contribution of tumor heterogeneity in a new animal model of … · published onlinefirst february...

13
Genomics Contribution of Tumor Heterogeneity in a New Animal Model of CNS Tumors Fuyi Chen 1 , Albert J. Becker 2 , and Joseph J. LoTurco 1 Abstract The etiology of central nervous system (CNS) tumor heterogeneity is unclear. To clarify this issue, a novel animal model was developed of glioma and atypical teratoid/rhabdoid-like tumor (ATRT) produced in rats by nonviral cellular transgenesis initiated in utero. This model system affords the opportunity for directed oncogene expression, clonal labeling, and addition of tumor-modifying transgenes. By directing HRasV12 and AKT transgene expression in different cell populations with promoters that are active ubiquitously (CAG promoter), astrocyte-selective (glial brillary acidic protein promoter), or oligodendrocyte-selective (myelin basic protein promoter) we generated glioblastoma multiforme and anaplastic oligoastrocytoma, respectively. Importantly, the glioblastoma multiforme and anaplastic oligoastrocytoma tumors were distinguishable at both the cellular and molecular level. Furthermore, proneural basic helix-loop-helix (bHLH) transcription factors, Ngn2 (NEUROG2) or NeuroD1, were expressed along with HRasV12 and AKT in neocortical radial glia, leading to the formation of highly lethal ATRT like tumors. This study establishes a unique model in which determinants of CNS tumor diversity can be parsed out and reveals that both mutation and expression of neurogenic bHLH transcription factors contribute to CNS tumor diversity. Implications: A novel CNS tumor model reveals that oncogenic events occurring in disparate cell types and/or molecular contexts lead to different tumor types; these ndings shed light on the sources of brain tumor heterogeneity. Mol Cancer Res; 12(5); 74253. Ó2014 AACR. Introduction Tumors of the central nervous system (CNS) have signif- icant intra- and intertumor heterogeneity in terms of cellular composition, cellular proliferation, invasiveness, and epige- netic status (1, 2). Integrative large-scale gene expression analysis of 200 glioblastoma multiforme tumors revealed four subtypes: proneural, neural, classical, and mesenchymal (1). A more recent glioblastoma multiforme classication based on a combination of epigenetics, copy-number variation, gene expression, and genetic mutations has led to identi- cation of as many as six glioblastoma multiforme subgroups (2). In addition, there are at least two prognostic subgroups of pediatric glioblastoma multiforme based on association with or without the aberrantly active Ras/AKT pathway (3, 4). Heterogeneity in glioblastoma multiforme tumors may be responsible for differential response to therapeutic interven- tions (1), and understanding the etiology of tumor hetero- geneity in animal models may become increasingly important to design therapeutic strategies effective at targeting molec- ularly dened tumor subtypes. Several animal models of brain tumor, including xenograft models, genetically engineered mouse models (GEMM), and models using virus-mediated somatic cell transgenesis have been used to address important aspects of CNS tumor biology. For example, GEMM-harboring mutations found in human gliomas have been used to assess tumorigenic effects of individual genes and mutations and to reveal the tumor cell-of-origin in some models (5, 6). Endogenous cell populations have been induced to form gliomas by both viral and nonviral somatic transgenesis of oncogenes or deletion of tumor suppressor genes in specic target cell populations (717). For example, oligodendrocyte precursor cells (OPC) can be the cell-of-origin for adult glioma (5, 6, 13) with OPC- originated glioma matching the human proneural subgroup of glioblastoma multiforme (6, 18). Friedmann-Morvinski and colleagues found that RNA interference (RNAi) knock- down of NF1 and p53 expression in either astrocytes glial brillary acidic protein (GFAPþ) or neurons (SynIþ) induced formation of mesenchymal glioblastoma multiforme subtypes, whereas the same RNAi in nestin-positive neural progenitors induced neural glioblastoma multiforme sub- types (16). To more fully explore causes of tumor diversity in cerebral cortex, we have developed an animal model in which multiple transgenes can be expressed in selected cell populations at different times in forebrain development. Authors' Afliations: 1 Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut; and 2 Department of Neu- ropathology, University of Bonn Medical Center, Bonn, Germany Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). Corresponding Author: Joseph J. LoTurco, University of Connecticut, 75 North Eagleville Road, Unit 3156, Storrs, CT 06268. Phone: 860-486-3283; Fax: 860-486-3303; E-mail: [email protected] doi: 10.1158/1541-7786.MCR-13-0531 Ó2014 American Association for Cancer Research. Molecular Cancer Research Mol Cancer Res; 12(5) May 2014 742 on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Upload: others

Post on 13-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

Genomics

Contribution of Tumor Heterogeneity in a New Animal Modelof CNS Tumors

Fuyi Chen1, Albert J. Becker2, and Joseph J. LoTurco1

AbstractThe etiology of central nervous system (CNS) tumor heterogeneity is unclear. To clarify this issue, a novel animal

model was developed of glioma and atypical teratoid/rhabdoid-like tumor (ATRT) produced in rats by nonviralcellular transgenesis initiated in utero. This model system affords the opportunity for directed oncogene expression,clonal labeling, and addition of tumor-modifying transgenes. By directingHRasV12 and AKT transgene expressionin different cell populations with promoters that are active ubiquitously (CAG promoter), astrocyte-selective (glialfibrillary acidic protein promoter), or oligodendrocyte-selective (myelin basic protein promoter) we generatedglioblastoma multiforme and anaplastic oligoastrocytoma, respectively. Importantly, the glioblastoma multiformeand anaplastic oligoastrocytoma tumors were distinguishable at both the cellular andmolecular level. Furthermore,proneural basic helix-loop-helix (bHLH) transcription factors, Ngn2 (NEUROG2) or NeuroD1, were expressedalongwithHRasV12 andAKT in neocortical radial glia, leading to the formation of highly lethal ATRT like tumors.This study establishes a unique model in which determinants of CNS tumor diversity can be parsed out and revealsthat both mutation and expression of neurogenic bHLH transcription factors contribute to CNS tumor diversity.

Implications: A novel CNS tumor model reveals that oncogenic events occurring in disparate cell types and/ormolecular contexts lead to different tumor types; these findings shed light on the sources of brain tumorheterogeneity. Mol Cancer Res; 12(5); 742–53. �2014 AACR.

IntroductionTumors of the central nervous system (CNS) have signif-

icant intra- and intertumor heterogeneity in terms of cellularcomposition, cellular proliferation, invasiveness, and epige-netic status (1, 2). Integrative large-scale gene expressionanalysis of 200 glioblastomamultiforme tumors revealed foursubtypes: proneural, neural, classical, and mesenchymal (1).A more recent glioblastoma multiforme classification basedon a combination of epigenetics, copy-number variation,gene expression, and genetic mutations has led to identifi-cation of as many as six glioblastoma multiforme subgroups(2). In addition, there are at least two prognostic subgroups ofpediatric glioblastoma multiforme based on association withor without the aberrantly active Ras/AKT pathway (3, 4).Heterogeneity in glioblastoma multiforme tumors may beresponsible for differential response to therapeutic interven-tions (1), and understanding the etiology of tumor hetero-

geneity in animalmodelsmay become increasingly importantto design therapeutic strategies effective at targeting molec-ularly defined tumor subtypes.Several animal models of brain tumor, including xenograft

models, genetically engineeredmousemodels (GEMM), andmodels using virus-mediated somatic cell transgenesis havebeen used to address important aspects of CNS tumorbiology. For example, GEMM-harboring mutations foundin human gliomas have been used to assess tumorigeniceffects of individual genes and mutations and to reveal thetumor cell-of-origin in some models (5, 6). Endogenous cellpopulations have been induced to form gliomas by both viraland nonviral somatic transgenesis of oncogenes or deletionof tumor suppressor genes in specific target cell populations(7–17). For example, oligodendrocyte precursor cells (OPC)can be the cell-of-origin for adult glioma (5, 6, 13)withOPC-originated glioma matching the human proneural subgroupof glioblastoma multiforme (6, 18). Friedmann-Morvinskiand colleagues found that RNA interference (RNAi) knock-down of NF1 and p53 expression in either astrocytes glialfibrillary acidic protein (GFAPþ) or neurons (SynIþ)induced formation ofmesenchymal glioblastomamultiformesubtypes, whereas the same RNAi in nestin-positive neuralprogenitors induced neural glioblastoma multiforme sub-types (16). To more fully explore causes of tumor diversityin cerebral cortex, we have developed an animal model inwhich multiple transgenes can be expressed in selected cellpopulations at different times in forebrain development.

Authors' Affiliations: 1Department of Physiology and Neurobiology,University of Connecticut, Storrs, Connecticut; and 2Department of Neu-ropathology, University of Bonn Medical Center, Bonn, Germany

Note: Supplementary data for this article are available at Molecular CancerResearch Online (http://mcr.aacrjournals.org/).

Corresponding Author: Joseph J. LoTurco, University of Connecticut, 75North Eagleville Road, Unit 3156, Storrs, CT 06268. Phone: 860-486-3283;Fax: 860-486-3303; E-mail: [email protected]

doi: 10.1158/1541-7786.MCR-13-0531

�2014 American Association for Cancer Research.

MolecularCancer

Research

Mol Cancer Res; 12(5) May 2014742

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 2: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

DNA transposon systems have been previously adapted tononviral somatic transgenesis in studies of neocortical devel-opment (19, 20), cellular reprogramming (21), and togenerate animal models of glioma (17). In utero electropo-ration is a relatively efficient method to deliver multiplecombinations of transgenes into neuronal and glial progeni-tors in the developing forebrain in utero (22). When com-bined with a DNA transposon system including cell type–specific promoters, it becomes possible to introducemultipletransgenes, including oncogenes, in different populations ofcells (19, 20, 23, 24). The active Ras pathway (25) and thephosphoinositide 3-kinase/AKT pathway (9, 26) have beenfrequently found in patients with human glioma and used invarious glioma animal models (9, 10, 12, 15). In the modeldescribed here, we introduced HRasV12 and AKT in cellpopulations in the radial glia lineage to ask whether diversityin glioma is linked to the population of cells induced toexpress oncogenic transgenes. We also tested whether addi-tion of neurogenic transcription factors to the startingprogenitor population modifies tumor type. We show byseveral measures, including morbidity, histology, develop-mental time course, tumor clonal pattern, and molecularsignature, that diverse tumors are generated when oncogeneexpression is directed into different cell populations. More-over, expression of the basic helix-loop-helix (bHLH) tran-scription factorsNgn2 orNeuroD1 along withHRasV12 andAKT results in atypical teratoid/rhabdoid-like tumor(ATRT), a pediatric tumor type rarely produced in existingtumor models.

Materials and MethodsPlasmidsA system of piggyBac transposon donor and helper plasmids

were produced and used in this study. To make the donorplasmids PBCAG-HRasV12 and PBCAG-AKT, HRasV12was PCR amplified from pTomo (15; Addgene plasmid26292), human AKT was amplified from 1036 pcDNA3Myr HA Akt1, (26; Addgene plasmid 9008), respectively,and inserted into the EcoRI/NotI sites of pPBCAG-enhancedgreen fluorescent protein (eGFP) (19). For construction ofPBGFAP-HRasV12/AKT, PBMBP-HRasV12/AKT, HRas-V12, and AKT were inserted into EcoRI/NotI sites ofPBGFAP-GFP and PBMBP-GFP, respectively (19). Forthe bHLH donor plasmids PBCAG-Ngn2 and PBCAG-NeuroD1, human Neurogenin2 cDNA clone (IMAGEID 5247719), human Neuronal Differentiation 1 cDNA(IMAGE ID 3873419) were purchased from Open Biosys-tem, PCR amplified and inserted into PBCAG-eGFP EcoRI/NotI sites. CAG-Ngn2 was made by inserting Ngn2 codingsequence into EcoRI/NotI sites of pCAG-eGFP.

AnimalsPregnant Wistar rats were obtained from Charles River

Laboratories, Inc. and maintained at the University of Con-necticut (Storrs, CT) vivarium on a 12-hour light cycle andfed ad libitum. Animal gestational ages were determined andconfirmed during surgery. Both male and female subjects

were used for tumor induction. For the purpose of construct-ing survival curves (Figs. 2A and 3A), rats were removedfrom the experiment for humane purposes when theybecame unable to eat or drink. At the end of the 40-weeksurvival experiment, percentage of survival for each precedingweek was calculated and percentage of survival curveswas generated. For tumors induced by PBCAG-Ngn2/PBCAG-HRasV12/AKT and PBCAG-NeuroD1/PBCAG-HRasV12/AKT, survival rates were also calculated for eachday. To statistically compare survival rates, a log-rank test,Breslow test, and Tarone–Ware test were performed usingSPSS (IBM SPSS statistics 21). All procedures and experi-mental approaches were approved by the University of Con-necticut Institutional Animal Care and Use Committee.

In utero electroporationIn utero electroporation was performed as previously

described (19, 20, 27). Electroporation was performed atembryonic day 14 or 15 (E14 or E15) and gestation age wasconfirmed during surgery. All plasmids were used at the finalconcentration of 1.0 mg/mL except PBCAG-cyan fluorescentprotein (CFP) and GLAST-PBase were used at the finalconcentration of 2.0 mg/mL.

Image acquisition and 3D reconstructionMulticolor imaging was performed as described using a

Zeiss Axio imager M2 microscope with Apotome with 488/546/350-nm filter cubes and the X-Cite series 120Q lightsource (20). All the images were further processed in AdobePhotoshop CS3 software. For three-dimensional (3D)reconstruction, P27 Wistar rats were deeply anesthetizedwith isoflurane and perfused transcardially with 4% para-formaldehyde/PBS (4% PFA). Samples were post fixedovernight in 4% PFA and sectioned at 65-mm thickness onvibratome (Leica VT 1000S). Sections were mounted ontomicroscope slides in sequential order, all aligned in the samedorsal–ventral and left–right arrangement. Images wereacquired with Stereo Investigator (Microbright Field). Afterimaging acquisition, outlines of the cerebral hemisphere andtumor clones were traced using color-coded contours for thehemisphere and each clone. Then traced images were trans-ferred to Neurolucida Explorer (Microbright Field), inwhich several contours from sequential sections wereappended to one another, and organized and stacked inorder. 3D model was created using Neurolucida Explorersoftware 3D visualization option to visualize the clonality ofselected clones of tumor cells.

ImmunohistochemistryAnimals were deeply anesthetized with isoflurane and

perfused transcardially with 4% paraformaldehyde/PBS(4% PFA). Samples were post fixed overnight in 4% PFA.For immunofluorescence, brains were sectioned at 65-mmthickness on a vibratome (Leica VT 1000S). Sections wereprocessed as free-floating sections and stained with GFAP(Cell Signaling Technology; 3670X), CC1 (Calbiochem;OP80), and NG2 (Chemicon; AB5320) antibodies. Imageswere acquired and processed as previous described (19, 20).

Determinants of Tumor Heterogeneity

www.aacrjournals.org Mol Cancer Res; 12(5) May 2014 743

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 3: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

For histologic analysis, immunohistochemistry and hema-toxylin and eosin staining were carried out on paraffin-embedded 4-mm sections as described in (28, 29) usingGFAP (DAKO;Z0334),MAP2c (Sigma;M4403), vimentin(DAKO; M0725), Synaptophysin (DAKO; M0776), Cyto-keratin (BMA Medicals; T-1302), Actin (DAKO; M0851),epithelial membrane antigen (DAKO; M0613) antibody.

Tumor cell cultureTumors were dissected from PBCAG-HRasV12/AKT–

transfected animals at the age P21. After dissection, tumortissues were chopped and trypsinized into single-cell sus-pension. Then cells were cultured in a serum-free mediumconsisting of Dulbecco's Modified Eagle Medium with L-glutamine, sodium pyruvate, B-27, N-2, basic fibroblastgrowth factor, and EGF (Invitrogen). Two days later, tumorcell adherent cultures were passaged into neurosphere sus-pension cultures at a density of 10 cells/mL and allowed togrow for 3 days to form tumor spheres.

RNA extraction, cDNA synthesis, and qRT-PCRAnimals ages P21 to P27 were deeply anesthetized with

isoflurane. Brains were quickly removed on ice. Tumorswere identified on the brain surface, removed, and thenchopped into cubes. Tissue from the opposite hemispherethat was tumor free was used as control. RNA extraction wasperformed using the Ambion RNAqueous Kit (Invitrogen)according to the manufacturer's instructions. cDNA syn-thesis was performed using the Transcriptor First StrandcDNA synthesis Kit (Roche) following manufacture's pro-tocol. Quantitative real-time PCR (qRT-PCR) was per-formed using Fast SYBR Green master mix (Applied Bio-system). Primer sequences are listed in SupplementaryTable S1. Triplicates were included from each sample. Foldof regulation of gene expression was calculated using theDDCt method. More specifically, the Ct values obtainedfrom tumor and control brain RNA samples are directlynormalized to a housekeeping gene (GAPDH). DDCt is thedifference in the DCt values between the tumor and controlsamples. The fold change in expression of the gene of interestbetween the tumor and control brain is then equal to 2^

(�DDCt). Unsupervised hierarchical clustering was per-formed on fold of regulation for each of the 24 genes profiledand a heat map was generated using the clustergram com-mand in the MATLAB bioinformatics toolbox (MathWorks). Other statistical analyses were performed usingKaleidaGraph version 4.0 (SynergySoftware 2006). A con-fidence interval of 95% (P < 0.05) was required for valuesto be considered statistically significant. All data are pre-sented as means and SEM.

ResultsA model of glioblastoma multiforme by piggyBac-mediated somatic transgenesisWedeveloped a nonviral somatic cell transgenesis approach

to produce tumors from endogenous rat neural progenitorsin vivo. HRasV12 and AKT transgenes were introduced intoembryonic neocortical neural progenitors, also known as

radial glial progenitors, by in utero electroporation of thelateral ventricles of embryonic rats. For nonviral cellulartransgenesis, we produced a system of plasmids consistingof a helper plasmid that expresses the piggyBac transposase inradial glial progenitors via the glutamate aspartate transporter(GLAST) promoter (GLAST-PBase) and thereby drivestransposon integration into the genomes of radial glia pro-genitors, and a set of donor plasmids containing transgenesflanked by transposon inverted terminal repeats (ITR) to beintegrated into the genome by piggyBac transposase activity.The donor plasmids included HRasV12 and AKT under thecontrol of one of three different promoters [CAG, myelinbasic protein (MBP), or GFAP; Fig. 1A] and a multicolorsystem of donor plasmids (20) with the CAG promoterdriving expression of eGFP, monomeric red fluorescentprotein (mRFP), or CFP to create multicolor clonal labeling(Fig. 1A; refs. 20, 30). Electroporation of this system ofplasmids unilaterally into the lateral cerebral ventricles ofE14–E15 rat embryos (Fig. 1A) invariably resulted by 21daysafter birth in the formation of large unilateral tumors (Fig.2F). Tumors were composed of both regions of uniformlycolored cells indicating significant regionalized clonal expan-sion (Fig. 1C and D), and regions containing a mixture ofdifferently colored cells indicating clonal mixing (Fig. 1F). Inaddition to tumors, streams of cells typically of one or twodifferent colors were found distal to the tumor core. Thesestreams were frequently in directionally oriented chainssuggesting migration and invasion into surrounding neuraltissue (Fig. 1E). Survival experiments showed that tumor-bearing animals began dying at 3 weeks of age and the rate ofdeath seemed stable by 31 weeks after birth for the CAGdonor plasmid–treated animals (Fig. 2E). Tumors wereconfirmed in all electroporated animals (30 of 30).Histologically, tumors induced by HRasV12 and AKT

driven by the CAG donor plasmids were composed offibrillary and gemistocytic elements (Supplementary Fig.S1A). The tumors diffusely infiltrated neighboring tissues(Supplementary Fig. S1B) and had cells with highly pleo-morphic cytologic appearance, and increasedmitotic activityindicative of malignancy (Supplementary Fig. S1C). Tumorcells frequently invaded the subarachnoidal space (Fig. 2G),and tumors had areas of necrosis indicative of high malig-nancy (Fig. 2H arrow). Tumors seemed by P7 and necroticareas were found as early as at P4. Immunohistochemicalanalysis revealed that tumor cells showed strong positivity forGFAP (Fig. 2I), vimentin (Fig. 2J), and mitogen-activatedprotein 2 (MAP2; Fig. 2K). Some neurons entrapped withintumors were positive for synaptophysin, whereas the bulkof the tumors did not show synaptophysin positivity.Interestingly, we did not observe vascular proliferation inthe rat tumors produced by the CAG donor plasmids, butobserved vascular proliferation in tumors induced by allother plasmid combinations used in this study. Moreover,when cultured in neurosphere suspension medium, tumorcells can form tumor spheres (Supplementary Fig. S2). Insum, the piggyBac transposon system effectively produces atumor model resembling human glioblastoma multiforme(WHO grade 4).

Chen et al.

Mol Cancer Res; 12(5) May 2014 Molecular Cancer Research744

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 4: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

MBPandGFAPpromoter–directed oncogene expressionproduces distinct tumor typesThe binary piggyBac system allows for directing inser-

tion of transgenes in one population of cells (i.e., radialglia neural progenitors), but then delayed expression ofoncogene expression in later-generated cell types in thelineage. This feature allowed us to introduce oncogenesinto neural progenitors at the embryonic lateral ventriclesurfaces, but then have their expression turned on later,primarily in either astrocytes, or oligodendrocytes. For thisexperiment, we constructed donor plasmids in whichHRasV12 and AKT expression was controlled by eithera mouse GFAP promoter fragment (19; PBGFAP-HRasV12/AKT) or a rat MBP promoter fragment (31;PBMBP-HRasV12/AKT). We first assessed the activity ofthe mouse GFAP and rat MBP promoters using GFP as areporter. We found that the GFAP promoter resulted in69.3% � 3% astrocytes, 19.4% � 2% neurons, 7.1% �2% oligodendrocytes, and 4.3% � 0.7% oligodendrocyteprecursor cells. In contrast, the MPB promoter construct(PBMBP-eGFP/mRFP) labeled 57.0% � 2% oligoden-drocytes, 16.2% � 0.7% astrocytes, 19.3% � 2% neu-

rons, and 7.5% � 0.7% oligodendrocyte precursor cells(Fig. 2C and D). One-way ANOVA showed a significantdifference between GFAP and MBP promoter–labeledastrocytes (P ¼ 6.91E-05) and oligodendrocytes (P ¼2.69E-05). A significant difference was also foundbetween NG2 cells labeled by GFAP and MBP promoters(one-way ANOVA, P ¼ 0.021172), but no significantdifference was found between the fraction of labeledneurons (one-way ANOVA, P ¼ 0.989884). The differ-ences in the cell populations labeled by the MBP andGFAP promoter fragments are consistent with enrichedtargeting of oligodendrocytes and astrocytes, respectively;however, each promoter was also capable of labeling somefraction of each cell type.We next compared and contrasted tumors produced by

the donor plasmids containing either the GFAP or MBPpromoters driving oncogene expression. For both donorplasmids, tumors were found in all animals and 65.2% (15of 23) of animals died by 40 weeks (Fig. 2E and F). A log-rank test showed no significant difference in survivalamong animals bearing tumors induced by GFAP, MBP,or CAG donor plasmids (log-rank test; P¼ 0.194 between

Figure 1. Multicolor rat glioma. A, schematic representation of piggyBac IUE approach and plasmids used. In utero electroporation was performedbetween E14 and E15 with plasmids listed. We used the multicolor piggyBac system to clonally label cells, oncogene donor plasmids to induce tumors,and both donor and conventional episomal plasmids encoding transcription factors to evaluate their role in gliomagenesis. After the animalswere born, tumor assessment was performed at different postnatal ages. B, representative images of a P22 rat brain hemisphere transfected withmulticolor piggyBac plasmids and PBCAG-HRasV12/ATK donor plasmids. Necrosis is labeled with N. C, overlay image showing large regions oftumor cells were uniformly labeled with mRFP. D, overlay image showing other regions of tumor were uniformly labeled with eGFP. E, streams of tumorcells invading tissue adjacent to tumors. F, region of the tumor containing a mixture of red- and green-labeled transformed cells indicatingregions of clonal mixing. G, 3D reconstruction of 5 tumor clones in a P27 rat transfected with PBGFAP-HRasV12/AKT. CT, cortex; CC, corpuscallosum. Scale bar, 500 mm in B and 100 mm in C, D, E, and F.

Determinants of Tumor Heterogeneity

www.aacrjournals.org Mol Cancer Res; 12(5) May 2014 745

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 5: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

Figure2. Distinct tumor types are inducedbydifferent donor plasmids. A,mouseGFAPpromoter fragment–labeled astrocytes.B, ratMBPpromoter fragment–labeled oligodendrocytes. C, quantification of proportions of labeled cells by GFAP and MBP promoter fragments (one-way ANOVA; �, P < 0.05;���, P < 0.01. NS, no significance difference). D, representative images of neuron, astrocyte, oligodendrocyte, and NG2 cell. E and F, Kaplan–Meier survivalcurvesand representative appearanceof tumor-bearingbrains. The log-rank test showednodifference in survival across all groups.G toK, characterization oftumors induced by PBCAG-HRasV12/AKT. G, subarachnoidal spread of tumor cells (H&E). H, necrosis (black arrow, H&E). I to K, tumor cells withfibrillary processes were positive for GFAP, vimentin, and MAP2. L to P, characterization of tumors induced by PBGFAP-HRasV12/AKT. L, overviewof H&E staining. Black arrow, vessels. M, necrosis (black arrow). N to P, GFAP, vimentin, and MAP2-positive processes. Q to U, characterizationof tumors induced by PBMBP-HRasV12/AKT. Q, prominent oligodendroglial components (H&E). R, round cell appearance of tumor cell (inset) andmitosis (black arrow, H&E). S, GFAP-positive astroglial process. T, oligodendroglial cells were vimentin-negative, whereas astroglial elements werevimentin-positive. U, MAP2-positive oligodendroglial component (black arrow, inset) and MAP2-positive astrocytes component (gray arrow). Scale bar,200 mm in A, B, L–N, P, Q, and S; 100 mm in G, I–K; 50 mm in H, O, R, and T; 20 mm in D; and 10 mm in inset in R and U.

Chen et al.

Mol Cancer Res; 12(5) May 2014 Molecular Cancer Research746

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 6: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

CAG and GFAP; P ¼ 0.554 between GFAP andMBP donor plasmid–transfected animals. For comparisonbetween CAG and MBP donor plasmid–transfected ani-mals, log-rank test, P ¼ 0.093; Breslow test, P ¼ 0.121;Tarone–Ware test, P ¼ 0.106). A histopathologic analysisof tumors in both GFAP and MBP promoter conditionsindicated frequent mitotic figures (Supplementary Fig.S1D, Fig. 2R, black arrow), similar to the CAG promotercondition described above, but also showed vascular pro-liferates consistent with highly malignant tumor types.Tumors were observed in 23 of 23 animals assessed fortumors 14 to 280 days after birth. In addition, GFAPdonor plasmid–induced tumors, unlike theMBP plasmids,resulted in tumors with prominent necrotic foci frequentlysurrounded by "pseudopalisading" tumor cells (Fig. 2M,arrow). Immunohistologic assessment of tumors inducedby GFAP donor plasmids indicated process-rich astroglialcells with delicate processes positive for GFAP, vimentin,and MAP2 (Fig. 2N–P). We conclude based on thehistopathologic findings that the GFAP donor plasmid–induced tumors are malignant astroglial tumors resemblinghuman glioblastoma multiforme (WHO grade 4).Although similar in many ways to tumors induced by theCAG donor plasmid, the multicolor clonal analysis indi-cated that the GFAP donor plasmids resulted in tumorswith larger clonal territories than the CAG donor plasmids.Similarly, the tumors in the GFAP donor plasmid condi-tion showed more clonally associated invasion and expan-sion into striatum than did tumors in the CAG donorplasmid condition.In contrast with the GFAP and CAG donor plasmid–

induced tumors, the MBP donor plasmid–induced tumorscontained a mixture of astroglial and oligodendroglial com-ponents, and lacked prominent necrotic foci and pseudo-palisading cell arrangements typical of the GFAP donorplasmid–induced tumors (Fig. 2M). In some areas of MBPdonor plasmid–induced tumors, there were processes withGFAP and vimentin positivity (Fig. 2S and T), but theseareas were less frequently observed than in the GFAP donorplasmid–induced tumors. In addition to the small astroglialcell fraction, the MBP donor plasmid–induced tumorscontained prominent "honeycomb"-like cell clusters (Fig.2Q) with round isomorphic nuclei located centrally toperinuclear halos (Fig. 2R, inset): a feature resemblingcytologic characteristics of human oligodendroglioma. Cellswere largely negative for vimentin (Fig. 2T), but positive forMAP2, an immunostaining pattern consistent with mixedoligodendroglial and astroglial components. MAP2 immu-nopositivity showed strong perinuclear cytoplasmic stainingwithout significant process labeling (Fig. 2U, black arrow,inset) typical of oligodendroglioma. Some MAP2-stainedcells in MBP donor plasmid–induced tumors displayed bi-or multipolar processes typical of astroglial cells (Fig. 2U,gray arrow). In sum, the histologic and immunohistochem-ical patterns observed in the tumors induced by MBPdonor plasmids were distinct from those observed with theGFAP and CAG donor plasmids, and are most similar tohuman anaplastic oligoastrocytoma (WHO grade 3).

Expression of bHLH transcription factors modifiestumor typeNext, we addressed whether addition of neurogenic tran-

scription factors, Neurogenin 2 (Ngn2) and Neuronal Dif-ferentiation 1 (NeuroD1), known to alter the fates of neuralprogenitors (32) would change tumor phenotype. We choseNgn2 and NeuroD1 because they have been previouslyshown to block the differentiation of neural progenitorsinto astrocytes and to promote differentiation into neurons(32). We hypothesized that transcription factors with neu-ron promoting effects would either inhibit tumor formation,or result in the formation of tumors distinct from CAGdonor plasmid–induced tumors. Indeed, expression of eitherNgn2 or NeuroD1 along with the CAG donor plasmidsresulted in a distinct tumor type that we never observed withany of the three other tumor-inducing plasmid combina-tions. Furthermore, animals transfected with the Ngn2- orNeuroD1-modifying plasmids showed earlier death andhigher rates of death than the other tumorigenic plasmidconditions (Fig. 3A). Log-rank tests showed animals trans-fected with Ngn2- or NeuroD1-modifying plasmids hadsignificantly shorter survivals (P < 0.0001) than animalstransfected with CAG, GFAP, or MBP donor plasmids(Fig. 3A).Upon postmortem analysis of cerebral tumors in Ngn2

and NeuroD1 donor plasmid conditions, we encounteredlarge cerebral tumors with highly irregular surfaces by 14 daysof age (Fig. 3B). Histologically, these large tumors containedcells that were poorly differentiated and showed regions ofextensive necrosis and proliferative features. The tumorscontained prominent rhabdoid cellular elements demon-strated in H&E sections (black arrow in Fig. 3H and L).The overall immunhistochemical profile indicated teratoidcharacteristics. Cells had characteristic "capping"-typeexpression patterns of vimentin (black arrow in Fig. 3I),and some cells were strongly positive for actin (Fig. 3J). Therewas also focal expression of epithelialmembrane antigen (Fig.3K). We also did not observe significant expression ofcytokeratin (Lu-5 epitope). Nuclear expression of integraseinteractor 1 (INI1; hSNF5/SMARCB1) was preserved in cellsin these tumors. Although poorly differentiated, the tumorsshowed regions of focal expression of astroglial GFAP (Sup-plementary Figs. S4B and S5C) and vimentin-positive pro-cesses (Fig. 3I and M) as well as scattered positivity for theneuronal markers MAP2 (Supplementary Figs. S4D andS5B) and synaptophysin (Supplementary Fig. S4C). Intrigu-ingly, synaptophysin positivity marked areas and clusters oftumor cells with cytologically neuronal features. The non-organoid distribution of respective elements argues againstentrapped neurons. MAP2c expression seemed to reflectdendritic neuronal structures. In sum, the tumors inducedby combining Ngn2 or NeuroD1 donor plasmids with theCAG donor plasmid histologlically most resembled atypicalteratoid rhabdoid tumor (ATRT) like tumors.Hertwig and colleagues (33) have reported that a panel of

7 signature genes can be used in error-free classification ofthree CNS tumors: glioma, primitive neuroectodermaltumor (PNET), and ATRT. We therefore assessed the

Determinants of Tumor Heterogeneity

www.aacrjournals.org Mol Cancer Res; 12(5) May 2014 747

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 7: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

expression of this panel of 7 genes in the tumors induced byHRasV12/AKT in combination with Ngn2. We foundsignificant upregulation in the expression of 6 out of 7 genesin the tumors induced by HRasV12/AKT in and Ngn2(Supplementary Fig. S6). Among the 6 upregulated genes,SPP1, which has been suggested as a diagnostic marker todistinguish ATRT from PNET and medulloblastoma (34)and as grade indicator of glioma (35), showed the greatest

upregulation (6.6-fold). The pattern of gene upregulationcombined with the histologic features suggests that thetumors induced by HRasV12/AKT in combination withNgn2 or NeuroD1 are ATRT-like tumors.As the bHLH transfection conditions resulted in a unique

and highly lethal tumor, we next addressed whether thisapparent phenotypic transformation from glioblastomamultiforme to ATRT-like tumors was due to a transient

Figure 3. ATRT like tumor was induced by addition of Ngn2 or NeuroD1 to PBCAG-HRasV12/AKT. A and B, Kaplan–Meier survival curves andrepresentative images of ATRT like tumor-bearing brains. ATRT like tumor-bearing animals had significantly short survival compared with glioblastomamultiforme and anaplastic oligoastrocytoma–bearing animals (log-rank test; �, P < 0.05; ���, P < 0.01). C to G, ATRT like tumor is induced by the additionof CAG-Ngn2 to PBCAG-HRasV12/AKT mixture. C, rhabdoid components (black arrow, inset), neuronal differentiation (gray arrows, H&E). D,vimentin rhabdoid cellular elements (black arrow, inset), and processes/glial differentiation (gray arrows). E, individual cells were actin-positive. F,clusters of cells express epithelial membrane antigen. G, few cells were positive for cytokeratin (Lu-5 epitope). H to K, ATRT like tumor is induced byaddition of PBCAG-NeuroD1 to PBCAG-HRasV12/AKT mixture. H, necrotizing tumor with mitoses, rhabdoid cellular differentiation (black arrow, inset),process-rich and epithelioid portions (H&E). I, vimentin-positive rhabdoid components (black arrow, inset) as well as vimentin-positiveprocesses (gray arrow). J, actin-positive cells. K, epithelial membrane antigen—clusters of positive cells. L to N, ATRT like tumor is induced by additionof PBCAG-Ngn2 to PBCAG-HRasV12/AKT mixture. L, H&E—tumor with high cellularity and undifferentiated appearance (mitotic figure, gray arrow;rhabdoid cell, black arrow, inset). M, vimentin staining, individual rhabdoid components (black arrow, inset) in an environment of a glial process-rich meshwork. N, clusters of actin-positive cells. Scale bar, 100 mm in C and E–N; 50 mm in D; and 10 mm in all insets.

Chen et al.

Mol Cancer Res; 12(5) May 2014 Molecular Cancer Research748

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 8: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

expression of bHLH factors in radial glia progenitors or toexpression in tumor cells. The piggyBac transposon systemallows for gating whether a transgene is integrated into atransfected cells or whether it remains episomal and is thuslost in cells that undergo subsequent proliferation (22). Wesubstituted the Ngn2 donor plasmids used above with aplasmid, CAG-Ngn2, in which the CAG-Ngn2 transgene isnot flanked by ITR sequences and so is not subject totransposase-mediated genomic integration (22). As a result,transgene expression from these episomal plasmids is lost in 1to 2 cell divisions (22, 23). Addition of episomal CAG-Ngn2was sufficient to produce tumors (Supplementary Fig.S3; Fig. 3C–E) with clusters of tumor cells expressingepithelia membrane antigen (Fig. 3F) and isolated cellsexpressing cytokeratin (Lu-5 epitope; Fig. 3G). NuclearINI1 was also preserved (data not shown). Thus, transientexpression of Ngn2 in radial glial progenitors and their

immediate progeny are sufficient to produce ATRT liketumors.

Distinct developmental patterns of tumorsThe four tumor-inducing conditions described above

(CAG donor plasmid, GFAP donor plasmid, MBP donorplasmid, and CAG donor plasmid with a bHLH transcrip-tion factor modifying donor plasmid) produced histologi-cally distinct tumors when assessed at similar postnataldevelopmental time points. We next sought to addresswhether these differences might be reflected in differencesin the developmental time course of each tumor type. Asshown in Fig. 4A–E, we found that the CAG donor plasmidcondition resulted in very early signs of abnormal cellproliferation with large aggregates of fluorescently labeledcells invading striatum and the ventricular and subventricularzones of neocortex by the day of birth (P0) in all animals

A A¢ B B¢ C D E

F F¢ G G¢ H I J

K K¢ L L¢ M N O

P P¢ Q R S T U

Figure 4. Induced tumors show distinct developmental time course. A to E, developmental time course for PBCAG-HRasV12/AKT–induced tumor. A, denselypacked cells were found in ventricular zone/subventricular zone, striatum, neocortex, and pia at P0.Magnified viewof boxed area is shown in A0. B, necrosis atP7. Magnified view of boxed area is shown in B0. C, representative image of PBCAG-HRasV12/AKT–transfected brain at P21. D, edge of PBCAG-HRasV12/AKT–induced tumor. DAPI (40,6-diamidino-2-phenylindole) is shown inmagenta. E, bipolar long spindle cells in PBCAG-HRasV12/AKT–induced tumors. F toJ, developmental time course for PBGFAP-HRasV12/AKT–induced tumor. F, a representative section from a P2 brain transfected with PBGFAP-HRasV12/AKT. Magnified view of boxed area is shown in F0. G, P9 section from PBGFAP-HRasV12/AKT–transfected brain. Magnified view of boxed area isshown in G0. H, representative image of PBGFAP-HRasV12/AKT–transfected brain at P21. I, edge of PBGFAP-HRasV12/AKT–induced tumor. DAPI isshown in magenta. J, bipolar pyramidal cells with long processes in tumors induced by PBGFAP-HRasV12/AKT. K to O, developmental time course forPBMBP-HRasV12/AKT–induced tumor. K, a representative section from a P3 brain transfected with PBMBP-HRasV12/AKT. Magnified view of boxed areais shown in K0. L, P7 section from PBMBP-HRasV12/AKT–transfected brain. Magnified view of boxed area is shown in L0. M, representative image ofPBMBP-HRasV12/AKT–transfected brain at P21. N, edge of PBMBP-HRasV12/AKT–induced tumor. DAPI is shown in magenta. O, small round cells withshort processes in tumors induced by PBMBP-HRasV12/AKT. P, a representative image section from a brain transfected with PBCAG-Ngn2, PBCAG-HRasV12/AKT at P0. Magnified view of boxed area is shown in P0. Q, a section from P21 animal showed tumor cells spreading whole cerebral hemisphere. R,edge of tumor induced by PBCAG-Ngn2, PBCAG-HRasV12/AKT. DAPI is shown in magenta. S and T, tumor cells frequently found in PBCAG-Ngn2–,PBCAG-HRasV12/AKT–induced tumors. U, extensive street-like necrosis. Scale bar, 1,000 mm inC, H,M, andQ; 500 mm inB, G, L, and P; 200 mm in A, F, andK; 100 mm in B0, D, G0 I, L0, N, P0, T, and U; and 50 mm in A0, E, F0, K0, J, O, S, and T.

Determinants of Tumor Heterogeneity

www.aacrjournals.org Mol Cancer Res; 12(5) May 2014 749

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 9: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

examined (3 of 3). In contrast, aggregates of proliferative cellswere not apparent in either the GFAP (0 of 3) orMBP donorplasmid (0 of 3) condition in the first few days after birth(P2–P3; Fig. 4F and K), but instead normally differentiatingneurons were apparent a few days after birth with onlyscattered cells outside of the neuronal cortical plate. By theend of the first postnatal week, masses of aberrantly prolif-erating cells seemed in both the MBP and GFAP donorplasmid conditions (Fig. 4G and L; 3 of 3 animals in MBP,GFAP donor–transfected animals, respectively). Thesemasses were often of a single clonally labeled color and werepresent in areas centered within white matter and the subpialzone (Fig. 4G and L). The time course of tumor growth wassimilar in the MBP and GFAP donor plasmid conditions;however, as indicated in the histopathologic analysis, themorphology of cells in the two conditions differed in mor-phology by P21 (Fig. 4H–J and M–O). In spite of thedelayed appearance of large tumors in the GFAP and MBPdonor plasmid conditions relative to the CAG donor plas-mid, the size of tumors in theMBP andGFAPdonor plasmidconditions by P21 reached sizes larger than those in the CAGdonor plasmid condition (Fig. 4C, H, and M). Finally,consistent with the early lethality and highly aggressivenature of the tumors induced by CAG donor plasmids withaddition of Ngn2-modifying plasmid, the tumors in thiscondition (Fig. 4P–U) were obvious by the day of birth (4 of4) and expanded rapidly into very large tumors with street-like necroses invading the entire cerebral hemisphere by 3weeks after birth (7 of 7). The developmental patternsobserved are consistent with when the promoters expressingthe oncogenes are most active; the ubiquitous CAG pro-moter is strongly active early in progenitors, whereas theGFAP and MBP promoters are most active later in thelineage when glial cells begin differentiating.

Gene expression differences in tumor typesThe Cancer Genome Atlas research network identified

four subtypes of glioblastoma multiforme subtypes by geneexpression profiles and gene mutation (1). Through thisanalysis, a group of 24 signature genes was identified thatcould be used to categorize the four subtypes (1). Wetherefore used expression levels of these 24 signature genesto further assess whether the four tumor patterns producedin our study could be distinguished based on the expressionof this set of genes. To do this, we used q-PCR andunsupervised hierarchical clustering analysis to compare andcategorize the tumors produced by the four conditions. Wefound, consistent with the agreement in histopathologicanalysis, that the tumors produced by the CAG and GFAPdonor plasmids clustered together in terms of expression ofthe 24 genes (Fig. 5). The tumors that were most distinct inhistopathology, those produced by theMBP donor plasmidsand modified by the Ngn2 donor plasmids, differed in geneexpression patterns relative both to each other and to theCAG and GFAP donor plasmid conditions (Fig. 5). Epi-dermal growth factor receptor (EGFR) is frequently ampli-fied and overexpressed in human glioblastoma multiformes;however, EGFR transcript was not prominent in all tumor

types. This might be because Ras and AKT are downstreamfactors of the EGFR pathway (15). We also found that p53was neither mutated nor deleted (data not shown), and p53transcript showed about 2-fold of upregulation. Thus, theheterogeneity we find in tumors produced by differingpromoter conditions and modifying transcription factorexpression is also reflected in different gene expressionpatterns.

DiscussionTwo, not mutually exclusive, explanations of tumor het-

erogeneity have been proposed (36). Overwhelming exper-imental and clinical evidence shows that different geneticmutations result in different tumor types including differentCNS tumor types (37). For example, Hertwig and colleagues(33) showed that infection of postnatal mouse neural stemcells with viruses containing V12HRAS or c-MYC couldresult in formation of 3 different tumor types dependingupon the combination and sequence in which oncogeneswere introduced. Similarly, Jacques and colleagues (38)showed that different combinations of conditional geneticdeletions in p53, retinoblastoma, and PTEN in mousesubventricular zone neural stem cells could induce formationof either PNET or glioma. Evidence for differing cell typesbeing a source of CNS tumor heterogeneity has also beenfound (39–42). For example, transduction of mutationallystabilized N-Myc into neural stem cells from perinatalmurine cerebellum and brain stem resulted in formationof medulloblastoma/primitive neuroectodermal tumors,whereas N-Myc transduced into neural stem cells (NSCs)isolated from forebrain resulted in diffuse glioma tumors(41). As different neural progenitor types have differentgene expression profiles that may modify tumor celldifferentiation, it seems likely that both cell-of-origin anddifferences in gene mutation contribute to tumor-typediversity in the CNS (36). Our study indicates that in thedeveloping neocortex, cell populations of origin can con-tribute to tumor diversity, and moreover, that expressionof nononcogenic transcription factors expressed in thesame population can also modify tumor type.Although pediatric glioblastoma multiforme shares histo-

logic similarities with adult glioblastoma multiforme, theyare now thought to be different entities with differentmolecular characteristics (2, 43). It has been shown, forexample, that pediatric glioblastoma multiformes display aspectrum of copy-number variations distinct from adultglioblastoma multiforme (44). Integrated molecular profil-ing experiments also reveal differences between pediatric andadult glioblastomamultiforme. For example, isocitrate dehy-drogenase (IDH) hotspot mutations are frequently found inadult high-grade glioma but not in pediatric tumors (2, 43).In addition, whereas PDGFRA is the predominant target offocal amplification in pediatric high-grade glioma, in adultglioblastoma, EGFR is the most common target (43). More-over, histone H3.3 mutations are frequently found in pedi-atric glioblastoma multiforme, whereas they are absent inadult glioblastoma multiforme (2, 45). The cell-of-origin of

Chen et al.

Mol Cancer Res; 12(5) May 2014 Molecular Cancer Research750

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 10: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

pediatric glioblastomamultiforme is unknown andmight bedifferent from adult glioblastoma multiforme as well. Wehave used the CAG donor plasmids to induce glioblastomamultiforme, which seemed as early as P7 in the rat and showsnecrotic areas as early as at P4, a developmental time periodin the precocial rat that corresponds to the neonatal period inhuman. Future experiments will be needed to determinewhether the early-arising tumors modeled in this system aremore similar to pediatric or adult glioblastomamultiforme intheir molecular identity and cell-of-origin.Our study indicates that the cell of mutation for both

ATRT like tumors and glioblastoma multiforme can beradial glial cells of embryonic neocortex. The in uteroelectroporation method we used targets radial glia and radialneural progenitor cells that line the lateral ventricles of the

lateral forebrain. This population of progenitors at theventricular surface of E14/15 rat forebrain contains sub-populations of progenitors capable of generating neurons ofdifferent types, and primarily glia or primarily neurons (23).We used the ubiquitous CAG donor plasmid to induceexpression of HRasV12 and AKT immediately in the radialprogenitor population. Ngn2 or NeuroD1 expressed by thesame immediately active promoter was sufficient to changethe tumor type generated. The tumor difference was appar-ent as early as the day of birth, approximately 1 week afterinduced gene expression. We also found that transientexpression of Ngn2 (by a nondonor episomal plasmid) inradial glia was sufficient to produce ATRTs. The effective-ness of transient Ngn2 expression, in combination withprevious findings that Ngn2 or NeuroD1 expressed in

Figure 5. Induced tumors showdifferent molecular signature. A,unsupervised cluster analysis ofqRT-PCR–based gene expressiondata for 24 genes selected fromVerhaak et al. (1) across differentdonor plasmid–induced tumors.The heatmap shows fold change ingene expression in induced tumorsrelative to tumor-free brain tissues.The 24 genes screened are listedon the left. Color scale representsfold changesof expressionwith redindicating upregulation and greenindicating downregulation of geneexpression. Gene expressionacross four conditions was shownin rows and expression of thewhole set of 24 gene in each tumorsample was shown in columns.B, averaged fold of regulation for6 representative genes selectedfrom A. Error bar, SEM. One-wayANOVA; �, P < 0.05; ���, P < 0.01.

Determinants of Tumor Heterogeneity

www.aacrjournals.org Mol Cancer Res; 12(5) May 2014 751

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 11: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

glioma cells induces cell death and neuronal differentiationwithout changing tumor type (46, 47), supports the idea thatNgn2 and NeuroD1 act in early-stage radial progenitors tochange the type of tumor generated. We have coexpressedNgn2 with GFAP donor plasmids and found that theresulted tumors were similar to tumors induced by GFAPdonor plasmids alone. But we did found rhabdoidal cells inthe resulted tumors (data not shown). However, the resultshave to be interpreted with caution because the mouseGFAP promoter fragment is also active in radial glia. In thefuture, combining Ngn2 with MBP donor plasmids maydistinguish the time of action of Ngn2.Loss of function in the INI1 gene is believed to be a

significant cause of ATRT in humans (48). INI1 knock-out mice develop tumors but these do not seem to beATRT (49). Hertwig and colleagues (33) showed thattransplantation of NSC/neural progenitor cells (NPCs)serially infected with c-MYC and V12HRAS could gen-erate ATRTs with gene expression profiles similar tohuman ATRT, suggesting that in rodent models addi-tional mutation types can lead to ATRT. Similarly, wedemonstrated in this study that ATRT like tumors aregenerated by HRasV12 and AKT transfection of neocor-tical radial glia, but only when coexpressed with thebHLH transcription factors Ngn2 or NeuroD1, two genesthat on their own are nononcogenic. This underscores thestrong possibility that tumor-type diversity may be influ-enced not only by the cell of mutation but also by thespecific molecular context present in that cell.Our current results also show that the GFAP and MBP

promoter–active populations in the radial glia lineagegenerate tumors with different molecular and histologicfeatures. Interestingly, all cell types are labeled by theGFAP and MBP donor plasmids, but yet the tumor typesgenerated were consistently different in histology andmolecular signature. This may suggest that tumor diversityis determined by the predominant cell type in a popula-tion that undergoes transformation. By mixing the GFAPand MBP donor plasmids in future experiments andtracking their clonal expansion, we may be able to dis-tinguish whether one tumor cell population is dominantover the other.

Several attributes of piggyBac transposon system dem-onstrated here make this model potentially useful for avariety of novel applications in tumor biology. The mainadvantage of the piggyBac IUE method is that it allows forintroduction of multiple transgenes. In this study, forexample, we simultaneously introduced a transposasehelper plasmid to target stable transgenesis in GLAST-positive cells, two oncogene-expressing donor plasmidswith their own promoters, 3 fluorescent reporter genes(Figs. 1, 2, and 3), and a transcription factor. The highcoexpression efficiency allowed us to direct expression indifferent subpopulations in sequence and to introduce aclonal labeling method and a modifying transcriptionfactor. This functionality should make this approach auseful platform for screening potential modifiers of tumordevelopment and for determining further how geneticmodifiers alter tumor development.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: F. Chen, J.J. LoTurcoDevelopment of methodology: F. Chen, J.J. LoTurcoAcquisition of data (provided animals, acquired and managed patients, providedfacilities, etc.): F. Chen, A.J. Becker, J.J. LoTurcoAnalysis and interpretation of data (e.g., statistical analysis, biostatistics, compu-tational analysis): F. Chen, A.J. Becker, J.J. LoTurcoWriting, review, and/or revision of the manuscript: F. Chen, A.J. Becker,J.J. LoTurcoAdministrative, technical, or material support (i.e., reporting or organizing data,constructing databases): J.J. LoTurco

AcknowledgmentsThe authors thank Dr. Akiko Nishiyama (University of Connecticut) for kindly

providingNG2 andCC1 antibodies, Dr.William Sellers (Harvardmedical school ) forsharing pcDNA3 Myr HA Akt1 construct, and Dr. Inder Verma (Salk Institute) forsharing pTomo construct.

Grant supportThis work was supported by NIH grants (RO1HD055655 and R01MH056524,

to J.J. LoTurco).The costs of publication of this article were defrayed in part by the payment

of page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received October 4, 2013; revised January 20, 2014; accepted January 24, 2014;published OnlineFirst February 5, 2014.

References1. VerhaakRG,HoadleyKA, PurdomE,WangV,QiY,WilkersonMD, et al.

Integrated genomic analysis identifies clinically relevant subtypes ofglioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR,and NF1. Cancer Cell 2010;17:98–110.

2. Sturm D, Witt H, Hovestadt V, Khuong-Quang DA, Jones DT, Koner-mann C, et al. Hotspot mutations in H3F3A and IDH1 define distinctepigenetic and biological subgroups of glioblastoma. Cancer Cell2012;22:425–37.

3. Haque T, Faury D, Albrecht S, Lopez-Aguilar E, Hauser P, Garami M,et al. Gene expression profiling from formalin-fixed paraffin-embed-ded tumors of pediatric glioblastoma. Clin Cancer Res 2007;13:6284–92.

4. Faury D, Nantel A, Dunn SE, Guiot MC, Haque T, Hauser P, et al.Molecular profiling identifies prognostic subgroups of pediatric glio-

blastoma and shows increased YB-1 expression in tumors. J ClinOncol 2007;25:1196–208.

5. Persson AI, Petritsch C, Swartling FJ, ItsaraM, Sim FJ, Auvergne R, et al.Non-stemcell origin for oligodendroglioma. Cancer Cell 2010;18:669–82.

6. Liu C, Sage JC, Miller MR, Verhaak RG, Hippenmeyer S, Vogel H, et al.Mosaic analysis with double markers reveals tumor cell of origin inglioma. Cell 2011;146:209–21.

7. Uhrbom L, Hesselager G, Nister M, Westermark B. Induction of braintumors in mice using a recombinant platelet-derived growth factor B-chain retrovirus. Cancer Res 1998;58:5275–9.

8. Holland EC, Hively WP, DePinho RA, Varmus HE. A constitutivelyactive epidermal growth factor receptor cooperates with disruption ofG1 cell-cycle arrest pathways to induce glioma-like lesions in mice.Genes Dev 1998;12:3675–85.

Mol Cancer Res; 12(5) May 2014 Molecular Cancer Research752

Chen et al.

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 12: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

9. Holland EC, Celestino J, Dai C, Schaefer L, Sawaya RE, Fuller GN.Combined activation of Ras and Akt in neural progenitors inducesglioblastoma formation in mice. Nat Genet 2000;25:55–7.

10. UhrbomL,Dai C,Celestino JC, RosenblumMK, Fuller GN,Holland EC.Ink4a-Arf loss cooperates with KRas activation in astrocytes andneural progenitors to generate glioblastomas of various morphologiesdepending on activated Akt. Cancer Res 2002;62:5551–8.

11. Hambardzumyan D, Amankulor NM, Helmy KY, Becher OJ, HollandEC.Modeling adult gliomas usingRCAS/t-va technology. TranslOncol2009;2:89–95.

12. Holmen SL, Williams BO. Essential role for Ras signaling in glioblas-toma maintenance. Cancer Res 2005;65:8250–5.

13. Lindberg N, Kastemar M, Olofsson T, Smits A, Uhrbom L. Oligoden-drocyte progenitor cells can act as cell of origin for experimentalglioma. Oncogene 2009;28:2266–75.

14. Alcantara Llaguno S, Chen J, Kwon CH, Jackson EL, Li Y, Burns DK,et al. Malignant astrocytomas originate from neural stem/progenitorcells in a somatic tumor suppressor mouse model. Cancer Cell2009;15:45–56.

15. Marumoto T, Tashiro A, Friedmann-Morvinski D, Scadeng M, Soda Y,Gage FH, et al. Development of a novel mouse glioma model usinglentiviral vectors. Nat Med 2009;15:110–6.

16. Friedmann-Morvinski D, Bushong EA, Ke E, Soda Y, Marumoto T,Singer O, et al. Dedifferentiation of neurons and astrocytes by onco-genes can induce gliomas in mice. Science 2012;338:1080–4.

17. Wiesner SM, Decker SA, Larson JD, Ericson K, Forster C, Gallardo JL,et al. De novo induction of genetically engineered brain tumors in miceusing plasmid DNA. Cancer Res 2009;69:431–9.

18. Lei L, Sonabend AM, Guarnieri P, Soderquist C, Ludwig T, Rosen-feld S, et al. Glioblastoma models reveal the connection betweenadult glial progenitors and the proneural phenotype. PLoS ONE2011;6:e20041.

19. Chen F, LoTurco J. A method for stable transgenesis of radial glialineage in rat neocortex by piggyBac mediated transposition. J Neu-rosci Methods 2012;207:172–80.

20. Siddiqi F, Chen F, Aron AW, Fiondella CG, Patel K, Loturco JJ. Fatemapping by PiggyBac transposase reveals that neocortical GLASTþprogenitors generate more astrocytes than nestinþ progenitors in ratneocortex. Cereb Cortex 2014;24:508–20.

21. Yusa K, Rad R, Takeda J, Bradley A. Generation of transgene-freeinduced pluripotent mouse stem cells by the piggyBac transposon.Nat Methods 2009;6:363–9.

22. LoTurco J, Manent JB, Sidiqi F. New and improved tools for in uteroelectroporation studies of developing cerebral cortex. Cereb Cortex2009;19 Suppl 1:i120–5.

23. Lu Y, Lin C, Wang X. PiggyBac transgenic strategies in the developingchicken spinal cord. Nucleic Acids Res 2009;37:e141.

24. Yoshida A, Yamaguchi Y, Nonomura K, Kawakami K, Takahashi Y,Miura M. Simultaneous expression of different transgenes in neuronsand glia by combining in utero electroporation with the Tol2 transpo-son-mediated gene transfer system. Genes Cells 2010;15:501–12.

25. Guha A, Feldkamp MM, Lau N, Boss G, Pawson A. Proliferation ofhuman malignant astrocytomas is dependent on Ras activation.Oncogene 1997;15:2755–65.

26. Ramaswamy S, Nakamura N, Vazquez F, Batt DB, Perera S, RobertsTM, et al. Regulation of G1 progression by the PTEN tumor suppressorprotein is linked to inhibition of the phosphatidylinositol 3-kinase/Aktpathway. Proc Natl Acad Sci U S A 1999;96:2110–5.

27. Centanni TM, Booker AB, Sloan AM, Chen F, Maher BJ, Carraway RS,et al. Knockdown of the dyslexia-associated gene Kiaa0319 impairstemporal responses to speech stimuli in rat primary auditory cortex.Cereb Cortex 2013 Feb 8. [Epub ahead of print].

28. Schick V, Majores M, Koch A, Elger CE, Schramm J, Urbach H, et al.Alterations of phosphatidylinositol 3-kinase pathway components in

epilepsy-associated glioneuronal lesions. Epilepsia 2007;48 Suppl5:65–73.

29. Schick V, Majores M, Engels G, Hartmann W, Elger CE, Schramm J,et al. Differential Pi3K-pathway activation in cortical tubers and focalcortical dysplasias with balloon cells. Brain Pathol 2007;17:165–73.

30. Weber K, Thomaschewski M, Warlich M, Volz T, Cornils K, Niebuhr B,et al. RGB marking facilitates multicolor clonal cell tracking. Nat Med2011;17:504–9.

31. WeiQ,MiskiminsWK,MiskiminsR.Cloning andcharacterization of therat myelin basic protein gene promoter. Gene 2003;313:161–7.

32. Cai L, Morrow EM, Cepko CL. Misexpression of basic helix-loop-helixgenes in the murine cerebral cortex affects cell fate choices andneuronal survival. Development 2000;127:3021–30.

33. Hertwig F, Meyer K, Braun S, Ek S, Spang R, Pfenninger CV, et al.Definition of genetic events directing the development of distinct typesof brain tumors from postnatal neural stem/progenitor cells. CancerRes 2012;72:3381–92.

34. Kao CL, Chiou SH, Ho DM, Chen YJ, Liu RS, Lo CW, et al. Elevation ofplasma and cerebrospinal fluid osteopontin levels in patients withatypical teratoid/rhabdoid tumor. Am JClin Pathol 2005;123:297–304.

35. Toy H, Yavas O, Eren O, Genc M, Yavas C. Correlation betweenosteopontin protein expression and histological grade of astrocyto-mas. Pathol Oncol Res 2009;15:203–7.

36. Visvader JE. Cells of origin in cancer. Nature 2011;469:314–22.37. Sieber OM, Tomlinson SR, Tomlinson IP. Tissue, cell and stage spec-

ificity of (epi)mutations in cancers. Nat Rev Cancer 2005;5:649–55.38. Jacques TS, Swales A, Brzozowski MJ, Henriquez NV, Linehan JM,

MirzadehZ, et al. Combinations of geneticmutations in the adult neuralstem cell compartment determine brain tumour phenotypes. EMBO J2010;29:222–35.

39. Gibson P, Tong Y, Robinson G, Thompson MC, Currle DS, Eden C,et al. Subtypes of medulloblastoma have distinct developmentalorigins. Nature 2010;468:1095–9.

40. InceTA,RichardsonAL,Bell GW,SaitohM,GodarS, KarnoubAE, et al.Transformation of different human breast epithelial cell types leads todistinct tumor phenotypes. Cancer Cell 2007;12:160–70.

41. Swartling FJ, Savov V, Persson AI, Chen J, Hackett CS, Northcott PA,et al. Distinct neural stem cell populations give rise to disparate braintumors in response to N-MYC. Cancer Cell 2012;21:601–13.

42. Brown K, Strathdee D, Bryson S, LambieW, Balmain A. The malignantcapacity of skin tumours induced by expression of a mutant H-rastransgene depends on the cell type targeted. Curr Biol 1998;8:516–24.

43. Paugh BS, Qu C, Jones C, Liu Z, Adamowicz-Brice M, Zhang J, et al.Integrated molecular genetic profiling of pediatric high-grade gliomasreveals key differences with the adult disease. J Clin Oncol 2010;28:3061–8.

44. Bax DA, Mackay A, Little SE, Carvalho D, Viana-Pereira M, Tamber N,et al. A distinct spectrum of copy number aberrations in pediatric high-grade gliomas. Clin Cancer Res 2010;16:3368–77.

45. Schwartzentruber J, Korshunov A, Liu XY, Jones DT, Pfaff E, Jacob K,et al. Driver mutations in histone H3.3 and chromatin remodellinggenes in paediatric glioblastoma. Nature 2012;482:226–31.

46. Zhao J, He H, Zhou K, Ren Y, Shi Z, Wu Z, et al. Neuronal transcriptionfactors induce conversion of human glioma cells to neurons and inhibittumorigenesis. PLoS ONE 2012;7:e41506.

47. Guichet PO, Bieche I, Teigell M, Serguera C, Rothhut B, Rigau V, et al.Cell death and neuronal differentiation of glioblastoma stem-like cellsinduced by neurogenic transcription factors. Glia 2012;61:225–39.

48. Biegel JA. Molecular genetics of atypical teratoid/rhabdoid tumor.Neurosurg Focus 2006;20:E11.

49. Klochendler-Yeivin A, Fiette L, Barra J, Muchardt C, Babinet C, YanivM. The murine SNF5/INI1 chromatin remodeling factor is essential forembryonic development and tumor suppression. EMBO Rep 2000;1:500–6.

www.aacrjournals.org Mol Cancer Res; 12(5) May 2014 753

Determinants of Tumor Heterogeneity

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531

Page 13: Contribution of Tumor Heterogeneity in a New Animal Model of … · Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531 Forhistologicanalysis,immunohistochemistryandhema-

2014;12:742-753. Published OnlineFirst February 5, 2014.Mol Cancer Res   Fuyi Chen, Albert J. Becker and Joseph J. LoTurco  CNS TumorsContribution of Tumor Heterogeneity in a New Animal Model of

  Updated version

  10.1158/1541-7786.MCR-13-0531doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mcr.aacrjournals.org/content/suppl/2014/02/05/1541-7786.MCR-13-0531.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mcr.aacrjournals.org/content/12/5/742.full#ref-list-1

This article cites 48 articles, 15 of which you can access for free at:

  Citing articles

  http://mcr.aacrjournals.org/content/12/5/742.full#related-urls

This article has been cited by 2 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mcr.aacrjournals.org/content/12/5/742To request permission to re-use all or part of this article, use this link

on June 20, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst February 5, 2014; DOI: 10.1158/1541-7786.MCR-13-0531