Xie L, Yao G, Shu H, Yao M, Wan D, Gu J, Yang S. The growth and metastasis of human hepatocellular carcinoma xenografts

Page 81: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function

Lu X, Qin W

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 257

are inhibited by small interfering RNA targeting to the subunit ATP6L of proton pump. Cancer Res. 2005 Aug 1;65(15):6843-9

Vitavska O, Merzendorfer H, Wieczorek H. The V-ATPase subunit C binds to polymeric F-actin as well as to monomeric G-actin and induces cross-linking of actin filaments. J Biol Chem. 2005 Jan 14;280(2):1070-6

Wong P, Lee C, Tannock IF. Reduction of intracellular pH as a strategy to enhance the pH-dependent cytotoxic effects of melphalan for human breast cancer cells. Clin Cancer Res. 2005 May 1;11(9):3553-7

Yokoyama K, Imamura H. Rotation, structure, and classification of prokaryotic V-ATPase. J Bioenerg Biomembr. 2005 Dec;37(6):405-10

Gerweck LE, Vijayappa S, Kozin S. Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther. 2006 May;5(5):1275-9

Hayashi Y, Katayama K, Togawa T, Kimura T, Yamaguchi A. Effects of bafilomycin A1, a vacuolar type H+ ATPase inhibitor, on the thermosensitivity of a human pancreatic cancer cell line. Int J Hyperthermia. 2006 Jun;22(4):275-85

Hurtado-Lorenzo A, Skinner M, El Annan J, Futai M, Sun-Wada GH, Bourgoin S, Casanova J, Wildeman A, Bechoua S, Ausiello DA, Brown D, Marshansky V. V-ATPase interacts with ARNO and Arf6 in early endosomes and regulates the protein degradative pathway. Nat Cell Biol. 2006 Feb;8(2):124-36

Miele L, Golde T, Osborne B. Notch signaling in cancer. Curr Mol Med. 2006 Dec;6(8):905-18

Petrangolini G, Supino R, Pratesi G, Dal Bo L, Tortoreto M, Croce AC, Misiano P, Belfiore P, Farina C, Zunino F. Effect of a novel vacuolar-H+-ATPase inhibitor on cell and tumor response to camptothecins. J Pharmacol Exp Ther. 2006 Sep;318(3):939-46

Rojas JD, Sennoune SR, Maiti D, Bakunts K, Reuveni M, Sanka SC, Martinez GM, Seftor EA, Meininger CJ, Wu G, Wesson DE, Hendrix MJ, Martínez-Zaguilán R. Vacuolar-type H+-ATPases at the plasma membrane regulate pH and cell migration in microvascular endothelial cells. Am J Physiol Heart Circ Physiol. 2006 Sep;291(3):H1147-57

Zuo J, Jiang J, Chen SH, Vergara S, Gong Y, Xue J, Huang H, Kaku M, Holliday LS. Actin binding activity of subunit B of vacuolar H+-ATPase is involved in its targeting to ruffled membranes of osteoclasts. J Bone Miner Res. 2006 May;21(5):714-21

De Milito A, Iessi E, Logozzi M, Lozupone F, Spada M, Marino ML, Federici C, Perdicchio M, Matarrese P, Lugini L, Nilsson A, Fais S. Proton pump inhibitors induce apoptosis of human B-cell tumors through a caspase-independent mechanism involving reactive oxygen species. Cancer Res. 2007 Jun 1;67(11):5408-17

Fais S, De Milito A, You H, Qin W. Targeting vacuolar H+-ATPases as a new strategy against cancer. Cancer Res. 2007 Nov 15;67(22):10627-30

Forgac M. Vacuolar ATPases: rotary proton pumps in physiology and pathophysiology. Nat Rev Mol Cell Biol. 2007 Nov;8(11):917-29

Gocheva V, Joyce JA. Cysteine cathepsins and the cutting edge of cancer invasion. Cell Cycle. 2007 Jan 1;6(1):60-4

Liao C, Hu B, Arno MJ, Panaretou B. Genomic screening in vivo reveals the role played by vacuolar H+ ATPase and cytosolic acidification in sensitivity to DNA-damaging agents such as cisplatin. Mol Pharmacol. 2007 Feb;71(2):416-25

Roy M, Pear WS, Aster JC. The multifaceted role of Notch in cancer. Curr Opin Genet Dev. 2007 Feb;17(1):52-9

Trédan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst. 2007 Oct 3;99(19):1441-54

Wang Y, Cipriano DJ, Forgac M. Arrangement of subunits in the proteolipid ring of the V-ATPase. J Biol Chem. 2007 Nov 23;282(47):34058-65

Otero-Rey EM, Somoza-Martín M, Barros-Angueira F, García-García A. Intracellular pH regulation in oral squamous cell carcinoma is mediated by increased V-ATPase activity via over-expression of the ATP6V1C1 gene. Oral Oncol. 2008 Feb;44(2):193-9

Gillies RJ, Robey I, Gatenby RA. Causes and consequences of increased glucose metabolism of cancers. J Nucl Med. 2008 Jun;49 Suppl 2:24S-42S

López-Lázaro M. The warburg effect: why and how do cancer cells activate glycolysis in the presence of oxygen? Anticancer Agents Med Chem. 2008 Apr;8(3):305-12

Marshansky V, Futai M. The V-type H+-ATPase in vesicular trafficking: targeting, regulation and function. Curr Opin Cell Biol. 2008 Aug;20(4):415-26

Morimura T, Fujita K, Akita M, Nagashima M, Satomi A. The proton pump inhibitor inhibits cell growth and induces apoptosis in human hepatoblastoma. Pediatr Surg Int. 2008 Oct;24(10):1087-94

Supino R, Petrangolini G, Pratesi G, Tortoreto M, Favini E, Bo LD, Casalini P, Radaelli E, Croce AC, Bottiroli G, Misiano P, Farina C, Zunino F. Antimetastatic effect of a small-molecule vacuolar H+-ATPase inhibitor in in vitro and in vivo preclinical studies. J Pharmacol Exp Ther. 2008 Jan;324(1):15-22

Fortini ME, Bilder D. Endocytic regulation of Notch signaling. Curr Opin Genet Dev. 2009 Aug;19(4):323-8

Hinton A, Sennoune SR, Bond S, Fang M, Reuveni M, Sahagian GG, Jay D, Martinez-Zaguilan R, Forgac M. Function of a subunit isoforms of the V-ATPase in pH homeostasis and in vitro invasion of MDA-MB231 human breast cancer cells. J Biol Chem. 2009 Jun 12;284(24):16400-8

Marceau F, Bawolak MT, Bouthillier J, Morissette G. Vacuolar ATPase-mediated cellular concentration and retention of quinacrine: a model for the distribution of lipophilic cationic drugs to autophagic vacuoles. Drug Metab Dispos. 2009 Dec;37(12):2271-4

Morissette G, Ammoury A, Rusu D, Marguery MC, Lodge R, Poubelle PE, Marceau F. Intracellular sequestration of amiodarone: role of vacuolar ATPase and macroautophagic transition of the resulting vacuolar cytopathology. Br J Pharmacol. 2009 Aug;157(8):1531-40

Sasazawa Y, Futamura Y, Tashiro E, Imoto M. Vacuolar H+-ATPase inhibitors overcome Bcl-xL-mediated chemoresistance through restoration of a caspase-independent apoptotic pathway. Cancer Sci. 2009 Aug;100(8):1460-7

Sun-Wada GH, Tabata H, Kawamura N, Aoyama M, Wada Y. Direct recruitment of H+-ATPase from lysosomes for phagosomal acidification. J Cell Sci. 2009 Jul 15;122(Pt 14):2504-13

Yan Y, Denef N, Schüpbach T. The vacuolar proton pump, V-ATPase, is required for notch signaling and endosomal trafficking in Drosophila. Dev Cell. 2009 Sep;17(3):387-402

You H, Jin J, Shu H, Yu B, De Milito A, Lozupone F, Deng Y, Tang N, Yao G, Fais S, Gu J, Qin W. Small interfering RNA targeting the subunit ATP6L of proton pump V-

Page 82: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function

Lu X, Qin W

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 258

ATPase overcomes chemoresistance of breast cancer cells. Cancer Lett. 2009 Jul 18;280(1):110-9

Buechling T, Bartscherer K, Ohkawara B, Chaudhary V, Spirohn K, Niehrs C, Boutros M. Wnt/Frizzled signaling requires dPRR, the Drosophila homolog of the prorenin receptor. Curr Biol. 2010 Jul 27;20(14):1263-8

Casey JR, Grinstein S, Orlowski J. Sensors and regulators of intracellular pH. Nat Rev Mol Cell Biol. 2010 Jan;11(1):50-61

Cruciat CM, Ohkawara B, Acebron SP, Karaulanov E, Reinhard C, Ingelfinger D, Boutros M, Niehrs C. Requirement of prorenin receptor and vacuolar H+-ATPase-mediated acidification for Wnt signaling. Science. 2010 Jan 22;327(5964):459-63

McCarty MF, Whitaker J. Manipulating tumor acidification as a cancer treatment strategy. Altern Med Rev. 2010 Sep;15(3):264-72

McHenry P, Wang WL, Devitt E, Kluesner N, Davisson VJ, McKee E, Schweitzer D, Helquist P, Tenniswood M. Iejimalides A and B inhibit lysosomal vacuolar H+-ATPase (V-ATPase) activity and induce S-phase arrest and apoptosis in MCF-7 cells. J Cell Biochem. 2010 Mar 1;109(4):634-42

Miranda KC, Karet FE, Brown D. An extended nomenclature for mammalian V-ATPase subunit genes and splice variants. PLoS One. 2010 Mar 10;5(3):e9531

O'Callaghan KM, Ayllon V, O'Keeffe J, Wang Y, Cox OT, Loughran G, Forgac M, O'Connor R. Heme-binding protein HRG-1 is induced by insulin-like growth factor I and associates with the vacuolar H+-ATPase to control endosomal pH and receptor trafficking. J Biol Chem. 2010 Jan 1;285(1):381-91

Toei M, Saum R, Forgac M. Regulation and isoform function of the V-ATPases. Biochemistry. 2010 Jun 15;49(23):4715-23

Vaccari T, Duchi S, Cortese K, Tacchetti C, Bilder D. The vacuolar ATPase is required for physiological as well as pathological activation of the Notch receptor. Development. 2010 Jun;137(11):1825-32

Chung C, Mader CC, Schmitz JC, Atladottir J, Fitchev P, Cornwell ML, Koleske AJ, Crawford SE, Gorelick F. The vacuolar-ATPase modulates matrix metalloproteinase isoforms in human pancreatic cancer. Lab Invest. 2011 May;91(5):732-43

Nishisho T, Hata K, Nakanishi M, Morita Y, Sun-Wada GH, Wada Y, Yasui N, Yoneda T. The a3 isoform vacuolar type H⁺ -ATPase promotes distant metastasis in the mouse B16 melanoma cells. Mol Cancer Res. 2011 Jul;9(7):845-55

This article should be referenced as such:

Lu X, Qin W. Vacuolar H(+)-ATPase in Cancer Cells: Structure and Function. Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):251-258.

Page 83: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Case Report Section Paper co-edited with the European LeukemiaNet

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 259

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

A case of Acute Lymphoblastic Leukemia with rare t(11;22)(q23;q13) Jill D Kremer, Anwar N Mohamed

Cytogenetics Laboratory, Pathology Department, Wayne State University School of Medicine, Detroit

Medical Center, Detroit MI, USA (JDK, ANM)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Reports/t1122q23q13MohamedID100059.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI t1122q23q13MohamedID100059.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Clinics

Age and sex

14 months old male patient.

Previous history

No preleukemia, no previous malignancy, inborn

condition of note. Patient has hemoglobin S trait.

Organomegaly

Hepatomegaly, splenomegaly, enlarged lymph

nodes, central nervous system involvement.

Blood WBC : 33 X 10

9/l

HB : 2.6g/dl

Platelets : 1 X 109/l

Blasts : 72%

Bone marrow : 100 bone marrow blast

replacement.

Cyto-Pathology Classification

Cytology

Acute lymphoblastic leukemia (ALL) with L1

morphology

Immunophenotype

Flow cytometry of bone marrow aspirate identified

a dim CD45 lymphoblast population (85%)

expressing HLA-DR, CD19 and partially

expressing CD10, CD22, CD9 and CD40.

Rearranged Ig Tcr

Not performed.

Pathology

Bone marrow aspirate appeared hypocellular with

95% lymphoblasts of L1 morphology, 2% myeloid

series, and 3% erythroid series.

Electron microscopy

Not performed.

Diagnosis

CD34 negative B-precursor ALL.

Survival

Date of diagnosis: 01-2011

Treatment: Methotrexate, Cytarabine, Vincristine,

Dexamethasone, PEG-aspargase

Complete remission : no

Treatment related death : no

Relapse : no

Status: Alive. Last follow up: 10-2011

Survival: 9 months

Karyotype

Sample: Bone marrow aspirate

Culture time: 24hr without stimulant and 48hr

with 10% conditioned medium.

Banding: GTG

Results

46,Y,der(X)t(X;9)(p11.1;q11),add(9)(q11),t(11;22)(

q23;q13)[20] (see Figure 1). Post induction bone

marrow study demonstrated a normal 46,XY

karyotype.

Page 84: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

A case of Acute Lymphoblastic Leukemia with rare t(11;22)(q23;q13)

Kremer JD, Mohamed AN

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 260

Figure 1: G-banded karyotype showing 46,Y,der(X)t(X;9)(p11.1;q11),add(9)(q11),t(11;22)(q23;q13). Arrows pointed to t(11;22).

Figure 2: FISH. A. Interphase hybridized with LSI MLL dual-color break apart probe showed a split signal pattern of MLL

(1O1G1F). B. Metaphase hybridized with BCR/ABL dual-fusion probe showed 2O2G signaling. C. For identification of chromosome 22, the same metaphase subsequently hybridized with LSI MLL probe showing relocation of the telomeric side

(orange signal) of MLL to 22q confirming t(11;22)(q23;q13) (arrows). Note: G= green; O= orange; F= fusion.

Other Molecular Studies

Technics:

Fluorescence in situ hybridization (FISH) using the

ALL panel DNA probes including CEP 4, 10, and

17 alpha satellite probes, LSI MLL dual-color break

apart probe, BCR/ABL and TEL/AML1 dual-fusion

translocation probes was performed (Abbott

Molecular, Downers Grove, IL).

Results:

Hybridization with MLL probe produced a

split/translocation pattern in 61% of interphase

cells. Metaphase FISH showed that the telomeric

region of MLL gene was translocated to 22q13

distal to BCR (Figure 2). The hybridization with the

BCR/ABL probe showed two signals each

(unfused), however on a previously G-banded

metaphase it appeared that the BCR signals

remained on chromosome 22 while one ABL signal

was translocated to der(X). The remaining probes

produced a normal hybridization pattern.

Comments The patient described here is a 14 month-old-male

presented with an upper respiratory tract infection

unresponsive to antibiotics. Subsequently he was

diagnosed with high risk B-precursor ALL due to

the positivity of MLL/11q23 rearrangement. The

patient was started on a Children's Oncology Group

induction chemotherapy protocol. Secondary to his

high risk status, the patient is being evaluated for a

bone marrow transplant. At time of diagnosis

Page 85: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

A case of Acute Lymphoblastic Leukemia with rare t(11;22)(q23;q13)

Kremer JD, Mohamed AN

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 261

Table 1: AML cases with t(11;22)(q23;q13) reported in literature.

Patient Primary

Malignancy Leukemia Karyotype Gene

4 Y/M [2] Non-Hodgkin

Lymphoma AML M1 48,XY,+8,+8,t(11;22)(q23;q13) MLL-EP300

5 Y/F [3] Neuroblastoma AML M2 46,XX,t(1;22;11)(q44;q13;q23),t(10;17)(q22;q21) MLL-EP300

65 Y/M [4] AML with MDS AMML 46,XY,t(11;22)(q23;q13)[15]/47,idem,+8[2] MLL-EP300

chromosome analysis revealed the presence

t(11;22)(q23;q13) in all 20 metaphases and

rearrangement of the MLL gene.

Translocations involving the MLL/11q23 region are

the most common genomic aberrations in infant

ALL seen in ~80% of cases (Raimondi, 2004).

Generally leukemia harboring MLL translocation is

clinically aggressive and associated with poor

prognosis. The most common chromosomes

involved in 11q23 translocations are t(4;11)

followed by t(11;19) and t(9;11). Additionally,

leukemia with MLL/11q23 translocations are

frequently associated with over expression of

FLT3, therefore, targeted therapy inhibitors of

FLT3 (a tyrosine kinase) may be beneficial for

those patients. Currently there are only three

reported cases in the literature with

t(11;22)(q23;q13), unlike our case all having

secondary acute myeloid leukemia with prior

therapy of topoisomerase II inhibitor (table 1).

Moreover, rearrangement of the MLL gene and

MLL-EP300 fusion gene were demonstrated in

those three cases (Ida et al., 1997; Ohnishi et al.,

2008; Duhoux et al., 2011). The clinical

presentation of our case is quit different from these

three cases. Although our case had a rearrangement

of the MLL/11q23 gene, the MLL-EP300 fusion

gene was not tested. Because the partner genes

involved in MLL/11q23 translocations are

markedly heterogeneous, it remains unclear

whether EP300 or other gene is involved in the

present case which may be responsible for the

different phenotype of this leukemia.

References Ida K, Kitabayashi I, Taki T, Taniwaki M, Noro K, Yamamoto M, Ohki M, Hayashi Y. Adenoviral E1A-associated protein p300 is involved in acute myeloid leukemia with t(11;22)(q23;q13). Blood. 1997 Dec 15;90(12):4699-704

Raimondi SC.. 11q23 rearrangements in childhood acute lymphoblastic leukemia. Atlas Genet Cytogenet Oncol Haematol. February 2004. URL : http://AtlasGeneticsOncology.org/Anomalies/11q23ChildALLID1321.html .

Ohnishi H, Taki T, Yoshino H, Takita J, Ida K, Ishii M, Nishida K, Hayashi Y, Taniwaki M, Bessho F, Watanabe T.. A complex t(1;22;11)(q44;q13;q23) translocation causing MLL-p300 fusion gene in therapy-related acute myeloid leukemia. Eur J Haematol. 2008 Dec;81(6):475-80. Epub 2008 Sep 6.

Duhoux FP, De Wilde S, Ameye G, Bahloula K, Medves S, Lege G, Libouton JM, Demoulin JB, A Poirel H.. Novel variant form of t(11;22)(q23;q13)/MLL-EP300 fusion transcript in the evolution of an acute myeloid leukemia with myelodysplasia-related changes. Leuk Res. 2011 Mar;35(3):e18-20. Epub 2010 Oct 25.

This article should be referenced as such:

Kremer JD, Mohamed AN. A case of Acute Lymphoblastic Leukemia with rare t(11;22)(q23;q13). Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):259-261.

Page 86: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Case Report Section Paper co-edited with the European LeukemiaNet

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 262

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Insertion as an alternative mechanism of CBFB-MYH11 gene fusion in a new case of acute myeloid leukemia with an abnormal chromosome 16 Yaser Hussein, Vandana Kulkarni, Anwar N Mohamed

Cytogenetics Laboratory, Pathology Department, Wayne State University School of Medicine, Detroit

Medical Center, Detroit MI, USA (YH, VK, ANM)

Published in Atlas Database: October 2011

Online updated version : http://AtlasGeneticsOncology.org/Reports/ins16q22p13p13MohamID100058.html Printable original version : http://documents.irevues.inist.fr/bitstream/DOI ins16q22p13p13MohamID100058.txt This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2012 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Clinics

Age and sex

17 years old female patient.

Previous history

No preleukemia, no previous malignancy, inborn

condition of note. Thalassemia trait carrier.

Organomegaly

Hepatomegaly, splenomegaly, enlarged lymph

nodes, no central nervous system involvement.

Blood WBC : 138.7 X 10

9/l

HB : 6.9g/dl

Platelets : 51 X 109/l

Blasts : 76%

Bone marrow : 100 Bone marrow biopsy was

hypercellular (100%) and replaced by myeloblasts

and monoblasts. Normal hematopoiesis was greatly

decreased and there was prominent

hemophagocytosis. The majority of the blasts were

myeloperoxidase positive however another smaller

component of blasts was nonspecific esterase

positive.

Cyto-Pathology Classification Cytology: Acute myeloid leukemia with abnormal

eosinophils (AML-M4eos).

Immunophenotype

Flow cytometry of bone marrow aspirate identified

a significant population of myeloblasts (49%)

expressing CD34, HLA-DR, CD9, CD13, CD33,

CD117 and partially expressing CD15, CD11b, and

CD64. A second population of monocytes is also

identified (37%) expressing CD4, CD14, CD15,

CD36 and CD64.

Rearranged Ig Tcr: Not performed.

Pathology

Bone marrow aspirate revealed myeloblasts,

monoblasts, monocytes, and increased eosinophils

many of which had abnormal granules (FAB AML-

M4eos).

Electron microscopy: Not performed.

Diagnosis

Acute myelomonocytic leukemia with abnormal

eosinophils (AML-M4eos) and CBFB/16q22

rearrangement.

Survival

Date of diagnosis: 03-2011

Treatment: Intrathecal methotrexate,

hydrocortisone, and cytarabine.

Treatment related death : no

Relapse : no

Status: Alive. Last follow up: 09-2011

Survival: 6 months

Page 87: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Insertion as an alternative mechanism of CBFB-MYH11 gene fusion in a new case of acute myeloid leukemia with an abnormal chromosome 16

Hussein Y, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 263

Karyotype

Sample: Bone marrow

Culture time: 24 hrs without stimulating agents

and 48 hrs with 10% conditioned medium.

Banding: GTG

Results

At time of diagnosis abnormal metaphase cells with

the following karyotype was found;

46,XX,ins(16)(q22p13p13)[20] (see Figure 1).

Remission bone marrow on 4/20/2011 and

9/13/2011 revealed a normal female karyotype;

46,XX[20].

Other Molecular Studies

Technics:

Fluorescence in situ hybridization (FISH) using LSI

CBFB dual color break-apart rearrangement DNA

probes (Abbott Molecular IL, USA), and

CBFB/MYH11 dual fusion translocation DNA

probe (Cytocell Inc. Cambridge, UK) were

performed.

Results:

The hybridization with the CBFB break-apart probe

produced a split pattern in 62% of interphase cells.

On metaphase cells, the 5'CBFB (SepctrumRed)

and 3'CBFB (SepctrumGreen) signals stayed on the

16q, instead of 5'CBFB being relocated to 16p as

seen in the standard inv(16). The CBFB signals

were separated but maintained the orientation

pattern of the 5' and 3' probe, suggesting they were

split by an insertion (Figure 2A). Subsequently,

using the CBFB-MYH11 probe on metaphases

showed that MYH11 signal on 16p moved and

juxtaposed to CBFB on 16q, confirming the

insertion of MYH11 into CBFB (Figure 2B).

Figure 1. G-Banded karyotype from the diagnostic bone marrow sample demonstrating the ins(16)(q22p13p13) (arrowed).

Page 88: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Insertion as an alternative mechanism of CBFB-MYH11 gene fusion in a new case of acute myeloid leukemia with an abnormal chromosome 16

Hussein Y, et al.

Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3) 264

Figure 2. A. Metaphase FISH using LSI CBFB/q22 breakapart rearrangement probe showing one normal fusion signal and split signals (red and green) on 16q (arrow). B. Metaphase hybridized with CBFB/MYH11 probe showing insertion of MYH11 green

signal (appearing yellow) within CBFB/16q22 red signal (arrow).

Comments The patient described here is a 17 year old female

presented with upper respiratory tract infection and

bruises for 2 weeks. Subsequently she was

diagnosed with AML (FAB M4 eos). Cytogenetics,

performed on bone marrow aspirate revealed a

unique structural abnormality of chromosome 16

which was interpreted as insertion; 46, XX,

ins(16)(q22p13p13). FISH confirmed that the

MYH11/p13 gene was inserted into the

CBFB/16q22 gene region (Figure 2B). The result of

this unusual structural rearrangement was the fusion

of CBFB /MYH11 genes commonly seen in

inv(16)(p13q22) bearing leukemia.

The CBFB/MYH11 gene fusion is strongly

associated with AML-M4 with abnormal

eosinophils. Generally, the fusion is generated from

inv(16)(p13q22) or t(16;16) with the inversion

being much more common than translocation (Le

Beau et al., 1983; Tobal et al., 1995). The case

presented here demonstrates that insertion is

another mechanism in producing CBFB/MYH11

gene fusion in AML-M4eos. To our best

knowledge, there is only one reported case of

AML-M4 having similar structural abnormality of

chromosome 16 and CBFB/MYH11 fusion

(O'Reilly et al., 2000). These two cases suggest that

insertion represents a variant rare rearrangement for

the formation of this fusion. FISH is highly

recommended to characterize unusual abnormalities

of chromosome 16 and to confirm the CBFB-

MYH11 fusion.

References Le Beau MM, Larson RA, Bitter MA, Vardiman JW, Golomb HM, Rowley JD. Association of an inversion of chromosome 16 with abnormal marrow eosinophils in acute myelomonocytic leukemia. A unique cytogenetic-clinicopathological association. N Engl J Med. 1983 Sep 15;309(11):630-6

Tobal K, Johnson PR, Saunders MJ, Harrison CJ, Liu Yin JA. Detection of CBFB/MYH11 transcripts in patients with inversion and other abnormalities of chromosome 16 at presentation and remission. Br J Haematol. 1995 Sep;91(1):104-8

O'Reilly J, Chipper L, Springall F, Herrmann R. A unique structural abnormality of chromosome 16 resulting in a CBF beta-MYH11 fusion transcript in a patient with acute myeloid leukemia, FAB M4. Cancer Genet Cytogenet. 2000 Aug;121(1):52-5

This article should be referenced as such:

Hussein Y, Kulkarni V, Mohamed AN. Insertion as an alternative mechanism of CBFB-MYH11 gene fusion in a new case of acute myeloid leukemia with an abnormal chromosome 16. Atlas Genet Cytogenet Oncol Haematol. 2012; 16(3):262-264.

Page 89: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

Instructions to Authors Manuscripts submitted to the Atlas must be submitted solely to the Atlas.

Iconography is most welcome: there is no space restriction.

The Atlas publishes "cards", "deep insights", "case reports", and "educational items".

Cards are structured review articles. Detailed instructions for these structured reviews can be found at:

http://AtlasGeneticsOncology.org/Forms/Gene_Form.html for reviews on genes,

http://AtlasGeneticsOncology.org/Forms/Leukaemia_Form.html for reviews on leukaemias,

http://AtlasGeneticsOncology.org/Forms/SolidTumour_Form.html for reviews on solid tumours,

http://AtlasGeneticsOncology.org/Forms/CancerProne_Form.html for reviews on cancer-prone diseases.

According to the length of the paper, cards are divided, into "reviews" (texts exceeding 2000 words), "mini reviews"

(between), and "short communications" (texts below 400 words). The latter category may not be accepted for indexing by

bibliographic databases.

Deep Insights are written as traditional papers, made of paragraphs with headings, at the author's convenience. No length

restriction.

Case Reports in haematological malignancies are dedicated to recurrent -but rare- chromosomes abnormalities in

leukaemias/lymphomas. Cases of interest shall be: 1- recurrent (i.e. the chromosome anomaly has already been described in

at least 1 case), 2- rare (previously described in less than 20 cases), 3- with well documented clinics and laboratory findings,

and 4- with iconography of chromosomes.

It is mandatory to use the specific "Submission form for Case reports":

see http://AtlasGeneticsOncology.org/Reports/Case_Report_Submission.html.

Educational Items must be didactic, give full information and be accompanied with iconography. Translations into French,

German, Italian, and Spanish are welcome.

Subscription: The Atlas is FREE!

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

Rules, Copyright Notice and Disclaimer Conflicts of Interest: Authors must state explicitly whether potential conflicts do or do not exist. Reviewers must disclose to

editors any conflicts of interest that could bias their opinions of the manuscript. The editor and the editorial board members

must disclose any potential conflict.

Privacy and Confidentiality – Iconography: Patients have a right to privacy. Identifying details should be omitted. If

complete anonymity is difficult to achieve, informed consent should be obtained.

Property: As "cards" are to evolve with further improvements and updates from various contributors, the property of the

cards belongs to the editor, and modifications will be made without authorization from the previous contributor (who may,

nonetheless, be asked for refereeing); contributors are listed in an edit history manner. Authors keep the rights to use further

the content of their papers published in the Atlas, provided that the source is cited.

Copyright: The information in the Atlas of Genetics and Cytogenetics in Oncology and Haematology is issued for general

distribution. All rights are reserved. The information presented is protected under international conventions and under

national laws on copyright and neighbouring rights. Commercial use is totally forbidden. Information extracted from the

Atlas may be reviewed, reproduced or translated for research or private study but not for sale or for use in conjunction with

commercial purposes. Any use of information from the Atlas should be accompanied by an acknowledgment of the Atlas as

the source, citing the uniform resource locator (URL) of the article and/or the article reference, according to the Vancouver

convention. Reference to any specific commercial products, process, or service by trade name, trademark, manufacturer, or

otherwise, does not necessarily constitute or imply its endorsement, recommendation, or favouring. The views and opinions

of contributors and authors expressed herein do not necessarily state or reflect those of the Atlas editorial staff or of the web

site holder, and shall not be used for advertising or product endorsement purposes. The Atlas does not make any warranty,

express or implied, including the warranties of merchantability and fitness for a particular purpose, or assumes any legal

liability or responsibility for the accuracy, completeness, or usefulness of any information, and shall not be liable whatsoever

for any damages incurred as a result of its use. In particular, information presented in the Atlas is only for research purpose,

and shall not be used for diagnosis or treatment purposes. No responsibility is assumed for any injury and/or damage to

persons or property for any use or operation of any methods products, instructions or ideas contained in the material herein.

See also: "Uniform Requirements for Manuscripts Submitted to Biomedical Journals: Writing and Editing for Biomedical

Publication - Updated October 2004": http://www.icmje.org.

http://AtlasGeneticsOncology.org

© ATLAS - ISSN 1768-3262

Page 90: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16
Page 91: Volume 16 - Volume 1 -Number 3Number 1 May - September 1997 …atlasgeneticsoncology.org/Journal/Arch2012Vol16Num3.pdf · Volume 1 -Number 3Number 1 May - September 1997 Volume 16

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS