18
Wilms tumor cells with WT1 mutations have characteristic features of mesenchymal stem cells and express molecular markers of paraxial mesoderm Brigitte Royer-Pokora 1, , Maike Busch 1 , Manfred Beier 1 , Constanze Duhme 1 , Carmen de Torres 2 , Jaume Mora 2 , Artur Brandt 1 and Hans-Dieter Royer 1,3 1 Institute of Human Genetics and Anthropology, Heinrich-Heine University, Du ¨ sseldorf D-40225, Germany, 2 Department of Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain and 3 Center of Advanced European Studies and Research, CAESAR, Bonn, Germany Received September 30, 2009; Revised and Accepted January 26, 2010 Wilms tumors (WTs) are genetically heterogeneous kidney tumors whose cells of origin are unknown. Tumors with WT1 mutations and concomitant loss of the wild-type allele represent a distinct subgroup, fre- quently associated with mutations in CTNNB1. Here, we describe the establishment and characterization of long-term cell cultures derived from five individual WTs with WT1 mutations. Three of these tumor cell lines also had CTNNB1 mutations and an activated canonical Wnt signaling pathway as measured by b-catenin/ T cell-specific transcription factor (TCF) transcriptional activity. Four of the five Wilms cell lines had a stable normal karyotype for at least 25 passages, and four lines showed loss of heterozygosity of chromosome 11p due to mitotic recombination in 11p11. Gene expression profiling revealed that the WT cell lines are highly similar to human mesenchymal stem cells (MSCs) and FACS analysis demonstrated the expression of MSC- specific surface proteins CD105, CD90 and CD73. The stem cell like nature of the WT cells is further supported by their adipogenic, chondrogenic, osteogenic and myogenic differentiation potentials. By generating multi- potent mesenchymal precursors from paraxial mesoderm (PAM) in tissue culture using embryonal stem cells, gene expression profiles of PAM and MSCs were described. Using these published gene sets, we found coexpression of a large number of genes in WT cell lines, PAM and MSCs. Lineage plasticity is indicated by the simultaneous expression of genes from the mesendodermal and neuroectodermal lineages. We con- clude that WTs with WT1 mutations have specific traits of PAM, which is the source of kidney stromal cells. INTRODUCTION Wilms tumor (WT) or nephroblastoma is one of the most common childhood malignancies, affecting 1/10 000 children. The classic triphasic WT contains a mixture of undifferen- tiated blastemal, differentiated epithelial cells and stromal elements, whereas individual cell types may predominate in some tumors. These different components are thought to arise from a multipotential mesenchymal renal stem/progeni- tor cell, which fails to undergo a proper differentiation program (1). The epithelial and blastemal elements in triphasic WT mimic structures seen during normal nephrogenesis, whereas the stromal-predominant/stromal-type WT contains ectopic mesenchymal elements not usually seen in kidneys, mostly striated and smooth muscle cells (rhabdomyogenesis), as well as chondrocytes, osteocytes or adipocytes. Another term often used for these tumors is mesenchymal tumors with ectopic differentiation. These cell types are reminiscent of the differentiation potential of human mesenchymal stem cells (MSCs), which have the capacity to differentiate along a number of connective tissue lineages, such as cartilage, bone, adipocyte or muscle (2). To whom correspondence should be addressed at: Institute of Human Genetics and Anthropology, Heinrich-Heine-University of Duesseldorf, Postfach 101007, Duesseldorf D40001, Germany. Tel: þ49 2118112350; Fax: þ49 2118112538; Email: [email protected] # The Author 2010. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected] Human Molecular Genetics, 2010, Vol. 19, No. 9 1651–1668 doi:10.1093/hmg/ddq042 Advance Access published on January 27, 2010 Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433 by guest on 18 March 2018

Wilms tumor cells with WT1 mutations have characteristic features of

  • Upload
    lehanh

  • View
    221

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Wilms tumor cells with WT1 mutations have characteristic features of

Wilms tumor cells with WT1 mutations havecharacteristic features of mesenchymal stemcells and express molecular markersof paraxial mesoderm

Brigitte Royer-Pokora1,�, Maike Busch1, Manfred Beier1, Constanze Duhme1,

Carmen de Torres2, Jaume Mora2, Artur Brandt1 and Hans-Dieter Royer1,3

1Institute of Human Genetics and Anthropology, Heinrich-Heine University, Dusseldorf D-40225, Germany,2Department of Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain and 3Center of Advanced European

Studies and Research, CAESAR, Bonn, Germany

Received September 30, 2009; Revised and Accepted January 26, 2010

Wilms tumors (WTs) are genetically heterogeneous kidney tumors whose cells of origin are unknown.Tumors with WT1 mutations and concomitant loss of the wild-type allele represent a distinct subgroup, fre-quently associated with mutations in CTNNB1. Here, we describe the establishment and characterization oflong-term cell cultures derived from five individual WTs with WT1 mutations. Three of these tumor cell linesalso had CTNNB1 mutations and an activated canonical Wnt signaling pathway as measured by b-catenin/T cell-specific transcription factor (TCF) transcriptional activity. Four of the five Wilms cell lines had astable normal karyotype for at least 25 passages, and four lines showed loss of heterozygosity of chromosome11p due to mitotic recombination in 11p11. Gene expression profiling revealed that the WT cell lines are highlysimilar to human mesenchymal stem cells (MSCs) and FACS analysis demonstrated the expression of MSC-specific surface proteins CD105, CD90 and CD73. The stem cell like nature of the WT cells is further supportedby their adipogenic, chondrogenic, osteogenic and myogenic differentiation potentials. By generating multi-potent mesenchymal precursors from paraxial mesoderm (PAM) in tissue culture using embryonal stemcells, gene expression profiles of PAM and MSCs were described. Using these published gene sets, wefound coexpression of a large number of genes in WT cell lines, PAM and MSCs. Lineage plasticity is indicatedby the simultaneous expression of genes from the mesendodermal and neuroectodermal lineages. We con-clude that WTs with WT1 mutations have specific traits of PAM, which is the source of kidney stromal cells.

INTRODUCTION

Wilms tumor (WT) or nephroblastoma is one of the mostcommon childhood malignancies, affecting 1/10 000 children.The classic triphasic WT contains a mixture of undifferen-tiated blastemal, differentiated epithelial cells and stromalelements, whereas individual cell types may predominate insome tumors. These different components are thought toarise from a multipotential mesenchymal renal stem/progeni-tor cell, which fails to undergo a proper differentiationprogram (1). The epithelial and blastemal elements in triphasic

WT mimic structures seen during normal nephrogenesis,whereas the stromal-predominant/stromal-type WT containsectopic mesenchymal elements not usually seen in kidneys,mostly striated and smooth muscle cells (rhabdomyogenesis),as well as chondrocytes, osteocytes or adipocytes. Anotherterm often used for these tumors is mesenchymal tumorswith ectopic differentiation. These cell types are reminiscentof the differentiation potential of human mesenchymal stemcells (MSCs), which have the capacity to differentiate alonga number of connective tissue lineages, such as cartilage,bone, adipocyte or muscle (2).

�To whom correspondence should be addressed at: Institute of Human Genetics and Anthropology, Heinrich-Heine-University of Duesseldorf, Postfach101007, Duesseldorf D40001, Germany. Tel: þ49 2118112350; Fax: þ49 2118112538; Email: [email protected]

# The Author 2010. Published by Oxford University Press. All rights reserved.For Permissions, please email: [email protected]

Human Molecular Genetics, 2010, Vol. 19, No. 9 1651–1668doi:10.1093/hmg/ddq042Advance Access published on January 27, 2010

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 2: Wilms tumor cells with WT1 mutations have characteristic features of

Mutations in the WT1 gene, a zinc finger transcription factorare found in 10–15% of WTs and are most frequent instromal-type WT (3–8). We have reported that WT1 germ-linemutations are often associated with loss of the second wild-type allele, confirming Knudson’s two-hit hypothesis for thedevelopment of this specific subclass of WT (3,7). Intralobarnephrogenic rests (ILNR), which are thought to be precursorlesions are found in WTs derived from patients with germ-lineWT1 mutations or WT1 deletions as in Wilms tumor, aniridia,genitourinary anomalies and mental retardation (WAGR)patients. It was shown that ILNR harbor homozygous WT1mutations, whereas the associated tumors have additionalCTNNB1 mutations, demonstrating the order of these geneticevents (9).

A high level of WT1 expression is observed at the time ofmesenchymal-to-epithelial transition during kidney develop-ment, pointing to its role in epithelial differentiation (10).Loss of a functional WT1 protein in mesenchymal cells ofmutant tumors might provoke a faulty differentiation intomyogenic, fat or cartilage lineages. WT1 is expressed in blas-tema and glomeruloid structures but is absent from stromalcells in WTs lacking mutations (8).

Mutations in exon 3 (destruction box) of CTNNB1 at aminoacid positions that are targets for phosphorylation were ident-ified in WTs (11–13). Owing to these mutations, b-catenincannot be phosphorylated resulting in its stabilization andmigration to the nucleus, where it activates Wnt target genestogether with T cell-specific transcription factor/lymphoidenhancer-binding factor. The regulation of b-catenin phos-phorylation is important for controlling Wnt signal transduction.CTNNB1 mutations are often associated with mutations in WT1(8,12,13) and in our recent analyses, we found that all WT1mutant tumors also had CTNNB1 mutations (14). These datasuggest that there is a high selection pressure for WT1 mutanttumors to acquire additional mutations in CTNNB1. Nuclearaccumulation of b-catenin, an indicator for activated Wnt sig-naling, was found in WTs with CTNNB1 mutations, althoughnot all mutant tumors showed nuclear b-catenin staining(8,11). In CTNNB1 mutant tumors nuclear localization wasobserved in undifferentiated mesenchymal cells (8).

Tumors with wild-type WT1 are a distinct subgroup oftenwith epigenetic alterations (loss of imprinting, LOI) in11p15 targeting the H19 and IGF2 genes (15). Furthermore,most of these WT1 wild-type tumors express high levels ofWT1 (C.D. and B.R.-P., unpublished data). Therefore, atleast two genetically distinct molecular subgroups of WTsexist, underscoring the heterogeneity of this type of tumor.

WTs are thought to be derived from kidney progenitor cells,but the identification of human kidney stem cells remainselusive. Several reports have described the isolation ofembryonic renal stem cells from mice and rats, which candifferentiate into myofibroblasts, endothelial and epithelialcells as well as myogenic, osteogenic, adipogenic and neurallineages (16–19). In addition, kidney side population cellswere isolated from embryonic and adult murine kidneys,capable of multilineage differentiation in vitro (20). Isolationof renal progenitors from human kidneys based on theexpression of CD133 also revealed that these cells arecapable of epithelial and endothelial differentiation (21).However, it has not been shown unequivocally that one type

of kidney progenitor cell exists, giving rise to all componentsof the kidney. Pure progenitor cells to address these questionsare not available.

The kidney is derived from the intermediate mesoderm,located between the lateral plate mesoderm and paraxial meso-derm (PAM). Nephrogenesis is a process of reciprocal induc-tive cell–cell interactions between the ureteric bud (UB),metanephric mesenchyme (MM) and stromal cells, ultimatelyresulting in mesenchymal-to-epithelial conversion andnephron formation. Most cells in the adult kidney areformed from either UB, giving rise to epithelial cells of thecollecting duct or MM to the rest of the epithelial cells ofthe nephron. In addition, stromal cells are important forkidney morphogenesis as shown by targeted disruption ofBf2/Foxd1 (22). Foxd1 mutants displayed a reduced numberof UB branches and nephrons, suggesting that Foxd1 isrequired for the formation of the renal capsule, establishingcorrect zones for induction and differentiation (23). Stromalcells in the kidney were believed to be derived from the inter-mediate mesoderm; however, fate mapping studies in thechick embryo showed that stromal cells of the developingkidney are derived from pax3þ PAM cells (24). Furthermore,fate mapping studies of pax3þ progenitor cells in reportermice (25) are consistent with those in the chicken andsupport the hypothesis that renal stromal cells derive frompax3þ PAM progenitors. Pax3 is a transcription factor firstexpressed in early somitogenesis, specifically in limbmuscle, PAM, neural crest and in the neural tube duringembryonic development and is implicated in migration anddifferentiation, as well as formation of hypaxial muscle.

The cells of origin for the different forms of WTs areunknown. In order to better understand the biology of WTs,a cell culture model system is needed. Primary WT cellshave a very limited lifespan in tissue culture and establishingcell lines from WTs has been notoriously difficult (26–30). Sofar, only a few WT-derived cell lines capable of long-termpropagation have been established, some with highlycomplex karyotypes (31–33). In contrast, WT cells could bepropagated to some extent after heterotransplantation intonude mice (34,35).

Here, we report a new method for the establishment of long-term homozygous WT1 mutant cell lines derived from WTsthat have different genetic alterations in the WT1 gene.Characterization of these novel cell lines provides experimen-tal evidence that the tumor initiating cells of WTs with WT1mutations may reside in PAM.

RESULTS

Establishment of cell lines from five individual WTs withWT1 mutations

As WTs with WT1 mutations show mesenchymal differen-tiation, we hypothesized that the WT cells have features ofMSCs. To test this hypothesis, we wished to establish long-term cell cultures from WTs with WT1 mutations. Initially,we cultivated explants from primary tumors in a tissueculture medium specifically developed for WTs (WTmedium) (27). Cells established with this medium showedepitheloid morphology, but could not be cultivated for more

1652 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 3: Wilms tumor cells with WT1 mutations have characteristic features of

than 1 month and lost their epitheloid morphology after 2–3passages (Fig. 1A). Next, we tested whether cells from WTswith WT1 mutations could be cultivated in a medium specifi-cally developed for the propagation of MSCs (MSC growthmedium, MSCGM). Therefore, tumor cell cultures wereinitiated in MSCGM and a parallel cell culture was set up inWT medium. The morphology of the WT cells in MSCGMwas spindle shaped and resembled MSCs (Figs 1B and 4A,left panels). Analyzing cells grown in these two differentmedia for the presence of WT1 mutations revealed that homo-zygous WT1 mutant cell lines could only be established inMSCGM, but not in WT medium. The presence of themutation was analyzed by restriction enzyme digest usingthe enzyme Mse1, which cuts only the mutant DNA. DNAfrom the original tumor sample contains more mutant DNAas seen by the presence of two strong bands at 110 and100 bp, whereas a faint band at 210 bp corresponding to theuncut wild-type DNA is seen (Fig. 1A, bottom, lane T).WT1 wild-type normal kidney (NK) cells (corresponding tothe band at 210 bp) were selectively enriched when tumorexplants from patient Wilms3 were cultivated in WTmedium and only wild-type DNA was observed already atpassage 2 (Fig. 1A, bottom, lanes P2 and P3). In contrast, atpassage 1 in MSGM, the culture consisted of a mixture ofcells with �50% mutant and wild-type WT1 alleles(Fig. 1B, bottom, P1). A homogeneous population of homozy-gous WT1 mutant cells was observed after passage six(Fig. 1B, bottom P6 and P8). As this patient has a tumor-specific WT1 mutation, normal cells have only wild-typeWT1 alleles (see below).

By using these culture conditions, we have now successfullyestablished cell lines from five different WTs which are

characterized by different homozygous WT1 mutations. Thenew WT-derived cell lines were cultivated for at least 60population doublings. The doubling time varied between 1.8and 3 days. The WT cell lines were termed Wilms1–5 inthis work.

Identification of WT1 and CTNNB1 mutations in WT celllines

A detailed description of the patient’s mutation status and thecell lines derived from their WTs is found in Table 1. Wilms1with a homozygous WT1 p.S50X stop mutation and anadditional p.S45F CTNNB1 mutation has been describedbefore (36). The Wilms2 cell line has a homozygousp.R362X WT1 stop mutation due to a mitotic recombinationin 11p11 and carries a p.S45Y CTNNB1 mutation. Wilms3harbors a tumor-specific homozygous WT1 mutation, resultingin a C-terminal extension of the WT1 protein and a wild-typeCTNNB1 gene. Wilms4, derived from a WAGR patient has atumor-specific WT1 mutation in the remaining allele and ap.DS45 CTNNB1 mutation. Wilms5 is unusual, as the germ-line p.R390X WT1 mutation was lost in the tumor. Thistumor is characterized by a novel homozygous WT1 mutationin exon 10 leading to an extended protein p.R433Pfs84 and awild-type CTNNB1 gene. The frameshift WT1 mutations inWilms3, 4 and 5 in exon 10 lead to a stop codon in the30-UTR at the same position yielding an extended WT1protein with 79 identical amino acids at the C terminus(Fig. 2A). The altered amino acid sequence does not followthe consensus sequence for a C2H2 type zinc finger(CX2 – 4C-X12-H-X3-H) and it is therefore unlikely that anintact Zinc finger 4 (ZF) is present in these mutant proteins.

Figure 1. Establishment of cell lines from primary WTs. (A) Top: Wilms3 cell culture initiated in WT medium. When the cells were kept in WT medium withoutsubculturing, they retained their epithelial morphology and showed signs of epithelial differentiation. Bottom: analysis of DNA from the Wilms3 culture atpassage 2 (P2) and 3 (P3) in comparison to the DNA extracted from the tumor (T) by restriction enzyme digest with Mse1 for the presence of the WT1 mutation.C: undigested wild-type control DNA. (B) Top: Wilms3 cell culture established in MSCGM, early passage cells at lower density. Bottom: disappearance ofwild-type WT1 during passaging: in P1, the culture shows wild-type and mutant WT1 alleles, whereas in P6 all cells show only mutant WT1.

Human Molecular Genetics, 2010, Vol. 19, No. 9 1653

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 4: Wilms tumor cells with WT1 mutations have characteristic features of

To test whether mutant WT1 transcripts are expressed, weperformed quantitative real-time PCR (Q-RT-PCR). Thelevel of WT1 mRNA varied between Wilms 1–5 and theQ-RT-PCR data were highly concordant with the microarraydata (Fig. 2B). The presence of WT1 mutations in expressedmRNAs was verified by sequencing and reported forWilms1 (36). The sequencing data for Wilms2, 3, 4 and 5

are not shown. All four major WT1 transcript isoforms wereexpressed, corresponding to transcripts lacking or containingexon 5 and lacking or containing the alternative splice sitecoding for three amino acids (KTS). The abbreviations forthese four isoforms are WT1A (2exon5/2KTS), WT1B(þexon5/2KTS), WT1C (2exon5/þKTS) and WT1D(þexon5/þKTS). Next, we studied whether mutant WT1

Table 1. Mutation status of WT cell lines

WT1 mutation status of patient in blood DNA WT1 mutation status in cell lines LOH CTNNB1 mutation

Wilms1 germ-line c.149 C.A, p.S50X Homozygous p.S50X 11p11–11pter Heterozygous p.S45FWilms2 germ-line, c.1084C.T, p.R362X Homozygous p.R362X 11p11–11pter Heterozygous p.S45YWilms3 wild-type Homozygous c.1293–1294insA, p.V432fsX87 11p11–11pter Wild-typeWilms4 WAGR, del 11p13 WT1 deletion and hemizygous c.1311–1312insC,

p.H438Pfs79No LOH Heterozygous p.DS45

Wilms5 germ-line c.1168 C.T, p.R390X Homozygous, c.1296–1299delGC, p.R433Pfs84 11p11–11pter loss of R390X Wild-typeWilms-cell line CLS1, unknown Wild-type No LOH Wild-type

Figure 2. WT cells with WT1 mutations express WT1 protein. (A) Amino acid sequence of normal and mutant WT1 proteins from Wilms3, Wilms4 and Wilms5cells. The frameshift WT1 mutations lead to identical stop codons in the 30-UTR yielding extended WT1 proteins with identical amino acids at the C terminus(yellow). The first altered amino acids are indicated (red). (B) WT1 mRNA expression levels in the five Wilms cell lines and MSCs. A highly concordant result isobtained by Q-RT-PCR and Agilent microarrays. The standard error was derived from three Q-RT-PCR experiments and from five individual microarrays. (C)Western blot analysis of WT1 expression in Wilms3 and M15 cells. n, nuclear extract; c, cytoplasm. (D) Fluorescence microscopy shows the subcellular local-ization of four transfected GFP-Wilms3 protein isoforms in live NIH-3T3 cells. The mutant WT1 isoforms are: A (2/2), B (þ/2), D (þ/þ) and C (2/þ).A more detailed description of mutant WT1 isoforms is given in the text.

1654 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 5: Wilms tumor cells with WT1 mutations have characteristic features of

proteins are expressed in the Wilms cell lines. Because thehighest level of WT1 mRNA was observed in Wilms3, weinitially analyzed whole cell lysates from this cell line usingan antibody specific for the N-terminus of WT1. In this exper-iment, WT1 was not detectable and we decided to enrich theproteins by fractionation into nuclear and cytoplasmic pro-teins. With the help of a highly sensitive western blot kit,we detected the mutant WT1 protein in cytoplasmic extractsof Wilms3, with a slightly higher level of the þexon5 thanthe 2exon5 isoform of WT1 (Fig. 2C). In comparison, inM15 cells, derived from mesonephric epithelial cells of themouse embryo, Wt1 is mostly found in the nucleus andthese cells express more of the 2exon5 isoform. After trans-fection of GFP–WT1 fusion proteins corresponding to thefour mutant WT1 isoforms from Wilms3, a predominant cyto-plasmic localization was observed for isoforms A (2/2), B(þ/2) and D (þ/þ), however isoform C (2/þ) was alsoobserved in the nucleus of NIH3T3 cells (Fig. 2D). Wilms1,2, 4 and 5 express very low levels of WT1 mRNA and theWT1 protein was not detectable by immunoblotting presum-ably due to the detection limit.

Karyotype and LOH analysis of Wilms cell lines

Karyotype analysis revealed a stable normal karyotype inWilms 1, 2, 4 and 5. Wilms3 cells had a mosaic karyotype atpassage 16: _4_7,XY,þ18[19]/46,XY[4]. FISH using chromo-some 18 centromere and telomere probes revealed that atpassage two, trisomy 18 was present in 59% of the cells,increasing to 74% by passage 16. Cells established from thenormal adjacent kidney of this patient had a normal karyotype,but FISH analysis revealed trisomy 18 in 5.1% of the cells.These cells were wild-type in WT1, indicating that they wereNK cells, as in this case the WT1 mutation is somatic andonly present in tumor cells. As blood DNA was unavailable,we could not investigate whether this patient has a germ-linemosaicism for trisomy 18. Constitutional trisomy 18 has beendescribed as a risk factor for WT development (37).

All Wilms cell lines had two copies of chromosome 11.Loss of heterozygosity (LOH) analyses showed that in fourcell lines a mitotic recombination event had occurredbetween markers in 11p11 and 11p13, whereas the cell linefrom the WAGR patient (Wilms4) has retained both 11p par-ental alleles. Loss of the maternal allele could be identified inWilms1 and 2, where parental DNA was available. Therefore,two copies of paternal alleles (paternal uniparental disomy,UPD) from 11p11! 11pter, containing the IGF2 gene, arepresent in these cells (Fig. 3). The IGF2 gene, a fetalgrowth factor is normally only expressed from the paternalallele. LOI of IGF2 leading to expression from both parentalalleles can be observed in WTs (15). High expression ofIGF2 mRNA was observed by array analysis in all fiveWilms cell lines and this was confirmed by Q-RT-PCR (Sup-plementary Material, Fig. S1).

WT cell lines with WT1 mutations have a similardifferentiation potential as human MSCs

The morphological similarity of the WT cell lines with MSCsprovoked the question, whether WT cells with WT1 mutations

have a mesenchymal differentiation potential. To address thisquestion, we performed in vitro differentiation experimentsusing established procedures and MSCs as positive controls.After induction of adipogenic differentiation, MSCs showedstrong Oil Red O staining, which labels lipid vesicles.Wilms3 showed a few stained areas (Fig. 4A, panel Wilms3,Oil Red O), whereas the other four Wilms cell lines did notshow any Oil Red O staining (see for example panelWilms1, Oil Red O, Fig. 4A). A low level of FABP4expression, a marker for fat differentiation was observed inuninduced Wilms3 and 5 and a slightly higher level inWilms2. In all Wilms cells except Wilms4, FABP4 expressionincreased after induction of adipogenesis (Fig. 4B).

Terminal chondrocyte differentiation was analyzed bySafranin O staining after growth in pellet cultures for 2–3weeks. Fibroblasts were used as negative controls (data notshown). Wilms1 and 2 could not be induced to terminal chon-drogenic differentiation, whereas Wilms3 could be induced asdemonstrated by positive Safranin O staining (Fig. 4A, panelWilms3, Safranin O). Wilms4 and 5 were not analyzed forthis differentiation pathway. Osteogenic differentiation wasstudied by determining the calcium concentration in Wilmscell culture supernatants in comparison to MSCs as positiveand fibroblasts as negative controls. Wilms1, 2 and 3showed some osteogenic differentiation with increased levelsof calcium, whereas Wilms4 and 5 showed very lowcalcium levels (Fig. 4C).

Next, we investigated the potential of these cells for muscledifferentiation. As positive control, we used human skeletalmuscle myoblasts (HSMM) because we were not able toinduce muscle differentiation in MSCs. Muscle differentiationof Wilms1–5 was initiated with reduced serum concentrationsand the development of multinucleated cells was checked byphase contrast microscopy. HSMM cells showed an almostcomplete conversion to multinucleated cells with concomitantgrowth arrest, whereas all Wilms cell lines continued to growin reduced serum. However, some multinucleated cells weredetected in Wilms1 and Wilms2 and some cells appearedstriated (data not shown). Using an antibody to titin, whichrecognizes a sarcomeric protein of differentiated striated

Figure 3. Loss of heterozygosity (LOH) analysis of Wilms2 cells. CA-repeatmarkers from the indicated positions on chromosome 11 were used to studyblood (B) and tumor (T) DNA from Wilms2. LOH for markers from 11p13to 11p15 was seen and the mitotic recombination occurred between markersin 11p11 (data not shown) and 11p13, as indicated by the crossed lines.This results in two paternal copies of IGF2. The two mutant WT1 copies(labeled with a red star) were also derived from UPD through mitotic recom-bination.

Human Molecular Genetics, 2010, Vol. 19, No. 9 1655

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 6: Wilms tumor cells with WT1 mutations have characteristic features of

muscle, in Wilms1 a few multinucleated cells were stained(Fig. 4A, right panels). Wilms2, 3, 4 and 5 did not show reac-tivity with this antibody, suggesting that a later stage ofmuscle differentiation cannot be achieved in these cells. We

conclude from these results that the WT cell lines with WT1mutations have a limited mesenchymal differentiationcapacity, which varies between the tumors with differentgenetic alterations.

Figure 4. Mesenchymal differentiation potential of Wilms cell lines. (A) Phase contrast microscopy of undifferentiated MSCs, Wilms1 and Wilms3 cells, leftpanels. Wilms1 and Wilms3 cells were stained with Oil Red O after induction of adipogenic differentiation, Oil Red O panels. MSCs served as positive control.Wilms1 and Wilms2 cells were stained with Safranin O after induction of chondrogenic differentiation, Safranin O panels. MSCs served as positive control.Immunofluorescence analysis of Wilms1 and Wilms2 cells after induction of muscle differentiation. Uninduced and induced cells were stained with an antibodyspecific for titin. HSMM served as positive control. Wilms1 cells have a mutant CTNNB1 gene and Wilms2 cells are wild-type for CTNNB1. (B) Semi-quantitative RT-PCR analysis of FABP4 mRNA expression after adipogenic differentiation of the five Wilms cell lines. Left panels, untreated controls, rightpanels, induced cells. The PCR reactions were stopped after 25, 30 and 35 cycles and analyzed by electrophoresis. GAPDH PCR reactions served as internalcontrols. (C) Osteogenic differentiation of WT cell lines. To monitor osteogenic differentiation, calcium concentrations in Wilms cell culture supernatants weredetermined after 3 weeks of induction cycles. MSCs served as positive control, which were taken as 100%. Calcium levels in Wilms cell culture supernatants aredisplayed relative to MSCs.

1656 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 7: Wilms tumor cells with WT1 mutations have characteristic features of

Gene expression profiling reveals a high degreeof similarity between WT cells with WT1 mutationsand human MSCs

To compare the WT cells and MSCs at the level of the tran-scriptome, we performed genome-wide expression analysisusing oligonucleotide arrays (Agilent). The expression profilesof Wilms1–5, MSCs, NK cells and CLS1, a WT cell linewithout a WT1 mutation were compared by average linkagehierarchical clustering of the gene expression list. This analy-sis grouped MSCs with the Wilms cell lines (Wilms1–5) at adistance from NK and CLS1 (Fig. 5). Using correlation coef-ficient and Euclidean metric as distance measurements pro-duced very similar trees. We conclude from these resultsthat the transcriptomes of MSCs and WT cells with WT1mutations have a high degree of similarity.

Next, we asked whether bona fide MSC genes are active inthe Wilms cell lines. Human MSCs express specific surfacemarkers which are commonly used for the identification andisolation of MSCs (2). We thus compared the expressionlevels of various characteristic MSC marker genes on microar-rays. We found that CD49a, CD44, CD73, CD90, CD105 andCD102 genes are strongly expressed in all five Wilms celllines (Fig. 6 and Supplementary Material, Fig. S2). In contrast,CLS1 with a wild-type WT1 gene did not express theseMSC-specific genes, except for CD44 which is also expressedin CLS1 (Fig. 6 and Supplementary Material, Fig. S2). Theexpression of CD73, CD105 and CD90 was confirmed byQ-RT-PCR and a good overall correlation was observedbetween microarray and Q-RT-PCR (Fig. 6). In addition,

FACS analysis further confirmed that CD73 and CD105 pro-teins were expressed on more than 99% of Wilms2, Wilms3,and MSC cells, whereas CD73 was observed on 15% ofCLS1 only and CD105 was not expressed. The MSC-markerCD90 was positive on 80% of Wilms2, 72% of Wilms3,45% of MSC and CLS1 was negative for this marker. TheseFACS results further confirmed the microarray data and theexpression of MSC markers on the WT cell lines. CD34 is amarker for hematopoietic stem cells and CD31 a marker forendothelial cells both of which were not expressed in MSCsand Wilms1–5, however, CLS1 WT cells strongly expressedthese genes (Supplementary Material, Fig. S2). In conclusion,WT1 mutant WT cells are highly related to MSCs.

To gain more specific information about the similarity ofWT cells with MSCs, we analyzed their gene expression pro-files and identified a shared gene signature. The Venn dia-grams (Fig. 7A) show that 2774 genes are expressed at ahigher level, whereas 2708 genes are expressed at a lowerlevel in comparison to CLS1 cells (Fig. 7A). We then per-formed an ontological analysis on the up-regulated genesusing EASE and identified a highly significant enrichment ofspecific GO terms (P , 0.001) in this gene set. The enrichedGO terms are: anatomical structure development, multicellularorganismal development, anatomical structure morphogenesis,developmental process, system development, organ develop-ment, organ morphogenesis and multicellular organismalprocess. Genes from the shared signature with an at least500-fold difference to CLS1 are shown in a heat map(Fig. 7B). We also performed a gene set analysis of theshared gene signature using Globaltest and identified a signifi-cant enrichment of GO terms relating to diverse signalingpathways (Fig. 7C). The heat maps show the expressionlevels of representative pathway genes in NK (N), CLS1 (C)and the Wilms cell lines (Wilms1–5). A large number ofgenes from the TGFb signaling pathway are expressed inthe shared signature, e.g. TGFBR2, INHBA, DCN, THBS1,THBS2, FST, SMAD7, TGFB1, BMP4, ACVR1, RHOA andMAPK1 (Fig. 7C, panel TGFb). The shared gene signaturealso contains significant numbers of genes from the Wnt,Hedgehog, MAPK, VEGF, mTOR and JAK-STAT signalingpathways (Fig. 7C). We expect that the expression of signalingpathway genes is correlated with pathway activity and futurework will investigate this issue.

It is not possible to definitely identify the cells from whichWTs originate in the tumor tissue of WT patients, as thesetumors are heterogeneous and show diverse differentiationpatterns. Here, we searched for alternative approaches toclarify this issue. A recent study presented the induction ofPAM with the generation of multipotent mesenchymal pro-genitors using ES cells that were cultivated on murine OP9stromal cells (38). In this work, the mesenchymal precursorswere isolated by flow sorting using a CD73-specific antibodyand subsequently analyzed by genome-wide expression profil-ing. By comparing MSCs derived from embryonal stem cellswith primary adult MSCs a specific gene signature consistingof 412 genes was identified. We compared the gene expressionprofiles of Wilms1–5 with this MSC-specific gene list andfound that the majority of MSC-specific genes were alsoexpressed in the five Wilms cell lines (SupplementaryMaterial, Table S1). These data further support the similarity

Figure 5. Gene expression analysis reveals a high degree of similarity betweenWT cells with WT1 mutations and mesenchymal stem cells. Hierarchicalcluster analysis of gene expression data from Wilms cell lines, MSCs, CLS1and NK. The numbers on the left side of the branched trees are approximatelyunbiased P-values. The numbers on the right side of the branched trees rep-resent bootstrap probabilities. Values above 95 indicate that the observedbranch is significant.

Human Molecular Genetics, 2010, Vol. 19, No. 9 1657

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 8: Wilms tumor cells with WT1 mutations have characteristic features of

of the WT cell lines to MSCs, suggesting that WTs with WT1mutations originate from cells of the mesenchymal lineage.

WT cells with WT1 mutations express typical molecularmarkers of PAM

Murine embryonal stem cells have been used for guided differ-entiation into three germ layers (39). This enabled the gener-ation of mesoendoderm, neuroectoderm, lateral platemesoderm and PAM in tissue culture. The different celltypes were sorted by flow cytometry using specific surfacemarkers and analyzed by genome-wide expression profiling.We used data from the published gene lists for mesoendoderm(CDH1þ/PDGFRAþ), neuroectoderm (SSEA12/KDR2/PDGFRA2), lateral plate mesoderm (CDH12/KDRþ/PDGFRA2) and PAM (CDH12/KDR2/PDGFRAþ) (39)and compared these with our gene expression data from thefive Wilms cell lines and MSCs. This analysis revealed thatWilms1–5 cells are negative for the mesendoderm markerCDH1 and the lateral plate mesoderm marker KDR/FLK1and positive for the paraxial marker PDGFRA. The immuno-fluorescence analysis of Wilms3 with PDGFRA showed thatall cells express this surface receptor (Supplementary

Material, Fig. S3A). Although there is some overlap in thegene expression profile between these lineages, the markerprofile expressed in the WT cells with WT1 mutations allo-cates these to PAM. The gene list of specific PAM cellsincludes PDGFRA, PDGFRB, MSX1, RUNX2, TWIST2,TBX4, TBX20, HAND1, DOK4 and several collagen genes aswell as the anterior–posterior markers HOXA10, HOXA11,HOXB6, HOXC10 and HOXD1. With the exception ofTBX4, TBX20 and HAND1, these PAM-specific genes arealso expressed in the Wilms1–5 cells and a heat map of theexpression of paraxial marker genes is shown in Supplemen-tary Material, Figure S3B.

As it was recently shown that kidney stromal cells arederived from PAM (24), we studied the expression of kidneystromal markers. It is known that cortical and medullary inter-stitial cells in E15.5 mouse kidney express Meis1 (40). Usingthe information available through the comprehensive geneexpression analysis of individual compartments of day E15.5mouse kidneys (40), we found that the Meis1 positive cellsalso express FoxD1 as well as Wt1 (Supplementary Material,Table S2). MEIS1 and FOXD1 are expressed in all Wilmscell lines as well as in MSCs. The expression of FOXD1was confirmed by Q-RT-PCR and a good correlation

Figure 6. Expression of characteristic mesenchymal stem cell markers. On the left, the RNA expression analysis of specific mesenchymal stem cell markersCD73, CD90 and CD105 in WT cell lines by microarray and validation by Q-RT-PCR is shown. The surface expression of CD73, CD105 and CD90 ofCLS1, MSC, Wilms2 and Wilms3 was analyzed by flow cytometry (FACS). Staining with isotype controls is indicated by gray histograms and the percentageof positive cells is indicated.

1658 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 9: Wilms tumor cells with WT1 mutations have characteristic features of

between the microarray data and Q-RT-PCR was observed(Supplementary Material, Fig. S1). The expression ofstromal marker genes and molecular markers of PAMsuggest that the tumor initiating cells for WTs with WT1mutations reside in PAM.

Identification of a WT-specific gene signature

Next, we wished to identify genes whose expression is specificfor the WT cells. We thus compared the gene expression pro-files of Wilms1–5 cells with that of MSCs and identified aWT-specific gene signature (WT signature). Microarray

analysis revealed a significant differential expression of 721genes between MSCs and Wilms1–5 cells. Of these differen-tially expressed genes, 322 had an at least 1.8-fold higher and399 an at least 1.8-fold lower expression level in Wilms1–5.A GO term enrichment analysis using EASE showed thatgenes from the tube development pathway were significantlyup-regulated including EDNRA, BMP2, BMP4, HHEX,SIM2, RHOJ, FOXF1, TBX3, PAX3 and LAMA5. Otherenriched GO terms not reaching statistical significance relateto organ morphogenesis (P-value 0.06), regulation of astrocytedifferentiation (P-value 0.085) and mesenchymal development(P-value 0.088). GO terms relating to keratin filament and

Figure 7. Identification and analysis of a gene signature shared by WT cells and mesenchymal stem cells. (A) A shared gene signature between WT cells withWT1 mutations and MSCs was identified by comparing their transcriptomes with CLS1 cells. The Venn diagram (upper panel) indicates the number of genes thatare significantly upregulated and the Venn diagram (lower panel) indicates the number of genes that are significantly downregulated in the shared signature. (B)The heatmap shows the genes from the shared signature whose expression levels are .500-fold higher or ,500-fold lower than in CLS1 cells. (C) Heatmaps ofthe expression levels of genes from the shared signature which were assigned to specific signaling pathways by Globaltest analysis. Individual signaling pathwaysare indicated on top of each heatmap. Details are given in the text. N, normal kidney cells; C, CLS1 WT cells with wild-type WT1; M, MSCs; 1–5 indicates theWT cell lines with mutant WT1.

Human Molecular Genetics, 2010, Vol. 19, No. 9 1659

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 10: Wilms tumor cells with WT1 mutations have characteristic features of

multicellular organismal development were significantlyenriched in the group of 399 genes with strongly reducedexpression levels in WT cells.

In order to get a deeper insight into the function of the WTsignature, we used a knowledge driven approach and extractedbiological information about individual genes by screening theliterature. On the basis of this comprehensive analysis, a largenumber of genes could be assigned to specific functionalgroups and these data are depicted in Table 2. We noted thatthe WT signature consists of a large group of genes encodingtranscription factors that play a role in embryonic develop-ment or pattern formation and another large group of geneswith a function in neurogenesis, axon development and axonguidance. Other functional groups relate to the cell cycle,diverse signaling pathways, blood vessel development,muscle differentiation and other biological processes(Table 2).

The most highly expressed genes from the WT signatureinclude PAX3, a gene expressed in the presomitic and PAM,developing somites, limb muscle, neural crest and neuraltube. The expression of this gene was confirmed byQ-RT-PCR (Supplementary Material, Fig. S1). PAX3 is alsoa master regulator of myogenesis. Other highly expressedgenes are HOXD10, which controls skeletal developmentand HOXD11 which is expressed in metanephric blastema ofthe kidney. Additional genes from the WT signature are alsoregulators of kidney development, for example the transcrip-tion factors HOXA11, HOXC11, SALL1, TCF21 and thebone morphogenic proteins, BMP2 and BMP4. The role ofHOXD11/HOXA11 in kidney development was demonstratedby double-knockout mice which display variable kidney hypo-plasias (41).

Other highly expressed genes have a function in malignanttumors. For example ARHGDIB, a Rho guanosindiphosphatdissociation inhibitor which affects tumor growth and malig-nant progression of gastric carcinomas, the DNA methyltrans-ferases DNMT1 and DNMT3B which are up-regulated in thecolorectal adenoma–carcinoma sequence, and ECT2 whichis over-expressed in glioblastomas where it is associatedwith poor patient survival. GLI and GLIS1 regulate stem cellproliferation and Hh signaling in melanomas and gliomas,IGFBP3 is highly expressed in pancreatic cancer and osteosar-comas, PIK3C2B a component of the PI3K signaling pathwayplays a role in oncogenic transformation, controls cell survi-val, cell migration and intracellular protein trafficking. Otherover-expressed genes include the oncogene PIM3, RAB15,STMN3 and TBX3, the latter is aberrantly expressed in asubset of breast cancers, as well as TWIST2 an inducer of epi-thelial to mesenchymal transition in tumors. PRAME andDSCR8 are tumor antigens, whereas MMP1 is responsiblefor breakdown of the extracellular matrix and tumor growth.It is important to note that PRAME (preferentially expressedin melanoma) is highly expressed in a wide variety ofcancers and is a prognostic marker for clinical outcome (42).

Coexpression of genes from the WT1 and/or CTNNB1mutant WT signature in WT cell lines

WTs with WT1 mutations show diverse differentiation patternsand for this reason the tumor tissue consists of heterogeneous

Table 2. WT signature genes

Gene symbol Fold change Gene symbol Fold change

Transcriptional regulation, transcription factor activityEVT4 5.4 MKX 6.1ETF7 3.2 NFE2L3 11.1FOXF1 10.3 NR2F1 3.6FOXL2 14.8 NR2F2 7.3GLI1 6.5 PAX3 102GLIS1 4.4 PLAG1 2.4HHEX 8 SALL1 17.4HMGA1 3.1 SALL2 3.5HOXA11 5 SIM2 17.1HOXA11S 13.1 SNFT 3.7HOXD1 10.8 TBX3 5.4HOXD10 350 TCF19 2.4HOXD11 122 TCF21/POD1 11.2HOXD9 7.8 TWIST2 5.1KLF5 6.2 ZNF521 9.4LZTS1 2.7

Neurogenesis, axon development, axon guidanceCDH18 36.6 NPAS2 5.5CDH4 3.6 NRGN 3.6CRMP1 7.7 OTXR 15.7DICS1 2.7 RGNEF 3DNM1 8.2 ROBO1 2.DTL 2.2 ROBO4 5.1GLI1 6.5 RTN1 9.7GLIS1 4.4 SEMA3D 30.3GREM1 9.8 SGIP1 5.7GPR162 2.6 SHB 2.4GPRC5P 5.7 SHC3 81.1GRASP 7.3 SIM2 17.1KLF5 6.2 STMN3 5.7MARCH4 2.6 SYNGR1 3.7NBEA 3.1 SYTL5 44.9NFE2L3 11.1 UCHL1 2.1

Cell cycleAURKB 2.7 KIF4A 2.4CDC20 2.6 KPNA2 2.4CDC25B 2.1 MCM2 2.3CDCA4 2 MCM7 2.2E2F7 3.2 NASP 5.5ECT2 2.1 PCBP4 1.9GTSE1 2.5 SEPT6 2.5KIF22 2

Wnt signaling, Wnt targets and antagonistsAPCDD1 149 PRICKLE2 2.1CRMP1 7.7 RANGAP1 2.1DKK3 (11p15) 2.6 SFRP1 14.3DKK2 28.7 TLE1 2.8FZD2 2.8 TLE4 2.1

Planar cell polarityDCHS1 11.9PRICKLE2 2.1

CiliogenesisARMC4 14.8

G-proteins, RAS/RAC signaling, GAP and GEPsARHGAP19 2 RANGAP1 2.1ARHGAP 29 4.7 RGNEF 3ARHGDIB 7.9 RHOJ 3.4DEF6 15.6 SGIP1 5.7DUSP4 4.1 SH2D5 8ECT2 2.1 SH3MD4 2.8GPR162 2.6 SHB 2.4GPRC5B 5.7 SHC3 81.1RAB15 3.4 SYTL5 44.9

Oncogene/overexpressed in tumors or metastasis relatedARHGDLB 7.9 SEMA3D 30.3DSCR8 172 STIP1 2.2

Continued

1660 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 11: Wilms tumor cells with WT1 mutations have characteristic features of

cell populations. In contrast, WT-derived cell lines represent ahomogeneous cell population. It is evident that microarraydata derived from WTs correspond to the average geneexpression pattern of the different cell types present in thetumor and the differentiation states of these cells. In order toinvestigate whether the WT1 mutant cell lines expresssimilar genes as WTs in vivo, we compiled a list of genescharacteristic for WTs with WT1 and/or CTNNB1 mutationsfrom existing microarray data (13,43–45). It is surprisingthat comparing the gene lists from these four publicationsnot a single gene is present in all four tumor signatures (Sup-plementary Material, Table S3). This could be explained bythe various array formats that were used or by different treat-ment regimens of the analyzed WTs (13,43–45). We foundthat 194 named genes of this merged gene list were alsopresent on our array format. Importantly 140 (72%) of theseWT signature genes are either expressed in all five or at

least in one of the Wilms cell lines (Supplementary Material,Table S3). Of note seven of these genes that are coexpressed intumors in vivo and the Wilms cell lines are PAM marker genes(Supplementary Material, Table S3, highlighted in blue). Wealso observed that WTs in vivo express a number of genesrelated to muscle differentiation, osteogenesis and endothelialcell function, which are not expressed in the Wilms cell lines(Supplementary Material, Table S3, highlighted in green andbrown). This could be explained by the fact that the Wilmscell lines have features of multipotent mesenchymal precur-sors and thus do not express differentiation specific genes. Insummary, these results demonstrate the overall similarity ofthe characteristic gene expression profile of the WT1/CTNNB1 mutant WTs in vivo with the established WT1mutant cell lines. Several genes were selected from this listand were analyzed by Q-RT-PCR using RNA from all celllines and for comparison RNA isolated from tumor materialof Wilms1, Wilms2 and Wilms4. Overall, a good correlationwas found between microarray data and Q-RT-PCR forAXIN2, DKK2, MET, SIM2 as well as PDGFRA, a markerfor PAM, which is also highly expressed in MSCs (Sup-plementary Material, Fig. S4).

b-Catenin/TCF-dependent Wnt signaling in WT cell lines

As Wilms1, 2 and 4 cells have a mutant CTNNB1 gene, weanalyzed whether b-catenin/TCF-dependent transcriptionalactivity is affected by these mutations. To address this issue,we used the TOPflash reporter assay, which measuresb-catenin-dependent Wnt signaling in living cells. As negativecontrol, we used the FOPflash reporter construct with mutantTCF4 consensus binding sites. After transfection of TOPflashand FOPflash constructs into the Wilms cell lines, the TOP/FOP ratio was determined. Wilms1, 2 and 4 showed a TOP/FOP ratio between 4 and 6.5, whereas Wilms3 and 5 with awild-type CTNNB1 gene had a ratio of 1 and 0.5, respectively.These data demonstrate that the b-catenin-dependent Wnt sig-naling pathway is activated in Wilms cell lines with CTNNB1mutations (Fig. 8A).

Activation of the Wnt signaling pathway causes nuclearaccumulation of b-catenin protein. We therefore analyzedthe subcellular localization of b-catenin by immunofluores-cence assays and immunoblotting. All five Wilms cell linesshowed cytoplasmic b-catenin and small amounts ofb-catenin in the nucleus as well as in cell–cell contacts(Fig. 8B). Very strong nuclear staining was observed inSW80 control cells (data not shown). The immunofluores-cence results were confirmed by western blot analysis whichdetected b-catenin in the nucleus and cytoplasm in allWilms cell lines (Fig. 8C).

To get an insight into the consequences of mutant b-cateninfor Wnt signaling-dependent gene regulation, we comparedthe gene expression profiles of the Wilms cells with mutantCTNNB1 mutations to those with a wild-type gene. Surpris-ingly, only 15 named genes were differentially expressed(Table 3). We identified AXIN2 in the group of genes whichare up-regulated in CTNNB1 mutant cells and confirmed thedifferential expression by Q-RT-PCR (SupplementaryMaterial, Fig. S4, higher expression in Wilms1, 2 and 4).AXIN2, is a Wnt target gene with a role in negative-feedback

Table 2. Continued

Gene symbol Fold change Gene symbol Fold change

ECT2 2.1 STMN3 5.7PIK3C2B 3.2 TMSL8 7.8PIM3 1.8 TWIST2 5.1PRAME 416 USP36 3

Endothel/angiogenesis/haematopoietic cell growthDKK3 2.8 PPP2R2B 23.2EDNRA 5.2 ROBO1 2.FLT1 19.7 ROBO4 5.1LTK 15 SHB 2.4NOSIP 2.4 SINK5 23.7NOX4 4.6 ZNF521 9.4

Extracellular matrixCOL5A5 14.4 ITGA4 4.9COL13A1 13.6 LAMA5 7.1COL18A1 10.4 MMP1 630

Hh signaling/target geneBMP2 8.6 GLI1 6.5BMP4 8.1 GLIS1 4.4GREM1 9.8 SFRP1 14.3

Cell–cell signaling/adheren junctionsBMP2 8.6 PCDHB9 6.9PCDHA1 3 PVRL2 2.9PCDHA11 9.7 SSTR1 51PCDHB11 4 SYT1 34.6PCDHB8 4.8

TGFb signalingBMP2 8.6 PPP2R2B 23.2BMP4 8.1ZNF521 9.4

RA signalingDTL 2.2 GRASP 7.3GPRC5B 5.7 KLF5 6.2

AdipogenesisFADS2 3.2

Chondrogenesis/osteogenesisTWIST2 5.1

Regulation of muscle differentiationPAX3 MYOZ3 2.9FKBP1B 2.3 SIM2 17.1

TWIST2 5.1

Manually curated and selected list of up-regulated genes, fold change versusMSC. The same genes may appear in more than one functional category, geneswith a high expression difference between Wilms cell lines and MSC arehighlighted in bold.

Human Molecular Genetics, 2010, Vol. 19, No. 9 1661

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 12: Wilms tumor cells with WT1 mutations have characteristic features of

regulation of Wnt signaling. In contrast, PAX8 expression isup-regulated in Wilms cells with a wild-type CTNNB1 gene.PAX8 participates in the regulation of mesenchymal–

epithelial transition in the kidney. Interestingly, two of theunknown transcripts expressed at a lower level also map tothe PAX8 gene locus but in an antisense orientation.

The Wnt-signaling pathway is known to be involved in WTswith WT1 and CTNNB1 mutations. We therefore askedwhether WTs with WT1/CTNNB1 mutations express thesame Wnt target genes as our Wilms cell lines. To investigatethis issue, we used a list of b-catenin/Wnt target genes fromthe Wnt homepage (www.stanford.edu/~rnusse/wntwindow.html) as well as new Wnt target genes described in the litera-ture and checked their expression levels on microarrays. Wethen compared our expression data with existing data of high-density microarrays of WTs (43,44) and found that most Wnttarget genes that are expressed in WTs in vivo are alsoexpressed in Wilms1–5 cells (Supplementary Material,Table S4). These results show that the WT cell lines faithfullyreflect the in vivo situation regarding activity of the Wnt sig-naling pathway. The establishment of WT cell culturemodels is an important prerequisite for further investigationsof WTs with WT1 mutations.

DISCUSSION

We report here for the first time the successful establishmentof long-term homozygous WT1 mutant cell cultures derivedfrom WTs with WT1 mutations. This was achieved by usinga tissue culture medium specifically developed for thegrowth of MSCs. This new method is of more general impor-tance as we were able to establish cell lines from five individ-ual WTs with diverse WT1 mutations. It is interesting to notethat long-term cell lines could not be established from WTswith a wild-type WT1 gene using MSCGM, demonstratingthat these tumors are biologically different (unpublished data).

Figure 8. Activation of the Wnt signaling pathway in WT cells with mutant CTNNB1 genes. (A) Transient transfection assay of the five Wilms cell lines with TOP-and FOPflash reporter plasmids and analysis of luciferase activity. The TOP to FOP ratio is displayed relative to the internal renilla control plasmid. Standard devi-ations were derived from three independent experiments. (B) Immunofluorescence analysis of b-catenin expression MSCs, Wilms2 and Wilms3 cells. b-catenin wasvisualized after incubation with an antibody specific for b-catenin followed by a fluorochrome-conjugated anti-rabbit secondary antibody. The cell lines and CTNNB1mutation status are indicated on top of each panel. Magnification �40. (C) Western blot analysis of b-catenin expression in the five WT cell lines. After incubationwith an antibody specific for b-catenin, the same blot was used to detect alpha-tubulin which is a cytoplasmic protein. c, cytoplasm; n, nuclear extract.

Table 3. Differentially down- and up-regulated genes in CTNNB1 mutant andwild-type WT cell lines

Gene Description Meanfc

Down-regulated genes in CTNNB1 mutant WTs (Wilms 1, 2 and 4)LOC654433 cDNA clone IMAGE:5266296 285.1AK126431 cDNA FLJ44467 fis, clone UTERU2025891 242.4NGEF Neuronal guanine nucleotide exchange factor 214.7PAX8 Paired box gene 8, transcript variant PAX8A 214.2LPAAT-THETA Lysophosphatidic acid acyltransferase theta 29.1LOC387895 cDNA clone IMAGE:6160413 28.2F2RL2 Coagulation factor II (thrombin)

receptor-like 227.1

BF217859 cDNA clone IMAGE:4103859 26.7TUBA2 Tubulin, alpha 2, transcript variant 2 22.3

Up-regulated genes in CTNNB1 mutant WTsPSCD3 Pleckstrin homology, Sec7 and coiled-coil

domains 32.3

MX2 Myxovirus (influenza virus) resistance 2(mouse)

5.5

PPFIBP2 PTPRF interacting protein, binding protein 2(liprin beta 2)

5.3

AXIN2 Axin 2 (conductin, axil) 5.6TUBB2B Tubulin, beta 2B 6.4HOXB3 Homeobox B3 6.5AXIN2 Conductin mRNA, complete cds 9.3IGFBP2 Insulin-like growth factor binding protein 2,

36 kDa12.1

TFAP2C Transcription factor AP-2 gamma 8.5RGC32 Response gene to complement 32 18.8WFDC1 WAP four-disulfide core domain 1 24.1CALB2 Calbindin 2, 29 kDa (calretinin) (CALB2) 37.5THC2338229 Unknown 44.7

1662 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 13: Wilms tumor cells with WT1 mutations have characteristic features of

Remarkably in four of our Wilms cell lines the homozygousmutant WT1 alleles are due to a similar mitotic recombinationevent between 11p11 and 11p13 leading to UPD from 11p11!11pter. This resulted in paternal UPD in two cases analyzedand consequently expression of the imprinted IGF2 gene in11p15 from both alleles. Only one cell line derived from theWAGR patient had retained both parental copies of the shortarm of chromosome 11.

The five Wilms cell lines can be grown for prolonged timein culture, irrespective of the presence or absence of an acti-vated mutant b-catenin, suggesting that the mutant WT1protein enables long-term survival. It is evident that themutant WT1 proteins cannot bind to their normal targetsequence in DNA, because they lack intact ZF domains.However, they have an intact N-terminal region which isrequired for protein–protein interactions. We showed herethat Wilms3 cells express a mutant WT1 protein which is mis-located in the cytoplasm. It is a likely possibility that mutantWT1 proteins fulfill novel functions which are not related totheir DNA binding activity. It has been shown that completeinactivation of Wt1 in knockout mice was associated withapoptosis of mesenchymal cells (46). In contrast, the Wilmscell lines with mutant WT1 are viable and do not show anysigns of apoptosis. It can be concluded from these resultsthat mutant WT1 is biologically active and therefore thesemutations can be classified as gain-of-function mutations. Inthis context, it is interesting to note that siRNA-dependentinhibition of Wt1 in kidney organ cultures isolated at day 11and cultured for 72 h resulted in abnormal proliferation (47).It is evident from these results that WT1 has a role in apoptosisand cell cycle regulation. We consider it possible thatgain-of-function mutations of WT1 may have an oncogenicfunction independent of b-catenin.

Origin and nature of WTs with WT1 mutations

WTs with WT1 mutations have stromal-type histology, but it isunclear where these WTs originate. A characteristic feature ofWTs with WT1 mutations is the presence of ectopic mesench-ymal differentiation, predominantly skeletal muscle but alsofat, and rarely cartilage and bone, reminiscent of humanMSCs and known derivatives of PAM. Here, we used a com-bination of approaches for an experimental analysis of theorigin and nature of WTs with WT1 mutations. A comparisonof genome-wide expression profiles showed that the WT celllines cluster with MSCs at a distance from NK cells orCLS1, the WT cells without a WT1 mutation. This analysisprovided the first evidence that WT cells are related toMSCs. We then analyzed a gene signature specific for multi-potent mesenchymal precursors which were derived fromhuman embryonic stem cells after the induction of PAM intissue culture (38) and showed that the majority of thesegenes were also expressed in the five Wilms cell lines. More-over, the Wilms cell lines express RNA and protein of thehighly specific MSC marker genes CD73, CD90 and CD105.To further substantiate the MSC-like nature of WT cellswith WT1 mutations, we induced adipogenic, chondrogenicand osteogenic differentiation as well as muscle differen-tiation. Our results demonstrate that the Wilms cell lineshave a limited capacity to differentiate into some of these

mesenchymal lineages. This reflects the in vivo situationquite well, as WTs with WT1 mutations show only incompletemesenchymal differentiation patterns. Collectively, these datasuggest that WTs with WT1 mutations could originate frommultipotent mesenchymal precursor cells of the PAMlineage. Similar evidence came from a comparison of geneexpression data from Wilms cell lines with existing geneexpression data from kidney development. Although theWilms cell lines expressed only a limited number of kidneyspecific genes, they all expressed the stromal marker genesFOXD1 and MEIS1.

The origin of stromal cells in the kidney has only recentlybeen resolved. It was reported that stromal cells in thekidney are derived from a pax3þ domain of PAM and notfrom the intermediate mesoderm as all other cells of theadult kidney (24). However, it is important to note that inPax3Cre/þ, R26R mice, b-galactosidase is also expressed incells that are derivatives of the metanephrogenic mesenchymeand therefore Pax3 expression is not exclusively found instromal cells (25). It is impossible to obtain PAM fromhumans; however, human embryonal stem cells can bedifferentiated into diverse germ layers including PAM (39).A PAM-specific gene set was derived after guided differen-tiation of ES cells and flow sorting of PAM cells with specificsurface markers (39). We analyzed the PAM cell-specific signa-ture in the five Wilms cell lines and detected a highly significantoverlap. By using this approach, we could show expression ofparaxial mesodermal genes, demonstrating their close relation-ship to this embryonal tissue. A low level of Wt1 is expressed inPAM derived from ES cells (S. Kume, personal communi-cation). Of note, we found that Wt1 is expressed in cortical(nephrogenic) and medullary interstitium, in the same compart-ment that expresses Meis1 and FoxD1, corresponding to stromalcells of the kidney (40 and Supplementary Material, Table S2).These data show that stromal-type WTs are related to kidneystromal progenitor cells, which originate in PAM. Our results,however, do not definitely prove that WTs originate in PAM.It is also a possibility that WTs initiate in immature MM andloss of a functional WT1 by mutation may change the fate ofthese cells towards the stromal lineage. Future work is requiredto resolve this issue.

The predominant differentiation type in stromal-type WTsis a rhabdomyogenic pattern. PAX3 is necessary and sufficientto induce myogenesis in MSCs (48). In addition, Pax32/2

mice have multiple skeletal defects, a delay in muscle differ-entiation and a decrease in muscle mass. Pax3 is essential forthe expression of Six1 and Eya2 during skeletal myogenesis(49), all three genes being expressed in the Wilms tumorcell lines. The expression of these genes in the WT celllines with WT1 mutations might explain their predominantrhabdomyogenic differentiation pattern in vivo and theirlimited muscle differentiation potential in vitro. Expressionof PAX3 in WT samples with myogenic differentiation aswell as in MM and in the stromal compartment of thedeveloping mouse kidney was recently demonstrated byimmunohistochemistry (50). As PAX3 promotes survival inembryonic and malignant cells and decreased PAX3 leads tocell death in embryonal rhabdomyosarcoma, melanoma andmyoblast cells, this gene may have an important contributionfor (tumor) cell survival.

Human Molecular Genetics, 2010, Vol. 19, No. 9 1663

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 14: Wilms tumor cells with WT1 mutations have characteristic features of

WT gene signature

The WT signature contains 322 up-regulated genes and thelargest group of genes belongs to the class of transcriptionfactors including six HOX genes. HOXD10 is expressed350-fold higher in WTs than MSCs and PAX3 102-foldhigher. This demonstrates that, despite their similarity withPAM and MSCs, the tumor cells also show significant differ-ences regarding the expression of transcription factors. It isevident that a tumor-specific transcriptional program is acti-vated in the Wilms cell lines and future work is needed toinvestigate the impact of these transcription factors for WTswith WT1 mutations. Another large group of genes has arole in neurogenesis, axon development or axon guidance, fol-lowed by genes involved in G-protein signaling. Ontologicalanalysis of the WT signature revealed tube development as asignificantly up-regulated GO term. Of note, the WT signaturecontains a number of genes which have been found to behighly expressed in tumors or are known members of onco-gene families. It is interesting that among these are severalgenes that are associated with poor prognosis or metastaticpotential; both properties are not usually an attribute of WT1mutant tumors.

b-Catenin/TCF-dependent Wnt signaling in CTNNB1mutant WTs

The role of b-catenin/TCF-dependent (canonical) Wnt signal-ing for nephron development and branching nephrogenesis iswell established (51,52). Canonical Wnt signaling wasobserved in epithelia of the branching UB and in mesenchymeduring transition into renal tubules and is rapidly down-regulated in maturing nephrons and becomes undetectable inpostnatal kidney. In transgenic animals with a Ctnnb1gain-of-function mutation, mesenchymal-to-epithelial tran-sition of induced renal progenitors is blocked by the continuedb-catenin activity (53). Therefore, a regulated activation withensuing inactivation of Wnt signaling is critical for normalnephrogenesis. Furthermore, Wnt signaling is implicated invertebrate development. During morphogenesis, massive cellmovements take place which establish and shape the germlayers (54). The Wnt/planar cell polarity pathway (PCP) regu-lates specific cell behaviors and the migration of paraxial andlateral plate mesoderm cells (55). Wnt5a, a ligand acting inPCP and the downstream target prickle are involved in cellmigration from the posterior streak; in the anterior streakthis is antagonized by Wnt3a to generate the non-migratorymedial mesoderm (56). WNT5a as well as PRICKLE2 arehighly expressed in the Wilms cell lines and therefore thesecells may have a defect in the cross talk with other cells.Recent data showed that Wnt signaling is a major target ofWT1 by competition of WT1 with TCF4 for binding to CBP(58). As in all the developmental processes Wnt signaling isonly temporarily active and if WT1 might have a role in itsdown-regulation, this would suggest that tumor developmentis caused by an inappropriately active Wnt signaling due tolack of repression by inactive WT1.

Another novel observation from our studies is that only 15of the 20 000 expressed genes show a significantly differentlevel in the CTNNB1 mutant and wild-type tumor cells. One

of the genes expressed at a higher level in the mutant cellsis a well-known TCF/b-catenin target gene, AXIN2, a negativeregulator of Wnt signaling. In contrast, PAX8, with a knownfunction in kidney development is up-regulated in CTNNB1wild-type WT cell lines. This small number of differentiallyexpressed genes in CTNNB1 mutant versus wild-type tumorcell lines suggests that the activation of other signaling path-ways is also important. Indeed, our comparative pathwayanalysis demonstrated that in the shared transcription profileof the five WT cell lines with MSCs, many members of thetransforming growth factor-b, mitogen-activated proteinkinase, calcium, Hedgehog, JAK-STAT, p53, mTOR and vas-cular endothelial growth factor signaling pathways areexpressed. If this gene expression pattern reflects pathwayactivity, WT cells and MSCs are regulated by the same signal-ing pathways.

Outlook

The establishment of long-term cell cultures derived fromWTs with WT1 mutations is essential for experimentalapproaches to unravel the impact of WT1 and CTNNB1 forthe development of WTs. The Wilms cell lines are a noveltool to study the function of genes over-expressed in the WTsignature and they allow to investigate the role of wild-typeand mutant WT1 for differentiation and Wnt signaling. Adetailed understanding of the biology of WTs with WT1mutations is an essential prerequisite for developing efficienttherapies.

MATERIALS AND METHODS

Patient and tumor characteristics

Patient numbering is identical to the cell line numbering. Allcell cultures were initiated from fresh WT samples obtainedfrom 1998 to date and only those with a germ line or tumor-specific WT1 mutation were included in this study. Patient 1was described previously (36). Briefly, the girl presentedwith bilateral tumors at 1 year of age with fetal rhabdomyoma-tous histology. The tumors did not respond to the SIOP-2001pre-surgery chemotherapy (4 weeks of Act D and VCR) andwere removed by kidney sparing surgery. After 1 yearwithout further cytotoxic treatment, she developed again bilat-eral tumors. The right tumor showed triphasic histology,whereas the left tumor showed fetal rhabdomyomatous his-tology and the cell culture was established from the lefttumor. Patient 2 presented at 1 year of age with unilateral,classical triphasic WT which was completely resectedupfront. In addition, this male patient had bilateral cryptorch-idism.

Patient 3 developed a unilateral WT at 11.5 months. He waspreoperatively treated according to SIOP-2001/GPOH proto-col (4 weeks of Act D and VCR). The stromal type tumorshowed fetal rhabdomyomatous histology. Patient 4 wasborn with bilateral cryptorchidism and bilateral corneal opaci-ties. A retarded psychomotor development was noticed, and atage of 11 months he presented with a renal mass in onekidney. The diagnosis of WAGR syndrome was suspectedbased on mental retardation, sporadic aniridia associated

1664 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 15: Wilms tumor cells with WT1 mutations have characteristic features of

with glaucoma and cryptorchidism. Cytogenetic analysis onperipheral blood lymphocytes demonstrated a 46, XY,del(11)(p13) karyotype, a deletion encompassing the WT1and PAX6 genes. Upon diagnosis, a partial nephrectomy ofthe lower pole of the left kidney was performed. Pathologyshowed classic nephroblastoma and multiple foci of intralobarnephroblastomatosis in the surrounding tissue. The biopsy ofthe right kidney showed abnormal tubular and vascular struc-tures with an interstitial inflammatory reaction which overallwas compatible with dysplastic features; there was no evi-dence of nephrogenic rests or tumor.

Patient 5 had hypospadias and developed unilateral WT atthe age of 2 years. He was preoperatively treated accordingto SIOP-2001/GPOH protocol (4 weeks of Act D and VCR).Pathology reported a tumor-like lesion of the kidney corre-sponding to a massive intralobar nephroblastomatosis with arhabdomyoblastic stroma component.

Cell culture

In Duesseldorf, the tumor cell culture was initiated by manualdissection of the tumor with scissors into 1 mm cubicles,which were placed into the respective growth media, withoutdisturbing for 7 days. Half of the medium was replaced withfresh growth medium every 3 days until outgrowth wasobserved. After that the medium was changed every 3 daysand subculturing was started after cells became almost conflu-ent. In Barcelona, the tumors were enzymatically digested andthe culture was initiated in DMEM. After the first passage, thecells were shipped to Germany. Thereafter, cells were kept atlow density with a medium change every 3 days. Growthmedium was either WT medium as described (27) or HumanMSCGM (Lonza). The following cell lines were purchased:human MSCs (Lonza), HSMM (Lonza) and WT-CLS1 (CellLines Services). The study was approved by the local EthicsCommittee (Nr. 2617) and parents gave written consent thatleftover tumor material can be used for research.

Mutation and LOH analysis

All WT1 exons were analyzed for mutations after amplificationusing the Expand High fidelity PCR system (Roche) and dena-turing high performance liquid chromatography (WAVE,Transgenomic) as a pre-screening method (primers and con-ditions in Supplementary Material, Table S5). To get anoptimal amplification of exon 1, 0.5 M Betain (Sigma) wasadded. Exons showing aberrant patterns were sequenced onan ABI 3100 automated capillary DNA sequencer (AppliedBiosystems) using cycle sequencing procedure and theBigDye terminator kit. Exon 3 of CTNNB1 was analyzed asdescribed before (11). LOH analysis was performed onblood and tumor DNA from the same patient. Four CA-repeatmarkers from chromosome 11p and q were used (primersequences and PCR conditions are described in SupplementaryMaterial, Table S6).

Protein extraction and western blot analysis

For preparation of nuclear and cytoplasmic extracts, 1 � 107

cells were washed once in PBS containing 1 mM MgCl2,

resuspended in hypotonic buffer (10 mM HEPES pH 7.9,300 mM sucrose, 50 mM NaCl, 1 mM EDTA pH 8.0, 1.5 mM

MgCl2, 0.6% NP40, 0.5 mM DTT, 0.5 mM PMSF, 1 mM

sodium metabisulfite, 1 mM benzamidine–HCl) and immedi-ately spun at 1700 g for 5 min. The supernatant wasremoved and saved as first wash fraction. The cells were resus-pended in the same buffer and incubated on ice for 30 min,followed by disruption of the cells in a homogenizer with aB pestle (at least 20 strokes). Nuclei were pelleted in arefrigerated microcentrifuge at 5700 g for 12 min and thesupernatant saved as cytoplasmic fraction. Nuclei were resus-pended in buffer B (50 mM Tris–HCl pH 7.5, 20% glycerol,10% sucrose, 0.42 M KCl, 5 mM MgCl2, 0.1 mM EDTA,2 mM DTT, 1 mM PMSF, 1 mM sodium metabisulfite, 1 mM

benzamidine–HCl) and placed on a rotating wheel at 48 for30 min. The nuclear extract was spun at 18400 g for 30 minat 48. The supernatant was removed as nuclear fraction.Protein concentration was determined with a Bradford assay(BioRad). 100 mg of the cytoplasmic and nuclear fractionswere separated on 10% SDS–PAGE and transferred to polyvi-nylidenfluorid membrane (BioRad). b-Catenin (1:500, E5,Santa Cruz Biotechnology) and a-tubulin (1:1000, T9026,Sigma) were detected using an ECL kit (GE Healthcare).For detection of the mutant WT1 protein, blocking was donein Super Block Blotting Buffer in PBS (Thermo Scientific)and the first antibody was an N-terminal specific antibody(1:220; clone 6F-H2, DAKO) and the second antibody was astabilized peroxidase conjugated goat anti-mouse antibody(H þ L, Thermo Scientific). Antibody-bound proteins weredetected with the Super Signal West Pico ChemiluminescentSubstrate (Thermo Scientific).

Differentiation analysis

Adipogenic, chondrogenic and osteogenic differentiation wasdone using reagents from Lonza and as described by the man-ufacturer. Non-induced control cells were kept in the respect-ive maintenance media. After the complete cycles of inductionand maintenance, cells were processed for adipogenic differ-entiation by staining with Oil Red O and cells from a parallelflask were used for RNA isolation (RNeasy, Qiagen). Forosteogenic differentiation, cells were harvested by scrapingthem in the presence of 0.5 M HCL and a calcium Liquicolorassay was performed according to the manufacturers instruc-tions (Stanbio Laboratory, USA). For chondrogenic differen-tiation, the pellet cultures were embedded in freezingmedium, cryo-sections were prepared and stained for glycosa-minoglycans with Safranin O. For the analysis of myogenicdifferentiation, HSMM cells (Lonza) were used as controls.Differentiation was induced with MSCGM (Lonza) sup-plemented with 2% horse serum for 1 week. Multinucleatedcells were documented with phase contrast microscopy. Forimmunofluorescence analysis, cells were seeded in fourchamber slides (BD Biosciences) and grown in reducedserum. After 1 week, cells were fixed with 2% paraformalde-hyde and 4% sucrose in PBS for 15 min at RT. Blocking wascarried out with PBS containing 4% FCS and 0.1% Tween 20for 30 min, followed by incubation with a titin-specific anti-body at a 1:100-fold dilution (H300, sc-28536; Santa CruzBiotechnology) in blocking solution for 1 h. Cells were

Human Molecular Genetics, 2010, Vol. 19, No. 9 1665

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 16: Wilms tumor cells with WT1 mutations have characteristic features of

washed with PBS and incubated with goat-anti-rabbit Alexafluor 488 antibody (Molecular Probes) for 1 h in the dark.Finally, cells were mounted with ProLong Antifade (Molecu-lar Probes) supplemented with DAPI and analyzed by fluor-escence microscopy.

For the semi-quantitative RT-PCR analysis of the FABP4gene for adipogeneic differentiation, the primers (for: 50-TATGAAAGAAGTAGGAGTGGGC-30; rev: 50-CCACCACCAGTTTATCATCCTC-30) were used for amplification and ali-quots of the reaction were removed after 25, 30 and 35 cycles.

Luciferase reporter assays in WT cells

Wilms cells were plated in six-well plates at a concentration of1.5 � 105 cells/well in MSCGM without antibiotics. At a con-fluence of 50–80%, the cells were cotransfected with TOP- orFOPflash-plasmids (Upstate Biotechnology) and eitherpRL-TK (Wilms1, 2, 4 and 5) or pRL-CMV (Wilms3)(Renilla luciferase) plasmid (Promega) for internal control oftransfection efficiency, using Lipofectamine LTX Reagentaccording to the protocol of the supplier (Invitrogen). Allassays were done at least in duplicates and the Firefly andRenilla luciferase activities were measured using the Dual-Luciferase Reporter Assay System (Promega) 24 h followingtransfection. The Renilla luciferase activity was used to nor-malize the transfection efficiency and the relative luciferaseactivity was used to calculate the TOP to FOP ratio.

Immunfluorescence and FACS

For staining of b-catenin, Wilms cell lines and MSC wereseeded in four chamber slides (BD Biosciences) at 0.5–2 �105 cells/well. At a confluence of 80%, cells were washedwith PBS and fixed with 3% paraformaldehyde for 15 min atroom temperature, followed by a PBS wash. Cells wereblocked and permeabilized in Blocking Solution consistingof 5% normal rabbit serum in PBS and 0.3% Triton X-100(Sigma) for 60 min at room temperature. Cells were incubatedovernight at 48C with the b-catenin antibody (1:150, # 9587,Cell Signaling). After washing with PBS, the slides were incu-bated 120 min in the dark with fluorochrome-conjugated anti-rabbit secondary antibody (1:500, Alexa Fluor 546; Invitro-gen). After mounting with Antifade (Invitrogen), they wereviewed and documented with a Zeiss Axioplan microscope.For the analysis of PDGFRA expression, the cells wereblocked with 50% goat serum, followed by incubation withthe antibody for 1 h at 48. The cells were washed with PBSand fixed with 3% formaldehyde, followed by incubationwith an FITC labeled anti-mouse antibody.

Flow cytometric analysis was performed on a FACS CantoFlow cytometer (BD Biosciences) and the results were ana-lyzed using DIVA software. Fluorescein isothiocyanate-conjugated monoclonal antibody to CD90 (clone F15-42-1-5;IgG1) and isotype controls were obtained from Beckman-Coulter (Krefeld, Germany); phycoerythrin-conjugatedmonoclonal antibody to CD73 (clone AD2; IgG1) from BDBiosciences (Heidelberg, Germany); phycoerythrin-conjugated monoclonal antibody to CD105 (clone 166707;IgG1) from R&D Systems (Wiesbaden, Germany).

RNA isolation, gene expression analysis and Q-RT-PCR

Total RNA was isolated from the cell lines using the RNeasyMini Kit (Qiagen). NK RNA was isolated from the cell cultureestablished from normal adjacent kidney from Wilms3 andgrown in WT medium. Labeling of the RNA in the One-Colorformat was performed as described by the manufacturer(Agilent Technologies). The cRNA was hybridized to 4 �44 K ‘Whole Human Genome Oligo Microarrays’ (Agilent)and ‘Spike-In positive controls’ (Agilent) were added asinternal controls. Microarrays were scanned with an AgilentTechnologies Scanner G2505B and scans were quantifiedusing Agilent Feature Extraction V10.1.1.1 Software. Theraw data were deposited in GEO, accession Nr. GSE18058.

The gene expression analysis of each cell line was repeatedtwice with the same RNA (technical reproduction) and twiceusing RNA from another passage (biological reproduction).In addition, after establishing two new cell lines, the geneexpression analysis was repeated with new RNA isolatedfrom all five cell lines in parallel. For the Q-RT-PCR,cDNA was synthesized using TaqMan Reverse TranscriptionReagents (Applied Biosystems) using total RNA. TheQ-RT-PCR was performed in triplicates using TaqMan geneexpression assay probes from Applied Biosystems for WT1(Hs00240913_m1), PAX3 (Hs00240950_m1), IGF2 (Hs01005963_m1), AXIN2 (Hs01063168_m1), FOXD1 (Hs00270117_s1), PDGFRA (Hs00183486_m1), SIM2 (Hs00231927_m1), DKK2 (Hs00205294_m1), MET (Hs01565580_m1),NT5E (CD73) (Hs00159686_m1), THY1 (CD90) (Hs00264235_s1), ENG (CD105) (Hs03986114_s1) and TaqMan Univer-sal PCR Master Mix (no AmpErase UNG). The analysis wasperformed on an ABI PRISM 7900HT Sequence DetectionSystem. The expression level of these genes in each samplewas normalized to an endogenous control gene, GAPDH(4333764T).

Statistical analysis

Four software packages were used for statistical analysis:LIMMA (R-library, Bioconductor, http://www.R-project.org)(57) for normalization and search for differentially expressedgenes, pvclust (R-library, http://www.is.titech.ac.jp/~shimo/prog/pvclust/) for cluster analysis, and globaltest (R-library, Bio-conductor) (58,59) and MEV (Multi-Experiment Viewer, TIGR)(60) for meta-analysis of KEGG pathways and GO terms, respect-ively. Before performing statistical tests, all array data were nor-malized using quantile normalization and filtered for genesconsidered to be unexpressed everywhere by removing thosegenes not exceeding a mean intensity of 200 in any cell line.

To determine relationships between cell lines, hierarchicalcluster analysis was applied to the filtered expression datausing pvclust. In addition to arranging the cell lines in acluster tree, pvclust allows to assess the significance of theobserved branches by producing bootstrap probabilities aswell as approximately unbiased P-values. Differential geneexpression between CLS1, MSC and WT was tested pairwise for each WT cell line separately, and only genes reportedby LIMMA as being differentially expressed in all tests for thecontrasts of interest were considered for further analysis bydividing them into sets of up- and down-regulated genes.

1666 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 17: Wilms tumor cells with WT1 mutations have characteristic features of

Biological meaning of these gene sets of differentiallyexpressed genes was investigated using MEV’s EASEmodule (Expression Analysis Systematic Explorer), testingfor the presence of over-represented GO terms. To test forassociation of pathways with the cell lines, a globaltest withKEGG pathways was performed on the filtered arrays, com-paring WT with MSC and the pooled WT and MSC datawith CLS1. For all statistical tests, a standard P-value of0.05 was used as significance level. Bootstrap P-values(pvclust) and adjusted resampling P-values (EASE) arebased on sample sizes of 10 000 and 1000, respectively.

SUPPLEMENTARY MATERIAL

Supplementary Material is available at HMG online.

ACKNOWLEDGEMENTS

We thank Dr B. Hildebrandt from the Institute of HumanGenetics, Dusseldorf for the cytogenetic analysis of theWilms cell lines and Dr R. Sorg, Institute for TransplantationDiagnostics and Cell Therapeutics, Dusseldorf for performingthe FACs analysis. We also thank Prof. Jurgens, Children Hos-pital, Pediatric Haematology and Oncology, University ofMunster, Germany and Dr Linderkamp, Childrens Hospital,Pediatric Haematology and Oncology, University of Hann-over, Germany, for tumor material.

Conflict of Interest statement: None declared.

FUNDING

This work was supported by grants from the DeutscheForschungsgemeinschaft [RO 501/12-1, RO 501/12-2].

REFERENCES

1. Beckwith, J.B., Kiviat, N.B. and Bonadio, J.F. (1990) Nephrogenic rests,nephroblastomatosis and the pathogenesis of Wilms tumor. Pediatr.Pathol., 10, 1–36.

2. Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K., Douglas, R.,Mosca, J.D., Moorman, M.A., Simonetti, D.W., Craig, S. and Marshak,D.R. (1999) Multilineage potential of adult human mesenchymal stemcells. Science, 284, 143–147.

3. Schumacher, V., Schneider, S., Figge, A., Wildhardt, G., Harms, D.,Schmidt, D., Weirich, A., Ludwig, R. and Royer-Pokora, B. (1997)Correlation of germ-line mutations and two-hit inactivation of the WT1gene with Wilms tumors of stromal-predominant histology. Proc. NatlAcad. Sci. USA, 94, 3972–3977.

4. Miyagawa, K., Kent, J., Moore, A., Charlieu, J.P., Little, M.H.,Williamson, K.A., Kelse, A., Brown, K.W., Hassam, S., Briner, J. et al.(1998) Loss of WT1 function leads to ectopic myogenesis in Wilms’tumour. Nat. Genet., 18, 5–17.

5. Nakadate, H., Yokomori, K., Watanabe, N., Tsuchiya, T., Namiki, T.,Kobayashi, H., Suita, S., Tsunematsu, Y., Horikoshi, Y., Hatae, Y. et al.(2001) Mutations/deletions of the WT1 gene, loss of heterozygosity onchromosome arms 11p and 11q, chromosome ploidy and histology inWilms’ tumors in Japan. Int. J. Cancer, 94, 396–400.

6. Shibata, R., Takata, A., Hashiguchi, A., Umezawa, A., Yamada, T. andHata, J. (2003) Responsiveness of chemotherapy based on the histologicaltype and Wilms’ tumor suppressor gene mutation in bilateral Wilms’tumor. Pathol. Int., 53, 214–220.

7. Schumacher, V., Schuhen, S., Sonner, S., Weirich, A., Leuschner, I.,Harms, D., Licht, J., Roberts, S. and Royer-Pokora, B. (2003) Two

molecular subgroups of Wilms’ tumors with or without WT1 mutations.Clin. Cancer Res., 9, 2005–2014.

8. Fukuzawa, R., Heathcott, R.W., Sano, M., Morison, I.M., Yun, K. andReeve, A.E. (2004) Myogenesis in Wilms’ tumors is associated withmutations of the WT1 gene and activation of Bcl-2 and the Wnt signallingpathway. Ped. Dev. Pathol., 7, 125–137.

9. Fukuzawa, R., Heathcott, R.W., More, H.E. and Reeve, A.E. (2007)Sequential WT1 and CTNNB1 mutations and alterations of b-cateninlocalisation in intralabor nephrogenic rests and associated Wilms tumours:two case studies. J. Clin. Pathol., 60, 1013–1016.

10. Pritchard-Jones, K., Fleming, S., Davidson, D., Bickmore, W., Porteous,D., Gosden, C., Bard, J., Buckler, A., Pelletier, J., Housman, D. et al.

(1990) The candidate Wilms’ tumour gene is involved in genitourinarydevelopment. Nature, 346, 194–197.

11. Koesters, R., Ridder, R., Kopp-Schneider, A., Betts, D., Adams, V.,Niggli, F., Briner, J. and von Knebel Doeberitz, M. (1999) Mutationalactivation of the b-catenin Proto-oncogene is a common event in thedevelopment of Wilms’ tumors. Cancer Res., 59, 3880–3882.

12. Maiti, S., Alam, R., Amos, C.I. and Huff, V. (2000) Frequent associationof b-catenin and WT1 mutations in Wilms tumors. Cancer Res., 60,6288–6292.

13. Li, C.-M., Kim, C.E., Margolin, A.A., Guo, M., Zhu, J., Mason, J.M.,Hensle, T.W., Murty, V.V.V.S., Grundy, P.E., Fearon, E.R. et al. (2004)CTNNB1 mutations and overexpression of Wnt//b-catenin target genes inWT1-mutant Wilms’ tumors. Am. J. Pathol., 165, 1943–1953.

14. Royer-Pokora, B., Weirich, A., Schumacher, V., Uschkereit, C., Beier, M.,Leuschner, I., Graf, N., Autschbach, F., Schneider, D. and von Harrach,M. (2008) Clinical relevance of mutations in the Wilms tumor suppressor1 gene WT1 and the cadherin-associated protein beta1 gene CTNNB1 forpatients with Wilms tumors: results of long-term surveillance of 71patients from International Society of Pediatric Oncology Study 9/Societyfor Pediatric Oncology. Cancer, 113, 1080–1089.

15. Ravenel, J.D., Broman, K.W., Perlman, E.J., Niemitz, E.L., Jayawardena,T.M., Bell, D.W., Haber, D.A., Uejima, H. and Feinberg, A.P. (2001) Lossof imprinting of insulin-like groth factor-II (IGF2) gene in distinguishingspecific biologic subtypes of Wilms tumor. J. Natl. Cancer Inst., 93,1698–1703.

16. Oliver, J., Barasch, J., Yang, J., Herzlinger, D. and Al-Awqat, Q. (2002)Mesenchyme contains embryonic renal stem cells. Am. J. Physiol. RenalPhysiol., 283, F799–F809.

17. Herzlinger, D., Koseki, C., Mikawa, T. and Al-Awqati, Q. (1992)Metanephric mesenchyme contains multipotent stem cells whose fate isrestricted after induction. Development, 114, 565–572.

18. Qiao, J., Cohen, D. and Herzlinger, D. (1995) The metanephric blastemadifferentiates into collecting system and nephron epithelia in vitro.Development, 121, 3207–3214.

19. Dekel, B., Zangi, L., Shezen, E., Reich-Zeliger, S., Eventov-Friedman, S.,Katchman, H., Jacob-Hirsch, J., Amariglio, N., Rechavi, G., Margalit, R.et al. (2006) Isolation and characterization of nontubular Sca-1þLin2

multipotent stem/progenitor cells from adult mouse kidney. J. Am. Soc.

Nephrol., 17, 3300–3314.20. Challen, G.A., Bertoncello, I., Deane, J.A., Ricardo, S.D. and Little, M.H.

(2006) Kidney side population reveals multilineage potential and renalfunctional capacity but also cellular heterogeneity. J. Am. Soc. Nephrol.,17, 1896–1912.

21. Bussolati, B., Bruno, S., Grange, C., Buttiglieri, S., Deregibus, M.C.,Cantino, D. and Camussi, G. (2005) Isolation of renal progenitor cellsfrom adult human kidney. Am. J. Pathol., 166, 545–555.

22. Hatini, V., Huh, S.O., Herzlinger, D., Soares, V.C. and Lai, E. (1996)Essential role of stromal mesenchyme in kidney morphogenesis revealedby targeted disruption of winged helix transcription factor BF-2. GenesDev., 10, 1467–1478.

23. Levinson, R.S., Batourina, E., Choi, C., Vorontchikhina, M., Kitajewski,J. and Mendelsohn, C.L. (2005) Foxd1-dependent signals controlcellularity in the renal capsule, a structure required for normal renaldevelopment. Development, 132, 529–539.

24. Guillaume, R., Bressan, M. and Herzlinger, D. (2009) Paraxial mesodermcontributes stromal cells to the developing kidney. Dev. Biol., 329,169–175.

25. Engleka, K.A., Gitler, A.D., Zhang, M., Zhou, D.D., High, F.A. andEpstein, J.A. (2005) Insertion of Cre into the Pax3 locus creates a newallele of Splotch and identifies unexpected Pax3 derivates. Dev. Biol., 280,396–406.

Human Molecular Genetics, 2010, Vol. 19, No. 9 1667

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018

Page 18: Wilms tumor cells with WT1 mutations have characteristic features of

26. Garvin, A.J., Surrette, F., Hintz, D.S., Rudisill, M.T., Sens, M.A. andSens, D.A. (1985) The in vitro growth and characterization of the skeletalmuscle component of Wilms’ tumor. Am. J. Pathol., 121, 298–310.

27. Garvin, A.J., Sullivan, J.L., Bennett, D.D., Stanley, W.S., Inabnett, T. andSens, D.A. (1987) The in vitro growth, heterotransplantation, andimmunohistochemical characterization of the blastemal component ofWilms’ tumor. Am. J. Pathol., 129, 353–363.

28. Hazen-Martin, D.J., Garvin, J., Gansler, T., Tarnowski, B.I. and Sens,D.A. (1993) Morphology and growth characteristics of epithelial cellsfrom classic Wilms’ tumors. Am. J. Pathol., 142, 893–905.

29. Hazen-Martin, D.J., Re, G.G., Garvin, A.J. and Sens, D.A. (1994)Distinctive properties of an anaplastic Wilms’ tumor and its associatedepithelial cell line. Am. J. Pathol., 144, 1023–1034.

30. Lobbert, R.W., Klemm, G., Gruttner, H.-P., Harms, D., Winterpracht, A.and Zabel, B.U. (1998) Novel WT1 mutation, 11p LOH, andt(7;12)(p22;q22) chromosomal translocation identified in a Wilms’ tumorcase. Genes Chromosomes Cancer, 21, 347–350.

31. Alami, J., Williams, B.R. and Yeger, H. (2003) Derivation andcharacterization of a Wilms’ tumor cell line, WiT 49. Int. J. Cancer, 107,365–374.

32. Haber, D.A., Park, S., Maheswaran, S., Englert, C., Re, G.G.,Hazen-Martin, D.J., Sens, D.A. and Garvin, A.J. (1993) WT1-mutatedgrowth suppression of Wilms tumor cells expressing a WT1 splicingvariant. Science, 262, 2057–2059.

33. Hard, G.C. (1984) Tumor biology. In vitro culture and transplantationmodels of Wilms’ tumor. Pochedly, C. and Baum, E.S. (eds), Wilms’Tumor Clinical and Biological Manifestations, Elsevier, New York,Amsterdam, Oxford, pp. 191–214.

34. Dekel, B., Metsuyanim, S., Schmidt-Ott, K.M., Fridman, E.,Jacob-Hirsch, J., Simon, A., Pinthus, J., Mor, Y., Barasch, J., Amariglio,N. et al. (2006) Multiple imprinted and stemness genes provide a linkbetween normal and tumor progenitor cells of the developing humankidney. Cancer Res., 66, 6040–6049.

35. Nagashima, Y., Nishihira, H., Miyagi, Y., Tanaka, Y., Sasaki, Y., Nishi,T., Imaizumi, K., Aoki, I. and Misugi, K. (1996) A nude mouse Wilms’tumor line (KCMC-WT-1) derived from an aniridia patient withmonoallelic partial deletion of chromosome 11p. Cancer, 77, 799–804.

36. Uschkereit, C., Perez, N., de Torres, C., Kuff, M., Mora, J. andRoyer-Pokora, B. (2007) Different CTNNB1 mutations as moleculargenetic proof for the independent origin of four Wilms tumors in a patientwith a novel germ-line WT1 mutation. J. Med. Genet., 44, 393–396.

37. Scott, R.H., Stiller, C.A., Walker, L. and Rahman, N. (2006) Syndromesand constitutional chromosomal abnormalities associated with Wilmstumour. J. Med. Genet., 43, 705–714.

38. Barberi, T., Willis, L.M., Socci, N.D. and Studer, L. (2005) Derivation ofmultipotent mesenchymal precursors from human embryonic stem cells.PLoS Med., 2, 554–560.

39. Shiraki, N., Higuchi, Y., Harada, S., Umeda, K., Isagawa, T., Aburatani,H., Kume, K. and Kume, S. (2009) Differentiation and characterization ofembryonic stem cells into three germ layers. Biochem. Biophys. Res.Com., 381, 694–699.

40. Brunskill, E.W., Aronow, B.J., Georgas, K., Rumballe, B., Valerius, M.T.,Aronow, J., Kaimal, V., Jegga, A.G., Grimmond, S., McMahon, A.P. et al.(2008) Atlas of gene expression in the developing kidney atmicroanatomic resolution. Dev. Cell, 15, 781–791.

41. Wellik, D., Hawkes, J. and Capecci, M.R. (2002) Hox11 paralogous genesare essential for metanephric kidney induction. Genes Dev., 16, 1423–1432.

42. Epping, M.T. and Bernards, R. (2006) A causal role for the human tumorantigen preferentially expressed antigen of melanoma in cancer. CancerRes., 66, 10639–10642.

43. Fukuzawa, R., Anaka, M.R., Weeks, R.J., Morison, I.M. and Reeve, A.E.(2009) Canonical WNT signalling determines lineage specificity in Wilmstumour. Oncogene, 28, 1063–1075.

44. Corbin, M., de Reynies, A., Rickman, D.S., Berrebi, D., Boccon-Gibod,L., Cohen-Gogo, S., Fabre, M., Jaubert, F., Faussillon, M., Yilmaz, F.et al. (2009) WNT/b-catenin pathway activation in Wilms tumors: aunifying mechanism with multiple entries? Genes Chromosomes Cancer,48, 816–827.

45. Zirn, B., Samans, B., Wittmann, S., Pietsch, T., Leuschner, I., Graf, N. andGessler, M. (2006) Target genes of the WNT/b-catenin pathway in Wilmstumors. Genes Chromosomes Cancer, 45, 565–574.

46. Kreidberg, J.A., Sariola, H., Loring, J.M., Maeda, M., Pelletier, J.,Housman, D. and Jaenisch, R. (1993) WT-1 is required for early kidneydevelopment. Cell, 74, 679–691.

47. Davis, J.A., Ladomery, M., Hohenstein, P., Michael, L., Shafe, A.,Spraggon, L. and Hastie, N. (2004) Development of an siRNA-basedmethod for repressing specific genes in renal organ culture and its use toshow that the Wt1 tumour suppressor is required for nephrondifferentiation. Hum. Mol. Genet., 13, 235–246.

48. Gang, E.J., Bosnakovski, D., Simsek, T., To, K. and Perlingeiro, R.C.R.(2008) Pax3 activation promotes the differentiation of mesenchymal stemcells toward the myogenic lineage. Exp. Cell Res., 314, 1721–1733.

49. Ridgeway, A.G. and Skerjanc, I.S. (2001) Pax3 is essential for skeletalmyogenesis and the expression of Six1 and Eya2. J. Biol. Chem., 276,19033–19039.

50. Hueber, P.A., Fukuzawa, R., El-Kares, R., Chu, L.L., Blumenkrantz, M.,He, S., Anaka, M.R., Reeve, A.E., Eccles, M.R., Jabado, N. et al. (2008)PAX3 is expressed in the stromal compartment of the developing kidneyand in Wilms tumors with myogenetic phenotype. Pediatr. Dev. Pathol.,(Epub ahead of print) Published Online: 1 August 2008, doi: 10.2350/08-05-0466.1.

51. Iglesias, D.M., Hueber, P.-A., Chu, L.L., Campbell, R., Patenaude, A.-M.,Dziarmaga, A.J., Quinlan, J., Mohamed, O., Dufort, D. and Goodyer, P.R.(2007) Canonical WNT signaling during kidney development.Am. J. Physiol. Renal Physiol., 293, F494–F500.

52. Schmidt-Ott, K.M. and Barasch, J. (2008) Wnt/b-catenin signallingin nephron progenitors and their epithelial progeny. Kidney Int., 74,1004–1008.

53. Park, J., Valerius, M. and McMahon, A. Wnt/b-catenin signalingregulates nephron induction during mouse kidney development.Development, 134, 2533–2539.

54. Roszko, I., Sawada, A. and Solnica-Krezel, L. (2009) Regulation ofconvergence and extension movements during vertebrate gastrulation bythe Wnt/PCP pathway. Semin. Cell Dev. Biol., doi:10.1016/j.semcdb.2009.09.004.

55. Sweetman, D., Wagstaff, L., Cooper, O., Weijer, C. and Munsterberg, A.(2008) The migration of paraxial and lateral plate mesoderm cellsemerging from the late plate primitive streak is controlled by differentWnt signals. BMC Dev. Biol., 8, 63.

56. Kim, M.K.-H., McGarry, T.J., Broin, P., Flatow, J.M., Golden, A.A.-J.and Licht, J.D. (2009) An integrated genome screen identifies the Wntsignalling pathway as a major target of WT1. Proc. Natl Acad. Sci. USA,106, 11154–11159.

57. Smyth, G.K. (2005) Limma: linear models for microarray data.Gentleman, R., Carey, V., Dudoit, S., Irizarry, R. and Huber, W. (eds),Bioinformatics and Computational Biology Solutions using R andBioconductor, Springer, New York, pp. 397–420.

58. Goeman, J.J., van de Geer, S.A., de Kort, F. and van Houwelingen, H.C.(2004) A global test for groups of genes: testing association with a clinicaloutcome. Bioinformatics, 20, 93–99.

59. Goeman, J.J., Oosting, J., Cleton-Jansen, A.-M., Anninga, J.K. and vanHouwelingen, H.C. (2005) Testing association of a pathway with survival.Bioinformatics, 21, 1950–1957.

60. Saeed, A.I., Sharov, V., White, J., Li, J., Liang, W., Bhagabati, N.,Braisted, J., Klapa, M., Currier, T., Thiagarajan, M. et al. (2003) TM4: afree, open-source system for microarray data management and analysis.Biotechniques, 34, 374–378.

1668 Human Molecular Genetics, 2010, Vol. 19, No. 9

Downloaded from https://academic.oup.com/hmg/article-abstract/19/9/1651/677433by gueston 18 March 2018