15
Cancer Genes and Networks TFE3 Xp11.2 Translocation Renal Cell Carcinoma Mouse Model Reveals Novel Therapeutic Targets and Identies GPNMB as a Diagnostic Marker for Human Disease Masaya Baba 1,2 , Mitsuko Furuya 3 ,Takanobu Motoshima 4 , Martin Lang 2 , Shintaro Funasaki 1 , Wenjuan Ma 1 , Hong-Wei Sun 5 , Hisashi Hasumi 2,6 , Ying Huang 2 , Ikuma Kato 3 , Tsuyoshi Kadomatsu 7 , Yorifumi Satou 8 , Nicole Morris 9 , Baktiar O. Karim 10 , Lilia Ileva 11 , Joseph D. Kalen 11 , Luh Ade Wilan Krisna 1 , Yukiko Hasumi 2 , Aiko Sugiyama 12 , Ryoma Kurahashi 4,7 , Koshiro Nishimoto 13 , Masafumi Oyama 13 , Yoji Nagashima 14 , Naoto Kuroda 15 , Kimi Araki 16 , Masatoshi Eto 17 , Masahiro Yao 6 , Tomomi Kamba 4 , Toshio Suda 18,19 , Yuichi Oike 7 , Laura S. Schmidt 2,20 , and W. Marston Linehan 2 Abstract Renal cell carcinoma (RCC) associated with Xp11.2 trans- location (TFE3-RCC) has been recently dened as a distinct subset of RCC classied by characteristic morphology and clinical presentation. The Xp11 translocations involve the TFE3 transcription factor and produce chimeric TFE3 proteins retaining the basic helixloophelix leucine zipper structure for dimerization and DNA binding suggesting that chimeric TFE3 proteins function as oncogenic transcription factors. Diagnostic biomarkers and effective forms of therapy for advanced cases of TFE3-RCC are as yet unavailable. To facil- itate the development of molecular based diagnostic tools and targeted therapies for this aggressive kidney cancer, we gener- ated a translocation RCC mouse model, in which the PRCC- TFE3 transgene is expressed specically in kidneys leading to the development of RCC with characteristic histology. Expres- sion of the receptor tyrosine kinase Ret was elevated in the kidneys of the TFE3-RCC mice, and treatment with RET inhibitor, vandetanib, signicantly suppressed RCC growth. Moreover, we found that Gpnmb (Glycoprotein nonmetastatic B) expression was notably elevated in the TFE3-RCC mouse kidneys as seen in human TFE3-RCC tumors, and conrmed that GPNMB is the direct transcriptional target of TFE3 fusions. While GPNMB IHC staining was positive in 9/9 cases of TFE3- RCC, Cathepsin K, a conventional marker for TFE3-RCC, was positive in only 67% of cases. These data support RET as a potential target and GPNMB as a diagnostic marker for TFE3- RCC. The TFE3-RCC mouse provides a preclinical in vivo model for the development of new biomarkers and targeted therapeutics for patients affected with this aggressive form of RCC. Implications: Key ndings from studies with this preclinical mouse model of TFE3-RCC underscore the potential for RET as a therapeutic target for treatment of patients with TFE3-RCC, and suggest that GPNMB may serve as diagnostic biomarker for TFE3 fusion RCC. 1 Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan. 2 Urologic Oncol- ogy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland. 3 Department of Molecular Pathology, Yokohama City University, Yokohama, Japan. 4 Department of Urology, Kuma- moto University, Kumamoto, Japan. 5 Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, Maryland. 6 Department of Urology, Yokohama City University, Yokohama, Japan. 7 Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan. 8 Laboratory of Retroviral Genomics and Tran- scriptomics, International Research Center for Medical Sciences (IRCMS), Center for AIDS Research, Kumamoto University, Kumamoto, Japan. 9 Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland. 10 Pathology/Histotechnology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland. 11 Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland. 12 DSK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan. 13 Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama, Japan. 14 Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan. 15 Department of Pathology, Kochi Red Cross Hospital, Kochi, Japan. 16 Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan. 17 Department of Urology, Kyushyu University, Fukuoka, Japan. 18 Laboratory of Stem Cell Regulation, International Research Center for Medical Sciences (IRCMS), Kumamoto Uni- versity, Kumamoto, Japan. 19 Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore. 20 Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland. Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). Corresponding Authors: W. Marston Linehan, National Cancer Institute, 10 Center Drive MSC 1107, Bethesda, MD 20892-1107. Phone: 240-858-3700; Fax: 301-402-0922; E-mail: [email protected]; Laura S. Schmidt, Phone: 240-858-3939; Fax: 301-480-3195; E-mail: [email protected]; and Masaya Baba, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan. Phone: 81-96-373-6836; Fax: 81-96-373-6869; E-mail: [email protected] Mol Cancer Res 2019;XX:XXXX doi: 10.1158/1541-7786.MCR-18-1235 Ó2019 American Association for Cancer Research. Molecular Cancer Research www.aacrjournals.org OF1 on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

TFE3 Xp11.2 Translocation Renal Cell Carcinoma Mouse Model … · 2019. 5. 22. · described in a pediatric renal cell carcinoma (RCC) case in 1986, and the fusion gene was confirmed

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • Cancer Genes and Networks

    TFE3 Xp11.2 Translocation Renal Cell CarcinomaMouse Model Reveals Novel Therapeutic Targetsand Identifies GPNMB as a Diagnostic Marker forHuman DiseaseMasayaBaba1,2, MitsukoFuruya3,TakanobuMotoshima4,Martin Lang2, ShintaroFunasaki1,Wenjuan Ma1, Hong-Wei Sun5, Hisashi Hasumi2,6, Ying Huang2, Ikuma Kato3,Tsuyoshi Kadomatsu7, Yorifumi Satou8, Nicole Morris9, Baktiar O. Karim10, Lilia Ileva11,Joseph D. Kalen11, Luh Ade Wilan Krisna1, Yukiko Hasumi2, Aiko Sugiyama12,Ryoma Kurahashi4,7, Koshiro Nishimoto13, Masafumi Oyama13, Yoji Nagashima14,Naoto Kuroda15, Kimi Araki16, Masatoshi Eto17, Masahiro Yao6, Tomomi Kamba4,Toshio Suda18,19, Yuichi Oike7, Laura S. Schmidt2,20, and W. Marston Linehan2

    Abstract

    Renal cell carcinoma (RCC) associated with Xp11.2 trans-location (TFE3-RCC) has been recently defined as a distinctsubset of RCC classified by characteristic morphology andclinical presentation. The Xp11 translocations involve theTFE3 transcription factor and produce chimeric TFE3 proteinsretaining the basic helix–loop–helix leucine zipper structurefor dimerization and DNA binding suggesting that chimericTFE3 proteins function as oncogenic transcription factors.Diagnostic biomarkers and effective forms of therapy foradvanced cases of TFE3-RCC are as yet unavailable. To facil-itate the development ofmolecular based diagnostic tools andtargeted therapies for this aggressive kidney cancer, we gener-ated a translocation RCC mouse model, in which the PRCC-TFE3 transgene is expressed specifically in kidneys leading tothe development of RCC with characteristic histology. Expres-sion of the receptor tyrosine kinase Ret was elevated in thekidneys of the TFE3-RCC mice, and treatment with RETinhibitor, vandetanib, significantly suppressed RCC growth.

    Moreover, we found thatGpnmb (Glycoprotein nonmetastaticB) expression was notably elevated in the TFE3-RCC mousekidneys as seen in human TFE3-RCC tumors, and confirmedthatGPNMB is the direct transcriptional target of TFE3 fusions.While GPNMB IHC staining was positive in 9/9 cases of TFE3-RCC, Cathepsin K, a conventional marker for TFE3-RCC, waspositive in only 67% of cases. These data support RET as apotential target and GPNMB as a diagnostic marker for TFE3-RCC. The TFE3-RCC mouse provides a preclinical in vivomodel for the development of new biomarkers and targetedtherapeutics for patients affected with this aggressive form ofRCC.

    Implications: Key findings from studies with this preclinicalmousemodel of TFE3-RCCunderscore the potential for RET asa therapeutic target for treatment of patients with TFE3-RCC,and suggest that GPNMB may serve as diagnostic biomarkerfor TFE3 fusion RCC.

    1Laboratory of Cancer Metabolism, International Research Center for MedicalSciences (IRCMS), Kumamoto University, Kumamoto, Japan. 2Urologic Oncol-ogy Branch, Center for Cancer Research, National Cancer Institute, NationalInstitutes of Health, Bethesda, Maryland. 3Department of Molecular Pathology,Yokohama City University, Yokohama, Japan. 4Department of Urology, Kuma-moto University, Kumamoto, Japan. 5Biodata Mining and Discovery Section,NIAMS, NIH, Bethesda, Maryland. 6Department of Urology, Yokohama CityUniversity, Yokohama, Japan. 7Department of Molecular Genetics, KumamotoUniversity, Kumamoto, Japan. 8Laboratory of Retroviral Genomics and Tran-scriptomics, International Research Center for Medical Sciences (IRCMS), Centerfor AIDS Research, Kumamoto University, Kumamoto, Japan. 9LaboratoryAnimal Sciences Program, Frederick National Laboratory for Cancer Research,Frederick, Maryland. 10Pathology/Histotechnology Laboratory, FrederickNational Laboratory for Cancer Research, Frederick, Maryland. 11Small AnimalImagingProgram, FrederickNational Laboratory forCancerResearch, Frederick,Maryland. 12DSK Project, Medical Innovation Center, Kyoto University GraduateSchool of Medicine, Sakyo-ku, Kyoto, Japan. 13Department of Uro-Oncology,Saitama Medical University International Medical Center, Saitama, Japan.14Department of Surgical Pathology, TokyoWomen's Medical University, Tokyo,Japan. 15Department of Pathology, Kochi Red Cross Hospital, Kochi, Japan.16Division of Developmental Genetics, Institute of Resource Development andAnalysis, Kumamoto University, Kumamoto, Japan. 17Department of Urology,

    Kyushyu University, Fukuoka, Japan. 18Laboratory of Stem Cell Regulation,International Research Center for Medical Sciences (IRCMS), Kumamoto Uni-versity, Kumamoto, Japan. 19Cancer Science Institute of Singapore, NationalUniversity of Singapore, Centre for Translational Medicine, Singapore. 20BasicScience Program, Frederick National Laboratory for Cancer Research, Frederick,Maryland.

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

    Corresponding Authors: W. Marston Linehan, National Cancer Institute, 10Center Drive MSC 1107, Bethesda, MD 20892-1107. Phone: 240-858-3700;Fax: 301-402-0922; E-mail: [email protected]; Laura S. Schmidt, Phone:240-858-3939; Fax: 301-480-3195; E-mail: [email protected]; and MasayaBaba, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.Phone: 81-96-373-6836; Fax: 81-96-373-6869; E-mail:[email protected]

    Mol Cancer Res 2019;XX:XX–XX

    doi: 10.1158/1541-7786.MCR-18-1235

    �2019 American Association for Cancer Research.

    MolecularCancerResearch

    www.aacrjournals.org OF1

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://crossmark.crossref.org/dialog/?doi=10.1158/1541-7786.MCR-18-1235&domain=pdf&date_stamp=2019-5-23http://mcr.aacrjournals.org/

  • IntroductionXp11.2 translocation, t(X;1)(p11.2;q21.2), was first

    described in a pediatric renal cell carcinoma (RCC) case in1986, and the fusion gene was confirmed to be PRCC-TFE3 in1995 (1, 2). TFE3 Xp11.2 translocation RCC (TFE3-RCC) wasdefined as an independent subtype of RCC by WHO in 2004and is characterized by distinctive morphologic features andXp11.2 rearrangements that create TFE3 gene fusions with avariety of partner genes (PRCC, SFPQ, ASPSCR, CLTC, NONO,RBM10, PARP14, LUC7L3, KHSRP etc.; refs. 2–17). TFE3encodes a transcription factor that has a basic helix–loop–helixleucine zipper (bHLH-Zip) structure through which TFE3dimerizes and interacts with M-box DNA sequences(TCAYRTGA) in transcriptional target genes. All TFE3 fusiongenes encode in-frame chimeric proteins, which retain thebHLH-Zip domain of TFE3 (16, 18). Nuclear accumulation ofTFE3 is one of the most significant histopathologic character-istics of TFE3-RCC (19, 20). The evidence is strong for TFE3fusions to be oncogenes with constitutively active transcrip-tional activity.

    TFE3-RCC is more common than was previously thought,comprising from 2% to 5% of adult cases (21, 22) and from25% to 40% of pediatric RCC cases (14, 23). TFE3-RCC is knownfor its aggressive malignant nature with a propensity to metasta-size when the primary tumor is small. There is currently nostandard or effective form of therapy for patients with advanceddisease (4, 16). Reduced awareness of TFE3-RCCand the technicalcomplexity of diagnosis including TFE3 staining and TFE3 genebreak-apart FISH have led to a decrease in awareness and earlydiagnosis of this disease (21, 24–26). It is, therefore, important todevelop novel diagnostic methods for TFE3-RCC. Although sev-eral diagnostic markers for TFE3-RCC have been reported, such asCathepsin K, melan A, and HMB45, the sensitivity and specificityof these conventional markers are limited and not robust enoughto confirm the diagnosis of TFE3-RCC (20, 26, 27). Transcrip-tional target genes of TFE3 that are upregulated following TFE3nuclear localization and activation could potentially be usefulmarkers for the diagnosis of TFE3-RCC.

    In this study, we have generated a TFE3-RCCmousemodel thatexpresses PRCC-TFE3, which is the first reported TFE3 fusionpartner and frequently observed in human disease, specificallyin kidney epithelial cells, and develops a variety of kidney epi-thelial neoplastic lesions including hyperplastic cysts, adenomas,and solid tumors. This mouse model provides a preclinical in vivosystem for development of new diagnostic markers and targetedtherapeutics. Genes that were upregulated in the kidneys of thismouse model were identified. We determined that GPNMB (gly-coprotein nonmetastatic B) is directly transcribed and upregu-lated by chimeric TFE3 and performed GPNMB IHC staining inhuman TFE3-RCCs to investigate its potential in the diagnosis ofthis form of RCC.

    Materials and MethodsGeneration of TFE3-RCC mouse model

    The cDNA of the human PRCC-TFE3 chimeric gene, which iscomposed of exon 1 of PRCC and exons 4–10 of TFE3, wasgenerated by overlap extension PCR, subcloned into an entryvector of the Gateway Protein Expression System (Invitrogen),and sequence verified. PRCC-TFE3 cDNA was cloned into a

    targeting vector, pRosa26-DEST (Addgene plasmid # 21189;ref. 28), which has a LoxP-Stop-LoxP (LSL) cassette precedingthe gene of interest, using the Gateway Protein Expression Systemaccording to the manufacturer's protocol. The targeting vector(pRosa26-DEST- PRCC-TFE3) was electroporated into mouseembryonic stem (ES) cells and selected for G418 resistance asdescribed previously. ES cells (LSL-PRCC-TFE3), in which theRosa26 locus was correctly targeted, were identified by Southernblot analysis and injected into blastocysts to produce chimeras.Backcrossing to C57BL/6 mice produced heterozygous F1 off-spring with germline transmission of the Rosa26-LSL-PRCC-TFE3knockin (KI) allele.Cadherin 16 (KSP)-Cre transgenicmice, whichexpress Cre recombinase under the cadherin 16 promoter specif-ically in renal epithelial cells (29), were crossed with Rosa26-LSL-PRCC-TFE3 KI (LSL-PRCC-TFE3) mice to generate PRCC-TFE3;KSP-Creþ mice. Mice were housed in Frederick National Labora-tory for Cancer Research animal facilities and euthanized by CO2asphyxiation for analyses according to the NCI-Frederick AnimalCare and Use Committee guidelines. Animal care proceduresfollowed the NCI-Frederick Animal Care and Use Committeeguidelines.

    Drug treatmentAll animal studies were conducted in accordance with the

    institutional guidelines for animal care and experimental neo-plasia and according to a protocol approved by the Animal Careand Use Committee of the Frederick National Laboratory forCancer Research. PRCC-TFE3;KSPCreþmice, 6–12months of age,were randomly assigned to vandetanib-treated group (n¼ 4) andvehicle-treated group (n ¼ 4). Vandetanib-treated animalsreceived vandetanib at 100 mg/kg by oral gavage 5 days/weekfor 16 weeks. Control animals received vehicle (PBS) by oralgavage at the same volume and frequency. MRI scans wereobtained at 4-week intervals and maximum tumor dimensionswere measured from the scans using Image J software and plottedto calculate tumor growth rates.

    MRIMRI was implemented for noninvasive detection of kidney

    lesions as described previously (30, 31), monitoring theirprogression and following the therapeutic response. Theimages were acquired on a 3.0T Clinical Scanner (Philips InteraAchieva) using a 40-mm diameter mouse-dedicated solenoidreceiver coil (Philips Research). A T2 weighted (T2w) TurboSpin Echo (TSE) sequence was acquired with an in-planeresolution of 0.180 � 0.180 mm and 0.5-mm slice thickness.A contrast medium gadolinium chelate, Dotarem, (Guerbet)was administrated intravenously at 0.1 mmol/kg and a post-contrast T1weighted (T1w) image was obtained using a gradi-ent echo scan with the same geometry as the T2w sequence. Afat saturation technique (Spectral Presaturation with InversionRecovery, SPIR) was implemented for both T1w and T2wsequences to suppress the fat and create a dark backgroundaround the kidneys to enhance contrast and distinguish fatfrom cystic and tumor masses in the kidneys.

    DNA microarray analysisTotal RNA was isolated from flash-frozen mouse kidneys using

    TRIzol reagent (Invitrogen). cDNA preparation and hybridizationof the probe arrays were performed according to the manufac-turer's instructions (Affymetrix). Affymetrix GeneChip Mouse

    Baba et al.

    Mol Cancer Res; 2019 Molecular Cancer ResearchOF2

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • Gene 2.0 ST Arrays were applied. Partek Genomics Suite 6.6 wasused for RMA-based data normalization and the subsequent dataanalysis, including PCA and ANOVA. Data are available at theNCBI GEO database under accession number (GSE130072).Gene set enrichment analysis (GSEA) was performed with GSEAsoftware as described previously (32, 33).

    Cell lines and biochemical analysisHEK293 cell lines that express HA-TFE3 andHA-PRCC-TFE3 in

    a doxycycline-dependent manner were established using the Flp-In T-Rex System (Invitrogen) as described previously (34) andcultured inDMEMwith 10% tetracycline-free FBS (Clontech) andselection antibiotics, 15 mg/mL blasticidin S and 150 mg/mLHygromicin B (Invitrogen). UOK109, UOK120, UOK124, andUOK146 cell lineswere derived fromprimary tumors of 4 patientswith TFE3-RCC treated in the Urologic Oncology Branch (UOB),NCI (Bethesda, MD) and carry the NONO-TFE3 or PRCC-TFE3gene fusions, as described previously (3, 8, 15). UOK111,UOK115, and UOK140 are cell lines derived from ccRCC tumorswith VHL gene mutations that were established in the UOB,NCI (35, 36). All cell lines were maintained in vitro in DMEMmedia supplemented with L-glutamine (4 mmol/L), sodiumpyruvate (110 mg/L), glucose (4.5 g/L), and 1X essential aminoacids, Penicillin–Streptomycin (100U/mL;Gibco), with 10%FBS(SigmaAldrich). Cell lineswere authenticated using short tandemrepeat DNA profiling (Genetica DNA Laboratories) and con-firmed to be Mycoplasma free. For gene expression profiling,HEK293 cell lines were cultured with or without 250 ng/mLdoxycycline. For qRT-PCR, total RNA was isolated using TRIzolreagent (Invitrogen) and was reverse transcribed to cDNA usingReverTra Ace qPCR RT Master Mix (Toyobo). qRT-PCR wasperformed with a LightCycler 96 Instrument (Roche) usingTHUNDERBIRD SYBR qPCR Mix (Toyobo) as described previ-ously (37). All reactions were performed with RPS 18 as aninternal control. Primer sequences are as follows: Gpnmb for-ward, 50- GCTACTTCAGAGCCACCATCACAA -30; Gpnmbreverse, 50- GGAGATGATCGTACAGGCTTCCA -30; Ret forward,50- CCACATGTTACCCGTGCAGTTC -30; Ret reverse, 50- CCAGG-CAGTCTGGGTCACAA -30; Nr4a1 forward, 50-AAACAAGGATT-GCCCTGTGG-30; Nr4a1 reverse, 50-CGCCCTTTTAGGCTGTC-TGT-30; Rps18 forward, 50- TTCTGGCCAACGGTCTAGACAAC-30; Rps18 reverse, 50- CCAGTGGTCTTGGTGTGCTGA -30. West-ern blotting analysis was performed as described previously (34).Primary antibodies were used as follows: GPNMB (R&D Systems)1:2,000 dilution, b-actin (Cell Signaling Technology) 1:1,000dilution.

    TCGA data analysisTheCancerGenomeAtlas (TCGA; refs. 12, 38) dataset was used

    to validate candidate transcriptional target genes of chimericTFE3, which were identified by RNAseq analysis.

    The dataset of four TFE3-RCC samples [TCGA-AK-3456-01,TCGA-BP-4756-01, TCGA-B8-5546-01 (all SFPQ-TFE3), TCGA-CJ-5681-01 (KHSRP-TFE3)] was compared with the dataset of465 ccRCC for gene expression levels of candidate genes usingcBioPortal (39) and six TFE3-RCC samples [TCGA-BQ-5882-01,TCGA-BQ-5887-01, TCGA-BQ-7050-01 (all PRCC-TFE3), TCGA-DZ-6131-01 (RBM10-TFE3), TCGA-G7-7501-01 (SFPQ-TFE3),TCGA-J7-8537-01 (DVL2-TFE3)] were compared with theremaining 283 samples with gene expression data within thepapillary RCC dataset. The papillary RCC dataset was further

    subdivided into papillary type I (161 samples) and papillary typeII RCC (79 samples).

    Luciferase reporter gene assayA 527bp fragment of the 50 region of the human GPNMB gene

    was amplified by PCR using KOD-Plus-Neo from the human BACclone (RP11-469O17; Advanced Geno Techs Co.). The PCRproduct amplified with primers containing restriction enzymesite (forward, ATGTACATGCTAGCACATAGTGAAACCTGCCTC-TACT; reverse, AACTTGATAAGCTTTGAATTCTCACGGACG-CAGG) was digested by NheI and HindIII, and ligated into apGL3-Basic Vector (Promega). Two M-box sequences in theGPNMB promoter construct were mutated using PrimeSTARMutagenesis Basal Kit (Takara) following the manufacturer'sprotocol. Primers for mutagenesis were as follows: M-box Mt1Forward: TAAGCTCGAGAGTTGTAAGAGGTTGAA, M-box Mt1Reverse: CAACTCTCGAGCTTATGACTCACTCCTT; M-box Mt2Forward: CCATCTCGAGATCCTCCCCGAGGCCCT, M-box Mt2Reverse: AGGATCTCGAGATGGTATTAAGCGGCAC. Reporterplasmids were cotransfected with phRL vector as an internalcontrol into a HEK293-Dx-PRCC-TFE3 cell line, which expressesPRCC-TFE3 in a doxycycline-dependent manner. Luciferase activ-ity was measured using Dual-Luciferase Reporter Assay Systemaccording to the manufacturer's protocols (Promega).

    Chromatin immunoprecipitation assayHEK293-Dx-HA-PRCC-TFE3 cell lines were cultured with or

    without doxycycline for 24 hours, and cross-linked with 1%formaldehyde at room temperature for 5 minutes followed byincubation with 125 mmol/L glycine. Nuclear lysates were son-icated with a Bioruptor USD-200 (Diagenode) for 10 minutestwice. To purify HA-PRCC-TFE3–bound chromatin, 50 mg ofchromatin was subjected to immunoprecipitation with Anti-HAAffinity Matrix (Roche) at 4�C overnight. Immunoprecipitateswere washed five times, eluted, and reverse–cross-linked. DNAwas purified with NucleoSpin Gel and PCR Clean-up (Takara)following the manufacturer's protocol. DNA enrichment in thechromatin immunoprecipitation (ChIP) sampleswas determinedby qRT-PCRwith THUNDERBIRD SYBR qPCRMix (Toyobo) anda LightCycler 96 Instrument (Roche) following the manufac-turer's protocol. Primers for theGPNMBpromoter region contain-ing twoM-boxmotifs are as follows: forward, GATGCCAAGAAG-GAGTGAGTCATA; reverse, ATCTGTGGTGCCTCCCTCTC. As aninternal negative control, a 142 bp region, which is 2 kbpupstream from the aforementioned GPNMB promoter regionwas selected. Primers for this negative control region are asfollows: forward, TCACTGGGACTTCAGGTACACATC; reverse,AGGGCCATTTTGGGTAAAGAA.Data are expressed as percentageof input DNA.

    Case selectionTFE3-immunoreactive RCCs were collected through consulta-

    tion systems by The Japanese Society of Pathology (http://pathology.or.jp/). Histologic features were evaluated by two patholo-gists with expertise in renal tumors (N. Kuroda and Y. Naga-shima). Clinicopathologic findings including immunostainingwere summarized in Table 1. Sporadic clear cell RCCs (n ¼ 56)and papillary RCCs (n ¼ 20), among which 20 ccRCC and 11papillary RCC were previously reported by us (40), were used forcomparison of GPNMB staining. The possibility that thesecases represented patients with von Hippel–Lindau disease,

    TFE3-RCC Mouse: A Model for Human Disease

    www.aacrjournals.org Mol Cancer Res; 2019 OF3

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://pathology.or.jp/http://pathology.or.jp/http://mcr.aacrjournals.org/

  • Birt–Hogg–Dub�e (BHD) syndrome, tuberous sclerosis complex,hereditary papillary RCC, or hereditary leiomyomatosis RCC wascarefully examined and excluded in all patients by thoroughmedical examination and family history. This studywas approvedby the Institutional Review Board of the Kumamoto University(Kumamoto, Japan; #1245), Yokohama City University(Yokohama, Japan), Saitama Medical University InternationalMedical Center (Saitama, Japan; #16-226), and Kochi Red CrossHospital (Kochi, Japan).

    FISHTo assess the status of TFE3 in the TFE3-RCCs, FISHanalysiswas

    performed by using a dual-color TFE3 break-apart probe (GSPLaboratory), which was labeled with FITC at 50 side (550 kb) andwith Texas Red at 30 side (570 kb) of TFE3. In each case, at least 60nonoverlapping nuclei in the tumor areawere counted. The signalpattern of a normal cell depends on sex, that is, one fused signal inmen, and two fused signals in women. Typical TFE3 rearrange-ment pattern of a TFE3-RCC cell is one split signal inmen, andonesplit signal and one fused signal in women. The split signal wasdefined according to the evaluationmethod described in previousstudies (25, 27). When the case exhibited a TFE3 split but did notshow a chimeric band of known partners in RT-PCR, a dual-colorSFPQ-TFE3 fusion FISHProbe (Cyto Test Inc)wasused. If a yellowsignal was observed in 30% or more of 100 counted cells, thetumor was defined as harboring SFPQ-TFE3.

    RT-PCRRNAs were extracted from the 9 TFE3-RCCs using QIAGEN

    RNeasy FFPE Kit (Qiagen) according to the manufacturer'sinstructions. Gene fusion products were amplified by RT-PCRusing the following primers: ASPSCR1 (exon 7) forward, 50-CCAAGCCAAAGAAGTCCAAG-30; SFPQ (exon 9) forward, 50-AGGTGGTGGTGGCAT AGGTT-30; PRCC (exon 3) forward,50-ATGCCTAAGCCTGGGGACGACTA-30; TFE3 reverse (exon4), 50-TGGACAGGTACTGTTTCACCTG-30; and TFE3 reverse(exon 6), 50-CCTTGACTACTGTACACATC-30.

    IHCThe resected tissues were fixed with 10% formalin and embed-

    ded in paraffin. Four-micron–thick paraffin sections were sub-jected to IHC. Sections were autoclaved at 121�C for 15 minutes.The sections were treated with the diluted antibodies at 4�Covernight. Working dilutions were 1:200 for GPNMB (goat poly-clonal antibody AF-2550 from R&D Systems) and Cathepsin K

    (Abcam), and 1:500 for TFE3 (Sigma,). The GPNMB immuno-staining was scored as (�), (1þ), and (2þ) according to themethod described previously (40).

    Statistical analysisExperimental data are summarized as themean values with SD.

    Statistical analyses were performed using a two-tailed unpaired ttest with or without Welch correction using GraphPad Prism 6.When the P value of the F test was less than 0.05,Welch correctionwas applied. Differences were considered to be statistically sig-nificant at a value of P less than 0.05. Survival data were estimatedand plotted with the Kaplan–Meier method; differences betweensurvival groups were assessed with the log-rank test. Nonlinearregression analysis for % tumor growth was performed withGraphPad Prism 6.

    ResultsPRCC-TFE3 expression in mouse kidney epithelial cellsproduces RCC

    PRCC-TFE3 knockin mice, generated by inserting humanPRCC-TFE3 cDNA preceded by a loxP-flanked neomycin cassetteinto the Rosa26 locus, were crossed with cadherin 16-Cre (KSP-Cre) transgenic mice to express PRCC-TFE3 specifically in mousekidney epithelial cells (Fig. 1A). PRCC-TFE3;KSP-Creþ mice andPRCC-TFE3;KSP-Cre� littermate control mice were evaluated fortheir phenotype. PRCC-TFE3;KSP-Creþmice showed significantlylarger and heavier kidneys at 4 and 7 months of age (Fig. 1B andC). MRI imaging revealed a disorganized kidney structure, mul-tiple cysts, and renal tumors in PRCC-TFE3;KSP-Creþ mice(Fig. 1D). Histopathologic analysis revealed a variety of prolifer-ating morphologies, dilation of tubules, cystic lesions, neoplasticregions protruding into the cystic lumen, and solid tumors ofvarious sizes (Fig. 1E). Complete necropsy with histopathologyrevealed no metastasis or primary tumors in organs other thankidney. All pathologic lesions in kidneys showed strong nuclearstaining of TFE3 (Fig. 1F). TFE3 nuclear staining was observedwithout obvious phenotype in renal papillae, pelvis, and ureter,but not in bladder, consistent with the CDH16-Cre (KSP-Cre)expression pattern (Supplementary Fig. S1A–S1F; ref. 29). Thecyst lumens were lined by monolayers of cuboidal and/or hyper-plastic cells with occasional adenomatous lesions proliferating inmultiple layers (Fig. 1E). Solid tumors displayed characteristics ofRCCwith eosinophilic cytoplasm, clear cytoplasm, and occasion-al mixture of both populations (Fig. 1G and H). The eosinophilic

    Table 1. Summary of clinicopathologic information of patients with TFE3-RCC

    Case# Sex Age

    Size(cm) Stage

    Fuhrmangrade Histologic finding FISH Fusion TFE3 GPNMB

    CathepsinK RET

    1 M 11 2.2 pT1a-Nx-Mx 3 Clear and papillary þ ASPL-TFE3 2 2 - 12 F 16 9.0 pT2a-pN1-cM1 2 Clear and tubular þ ASPL-TFE3 2 1 1 23 F 19 2.5 pT1a-Nx-Mx 2 Clear and papillary, calcification þ SFPQ-TFE3 2 2 1 24 F 20 6.0 pT1b-Nx-Mx 3 Eosinophilic, alveolar, tubular þ PRCC-TFE3 2 2 - 15 M 26 3.3 pT1a-Nx-Mx 2 Eosinophilic, alveolar, tubular þ PRCC-TFE3 2 2 2 -6 F 33 1.0 pT1a-Nx-Mx 2 Clear and alveolar þ SFPQ-TFE3 2 1 1 -7 M 52 2.7 pT1a-pN12-cM0 2 Clear and papillary þ PRCC-TFE3 2 2 - 18 F 66 5.5 pT1b-Nx-Mx 1 Clear and multicystic, calcification þ SFPQ-TFE3 1 2 2 29 F 73 2.5 pT3-Nx-Mx 3 Alveolar, papillary,mixed clear and

    eosinophilic, psammoma bodyþ ASPL-TFE3 2 2 1 2

    Baba et al.

    Mol Cancer Res; 2019 Molecular Cancer ResearchOF4

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • cells tended to be arranged in tubules, trabeculae, and lobules,and were supported by a fine fibrovascular stroma. On the otherhand, clear cells were arranged in lobules, and frequently exhib-ited an alveolar and/or multicystic pattern. Psammoma bodies,calcified lesions that are characteristic of TFE3-RCC (41), wereoccasionally observed (Fig. 1I). An increased cell proliferation ratein the kidneys of PRCC-TFE3;KSP-Creþ mice was indicated by asignificantly increased BrdU incorporation (Fig. 1J and K) as wellas higher number of Ki67þ cells (Supplementary Fig. S1G andS1H). Increased BrdU incorporation and Ki67þ cells in cyst-liningcells indicated that the cystic regions were produced by uncon-trolled hyperproliferation of tubular epithelial cells. Intriguingly,multiple stages of RCC development from cystic regions withabnormally proliferating monolayers to solid tumors wereobserved in a single kidney of PRCC-TFE3;KSP-Creþ mice. Therenal tumor growth rate was calculated from MRI measurements(Fig. 1L and M). Renal neoplastic lesions in this mouse modeldisplay heterogeneous histologic features, which are also seen inhuman TFE3-RCC. As expected, growth rates of individual tumorsmeasured by MRI imaging were variable, probably because oftheir heterogeneous features. As indicated from the histology andMRI imaging, kidney structures were disorganized because ofabnormal epithelial cell proliferation and enlarged tumors inaged mice, resulting in renal failure and early death comparedwith control PRCC-TFE3;KSP-Cre� mice. The mean survival ofPRCC-TFE3;KSP-Creþ mice was 11 months (Fig. 1N). The bloodurea nitrogen (BUN) levels were significantly higher in PRCC-TFE3;KSP-Cre(þ) mice when moribund over the age of 10months, compared with PRCC-TFE3;KSP-Cre(-) control mice(Fig. 1O). This newly established PRCC-TFE3;KSP-Creþ mousemodel will be useful for development of novel therapeutics,identification of potential diagnostic biomarkers, and as a pre-clinical model for testing new therapies for TFE3-RCC.

    Identification of GPNMB and RET as upregulated genes inPRCC-TFE3;KSP-Creþ kidneys

    To identify candidate genes for biomarkers or therapeutic targetmolecules that could be useful for the diagnosis or molecularbased therapy of TFE3-RCC, we performed microarray analysis ofkidneys from 4-month-old and 7-month-old PRCC-TFE3;KSP-Creþmice and control PRCC-TFE3;KSP-Cre�mice (Fig. 2A). Mostof 78 significantly upregulated genes in kidneys from 4-month-old PRCC-TFE3;KSP-Creþ mice were also upregulated in kidneysfrom 7-month-old PRCC-TFE3;KSP-Creþ mice (Fig. 2A; Supple-mentary Table S1A; Table S2A). Aprevious report of a TFEB-drivenRCCmouse model demonstrated activation of the Wnt-b cateninand ErbB signaling pathways (42). However, GSEA analysis didnot demonstrate either a Wnt-b catenin or ErbB activation signa-ture in PRCC-TFE3;KSP-Creþmouse kidneys, indicating that TFEBand PRCC-TFE3 have distinct oncogenic functions (Supplemen-tary Fig. S2). One of the most significantly elevated genes inkidneys from both 4 month and 7-month-old PRCC-TFE3;KSP-Creþ mice was Gpnmb that encodes a type I transmembraneglycoprotein (Fig. 2B–D; Supplementary Table S1A; TableS2A). GSEA analysis demonstrated significant enrichment ofgenes associatedwith EGFR signaling activation andRET signalingactivation among those genes upregulated in kidneys from PRCC-TFE3;KSP-Creþ mice (Fig. 2E and F). In addition, a Kras-activatedsignature was seen in PRCC-TFE3;KSP-Creþ mouse kidneys (Sup-plementary Fig. S3). Although Egfr was not significantly upregu-lated in PRCC-TFE3–expressing kidneys, expression of the Ret

    proto-oncogene, a receptor tyrosine kinase that shares down-stream signaling with EGFR, including the Kras pathway, wassignificantly higher in PRCC-TFE3;KSP-Creþ kidneys than incontrol kidneys (Fig. 2B, C, and G). IHC staining revealed robustGpnmb staining that overlapped with strong TFE3 nuclear stain-ing in kidneys from PRCC-TFE3;KSP-Creþ mice, while controlkidneys demonstrated negative staining for both TFE3 andGpnmb (Fig. 2H–M; Supplementary Fig. S4). In addition, IHCstaining demonstrated strong Ret expression in kidney tumors ofPRCC-TFE3;KSP-Creþ mice and negative staining in control kid-neys (Fig. 2N andO; Supplementary Fig. S4), whichmotivated usto test therapeutic agents that target Ret in this TFE3-RCC mousemodel. Vandetanib is a multiple tyrosine kinase inhibitor thatefficiently inhibits EGFR, VEGFR, and RET. We administeredvandetanib to PRCC-TFE3;KSP-Creþ mice. Vandetanib treatment(100 mg/kg) demonstrated a therapeutic effect on tumor growthcompared with vehicle treatment with no evidence of toxicity(Fig. 2P–U). Nonlinear regression analysis demonstrated statis-tically significantly reduced tumor growth in the vandetanib-treated group compared with the control-treated group(Fig. 2V). These data underscore the usefulness of this TFE3-RCCmouse model not only for the development of biomarkers andtargeted therapeutics but also as a preclinical model to test drugsor imaging methods. Furthermore, we have performed IHCstaining of RET on human TFE3-RCC samples and found that7 of 9 cases (77.8%) were positive for RET staining(Fig. 2W; Table 1), potentially supporting further evaluation ofvandetanib treatment in advanced human TFE3-RCC cases.

    GPNMB is a direct transcriptional target gene of chimeric TFE3To investigate themolecularmechanisms ofGpnmboverexpres-

    sion in PRCC-TFE3;KSP-Creþ kidneys, we established stable celllines derived fromHEK293 cells, which express wild-type TFE3 orPRCC-TFE3 in a doxycycline-dependent manner (Fig. 3A and B).Induced wild-type TFE3 localized predominantly in the cyto-plasm. On the other hand, PRCC-TFE3 localized predominantlyin the nucleus (Fig. 3A). These results support the concept that thePRCC-TFE3 chimeric protein works as an oncoprotein with con-stitutively active transcriptional activity. Induction of PRCC-TFE3and, to a lesser extent, wild-type TFE3, resulted in upregulatedGPNMB expression at bothmRNA and protein levels (Fig. 3C andD). To determine whether GPNMB is the direct transcriptionaltarget of chimeric TFE3, we inserted the promoter region ofhuman GPNMB, from �467 to the transcription start site (TSS),into a luciferase reporter plasmid. Two M-box motifs (M-box1and M-box2), which are putative TFE3-binding sequences, wereincluded in this construct. We transfected reporter plasmidscarrying wild-type or mutant M-box motifs into doxycycline-inducible PRCC-TFE3–expressing HEK293 cells (Fig. 3E). Asshown in Fig. 3F, the promoter activity of GPNMB was upregu-lated by the induced expression of PRCC-TFE3. Importantly, thisPRCC-TFE3–dependent GPNMB promoter activity was attenuat-ed in cells transfected with mutated M-box1 or M-box2 reporterplasmids. Furthermore, complete absence of PRCC-TFE3–dependent promoter activity was seen in cells transfected witha reporter plasmid in which both M-box1 and M-box2 weremutated (Fig. 3F). These findings indicate that GPNMB is tran-scriptionally upregulated by PRCC-TFE3 through both M-boxmotifs in its promoter. In addition, we have confirmed the directbinding of PRCC-TFE3 to the minimal GPNMB promoter regioncontaining these two M-box motifs by ChIP followed by qPCR

    TFE3-RCC Mouse: A Model for Human Disease

    www.aacrjournals.org Mol Cancer Res; 2019 OF5

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • Baba et al.

    Mol Cancer Res; 2019 Molecular Cancer ResearchOF6

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • (Fig. 3G and H). These experimental data clearly indicate thatGPNMB is a direct transcriptional target gene of PRCC-TFE3.

    Specific expression of GPNMB in TFE3-RCCBecauseGPNMBwas proven to be a direct transcriptional target

    of chimeric TFE3, we evaluated the utility of GPNMB as a diag-nostic marker for TFE3-RCC. To determine whether GPNMBexpression was specific for TFE3-RCC, we compared GPNMBexpression in RCC cell lines established from human TFE3-RCCs,and in cell lines from sporadic clear cell RCCs (ccRCC). The TFE3RCCcell linesUOK109,whichwedeveloped from tumormaterialremoved from a 39-year-old female (15), UOK120, developedfrom tumor material from a 30-year-old male (3), UOK124,developed from a 21-year-old female (3), and UOK146, devel-oped from tumor material from a 45-year-old male (3, 35),expressed significantly higher amounts of GPNMB protein, whileccRCC cell lines UOK111, UOK115, and UOK140 (15) expressedno detectable GPNMB (Fig. 4A and B). To further confirm thespecificity of GPNMB expression in TFE3-RCC, we utilized thegene expression database for ccRCC (KIRC) from TCGA proj-ect (12, 38) from which there were 4 cases of TFE3-RCC. Wecompared the GPNMB mRNA expression levels in 465 cases ofccRCC with 4 cases of TFE3-RCC. GPNMB expression levels werestatistically significantly higher (P < 0.0001) in TFE3-RCC thanccRCC (Fig. 4C). We also compared the expression levels ofCathepsin K, which has been used as a diagnostic marker forTFE3-RCC, but were unable to see statistically significant Cathep-sin K expression differences between ccRCC and TFE3-RCC(Fig. 4C), thereby underscoring the utility of GPNMB as a diag-nostic marker. Although TFE3-RCC can present with a variety ofhistologies, the most frequently observed histologic character-istics are papillary, tubular, or alveolar structures composed ofclear cells, which may lead to misdiagnosis of TFE3-RCC asccRCC or papillary RCC. Hence, we compared GPNMB mRNAexpression in TFE3-RCC and papillary RCC using the TCGAgene expression database for papillary RCC (KIRP; ref. 14)comprised of 6 cases of TFE3-RCC, 161 cases of papillary typeI RCC, and 79 cases of papillary type II RCC. As shownin Fig. 4D, while GPNMB expression levels were statistically

    significantly higher (P < 0.0001) in TFE3-RCC than in eitherpapillary type I RCC, papillary type II RCC (P < 0.0001), or totalpapillary RCC (P < 0.0001), there were no statistically signif-icant differences in Cathepsin K expression between TFE3-RCCand either papillary type I RCC, papillary type II RCC, or totalpapillary RCC. (Fig. 4D). These data show that GPNMB expres-sion is a useful biomarker for distinguishing TFE3-RCC fromother types of RCCs.

    GPNMB immunostaining as potential diagnostic marker forTFE3-RCC

    To evaluate the IHC expression pattern of GPNMB in formalin-fixed paraffin-embedded (FFPE) TFE3-RCCs, we collected 9 caseswith characteristic histologies of TFE3 translocation RCC. Theydisplayed papillary, tubular, or alveolar histology with frequentcalcification and were composed of clear cells or mixtures of clearcells and eosinophilic cells (Fig. 5A–C). All cases demonstratednuclear TFE3 immunoreactivity, and were confirmed to be TFE3-RCCby FISHandRT-PCRanalyses (Fig. 5D–F). Clinicopathologicfindings are summarized in Table 1. All cases of TFE3-RCC wereconfirmed to have nuclear TFE3 staining (Table 2). All TFE3-RCCsshowed positive immunoreactivity for GPNMB (Fig. 5G–I; Table 2), two cases with 1þ staining, and 7 cases with 2þstaining. Cathepsin-K was positively stained in only 6 of 9 TFE3-RCC cases (Fig. 5J–L; Table 2). There was no obvious correlationbetween Fuhrman grade and immunoreactivity of GPNMB orCathepsin-K (Table 1). To further confirm the specificity ofGPNMB immunostaining as a potential diagnostic marker forTFE3-RCC, we have stained 56 ccRCCs and 20 papillary RCCs forGPNMB [(40) and this report]. Comparisons between TFE3-RCCand ccRCC and between TFE3-RCC and papillary RCC werestatistically significant (P

  • Figure 2.

    Overexpression of Gpnmb and Ret in TFE3-RCC mouse model. A, Numbers of genes differentially expressed between PRCC-TFE3;KSP-Creþ kidneys and PRCC-TFE3;KSP-Cre�kidneys. Volcano plot of gene expression changes for 4-month-old (4M; B) and 7-month-old (7M; C) PRCC-TFE3;KSP-Cre� kidneys and PRCC-TFE3;KSP-Creþ kidneys. The x-axis specifies the fold changes [log2(Cre(þ)/Cre(-))] and the y-axis specifies the negative log to the base 10 of the t test q-values. Redand blue dots represent genes expressed at significantly higher or lower levels (>2-fold vs. < -2-fold) in PRCC-TFE3;KSP-Creþ (Continued on the following page.)

    Baba et al.

    Mol Cancer Res; 2019 Molecular Cancer ResearchOF8

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • DiscussionTFE3 Xp11.2 translocation RCC (TFE3-RCC) was initially

    described in 1996 (3, 43) and established as an independentsubtype of RCC in 2004 by the World Health Organization (44).Because of the relatively low incidence of TFE3-RCC (14), thebiological characteristics of TFE3-RCC have not been fully clar-ified and effective forms of therapy for patients with advanceddisease have yet to be established (16). Although many cases ofTFE3-RCCdisplay the characteristic histology, other cases of TFE3-RCC are misdiagnosed as ccRCC, papillary RCC, or unclassifiedRCC (12, 25, 26). Given the aggressive nature of this disease, it isof great importance to develop a concise, sensitive, and specificmethod for diagnosis and treatment of TFE3-RCC. Here, we havegenerated the first TFE3-RCC mouse model, which we haveutilized to further characterize the chimeric PRCC-TFE3 proteinand identify a robust and reliable diagnostic marker for TFE3-RCC, and as a model for the development of targeted therapeuticapproaches for this disease.

    Results from the evaluation of our PRCC-TFE3–expressingmouse model have confirmed that chimeric PRCC-TFE3 is anoncogene, which is responsible for RCC development in vivo.Because all the chimeric genes reported to date in TFE3-RCCencode the carboxy-terminal half of TFE3, which retains the basichelix–loop–helix leucine zipper structures through which TFE3dimerizes and binds to DNA (16, 18), it is predicted that thesechimeric genes function as oncogenic transcription factors (45,46). Indeed, overexpressed PRCC-TFE3 and SFPQ-TFE3 (data notshown) demonstrated predominant nuclear localization, whileoverexpressed wild-type TFE3 localized in the cytoplasm of HEK293 cells. This finding suggests that chimeric TFE3 proteinsacquire the ability to localize in the nucleus and function asconstitutively active transcription factors. Aberrant upregulationof PRCC-TFE3 transcriptional target genes, followed by pertur-bationof the transcriptional network ismost likely responsible forTFE3-RCC development. Further analysis of the transcriptionalnetwork alterations caused by PRCC-TFE3 expression may pro-vide clues to understanding the molecular mechanisms of TFE3-RCC development. Indeed, our TFE3-RCC mouse model dem-onstrated Ret overexpression and therapeutic responses to van-detanib treatment. We utilized publicly available databasesand searched for putative transcription factors, which may regu-late Ret gene expression (data not shown). There was no clearevidence for TFE3 mediated Ret regulation. However, among the

    many putative transcription factors, which may regulate Rettranscription, Nr4a1 expression was dramatically elevated inPRCC-TFE3–expressing kidneys (Supplementary Fig. S5). Nr4a1expression was significantly higher in TFE3-RCC than ccRCC andpapillary RCC (Supplementary Fig. S5). It will be of great impor-tance to clarify the details of the transcriptional network, includ-ing Nr4a1, perturbed by chimeric TFE3.

    Another type of translocation RCC has been described involv-ing chromosome 6p21 translocation, in which a second MITfamily transcription factor, TFEB, is fused to and transcribed bya strong promoter of theMALAT1 gene (47, 48). In chromosome6p21 translocation RCC, overexpression of wild-type TFEB drivesRCC development. Indeed, kidney-specific overexpression ofwild-type TFEB in a transgenic mouse model produced clear celland papillary RCCs (42). This mouse model displays aberrantactivation of the Wnt signaling and ErbB signaling pathways,whichwas not observed inour PRCC-TFE3mousemodel byGSEAanalysis (Supplementary Fig. S2). The difference between thesetwomodels might reflect differences between wild-type TFEB andchimeric PRCC-TFE3. As we have shown, PRCC-TFE3 localizes inthe nucleus inmost of the cells, whereas wild-type TFE3 and TFEBlocalize predominantly in the cytoplasm under normal condi-tions (37, 49–51). In addition, PRCC that displaces the N-termi-nal half of TFE3 may contribute to altered transcriptional activityaffecting different transcriptional targets comparedwithwild-typeTFE3. Indeed, when we compared significantly upregulated genesin PRCC-TFE3–expressing kidneys and TFEB-expressing kid-neys (42), less than 10% of PRCC-TFE3 upregulated genes werecommonly upregulated in TFEB-expressing kidneys (Supplemen-tary Table S3).Our unique PRCC-TFE3mousemodel will providea powerful tool for further clarification of molecular mechanismsresponsible for TFE3-RCC development.

    In this TFE3-RCC mouse model, PRCC-TFE3 expression wasinduced by cadherin 16 promoter–driven Cre recombinasewith broad expression in the distal nephron and some regionsof the proximal tubules (29, 30). Notably, PRCC-TFE3–expres-sing kidneys demonstrated different histologic features, dilatedmonolayer tubules with hyperproliferation, adenomatous epi-thelial cells growing as multiple layers into the lumen of dilatedtubules, and solid tumors with a variety of sizes. The diversity ofhistologic features resulting from PRCC-TFE3 expression inmouse kidney may represent different stages of RCC develop-ment, which have acquired further genetic and/or epigenetic

    (Continued.) kidneys (q < 0.05). Gpnmb was one of the most significant genes showing increased expression in both 4-month-old and 7-month-old PRCC-TFE3;KSP-Creþ kidneys. Ret was identified as one of the highly expressed and druggable receptor tyrosine kinases in both 4-month-old and 7-month-old PRCC-TFE3;KSP-Creþ kidneys. D, Expression of Gpnmb was quantified by qRT-PCR analysis of 8-month-old PRCC-TFE3;KSP-Cre� mouse kidneys (n ¼ 6) and PRCC-TFE3;KSP-Creþ mouse kidneys (n ¼ 7). Data are represented as a box-and-whisker plot (unpaired t test). E and F, GSEA of microarray data from 4-month old PRCC-TFE3; KSP-Creþ mouse kidneys (n ¼ 4) versus 4-month-old PRCC-TFE3; KSP-Cre� mouse kidneys (n ¼ 4; E) and 7-month-old PRCC-TFE3; KSP-Creþ mousekidneys (n ¼ 4) versus 7-month-old PRCC-TFE3; KSP-Cre� mouse kidneys (n ¼ 3; F). Significant enrichment of genes associated with EGFR activation (E) andRET activation (F) was seen in kidneys from PRCC-TFE3; KSP-Creþ mice. NES, normalized enrichment score; NOM P value, Nominal P value. G, Expression of Retwas quantified by qRT-PCR analysis of 8-month-old PRCC-TFE3;KSP-Cre� mouse kidneys (n ¼ 6) and PRCC-TFE3;KSP-Creþ mouse kidneys (n ¼ 7). Data arerepresented as a box-and-whisker plot (unpaired t test). H and I, H&E staining of 7 month-old PRCC-TFE3;KSP-Cre� (H) and PRCC-TFE3;KSP-Creþ mouse kidneys(I). (J–O) Representative immunostaining for TFE3 (J and K), Gpnmb (L and M), and Ret (N and O) on serial sections of 7-month-old PRCC-TFE3;KSP-Cre� (J, L,N) and PRCC-TFE3;KSP-Creþ (K, M, O) kidneys. Bottom panels are higher magnified images of the rectangular areas in top panels. (P–V) Treatment of PRCC-TFE3;KSP-Creþ mice with vandetanib, an inhibitor of RET. Representative coronal T2 weighted MRI images of vehicle-treated (P and Q) and vandetanib-treated (100mg/kg; R and S) PRCC-TFE3;KSP-Creþ mice chronologically taken on day 0 and day 116 of the study. Red arrows indicate the tumors that were tracked forgrowth. Representative tumor growth curves of vehicle-treated (T) and vandetanib-treated (U) PRCC-TFE3;KSP-Creþ mice shown in P and Q, and R and S,respectively. The largest dimension of each tumor was measured from sequential MRI images taken on day 0, 30, 60, 95, and 116 of treatment. V, Nonlinearregression analysis of % tumor growth in the vehicle-treated group (black dots) and vandetanib-treated group (red square; ���� , P < 0.0001) W, Representativeimmunostaining for RET on human TFE3-RCC demonstrates significant cytoplasmic staining.

    TFE3-RCC Mouse: A Model for Human Disease

    www.aacrjournals.org Mol Cancer Res; 2019 OF9

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • changes in addition to PRCC-TFE3 expression. Of note, recentadvances in next-generation sequencing technology haverevealed the heterogeneity of cancer. Clonal heterogeneity withdifferent combinations of driver gene mutations has been wellcharacterized in clear cell RCC (52, 53). We hypothesize thatTFE3-RCC may also display clonal heterogeneity with a variety

    of driver gene mutations. In fact, our TFE3-RCCmice developedvariable histologies, tumor doubling times, and responses totargeted therapeutics. This diverse phenotype may result from avariety of driver gene mutations. Identification and character-ization of additional TFE3-RCC driver gene mutations willcontribute to a better understanding of the causes of TFE3-

    WT-TFE3

    WT-TFE3WT-TFE3 WT-TFE3

    PRCC-TFE3

    PRCC-TFE3PRCC-TFE3 PRCC-TFE3

    Doxy + +

    TFE3

    HA

    Actin

    −+ +− −

    −Doxy

    GPNMB

    H3

    8

    6

    4

    2

    0

    Doxy (−)Doxy (+)

    Doxy (−)Doxy (+)

    Doxy (−)Doxy (+)

    Rel

    ativ

    e ex

    pres

    sion

    P = 0.0092

    P = 0.0084

    Relative luciferase activityn.s.

    n.s.

    Vec

    WtWt

    Mt1Mt1

    Mt2Mt2

    Mt1/2Mt1/2

    M-box1:CACATGAM-box2:TCACATGA

    –467 M-box1

    M-Box1/2

    M-Box

    M-Box

    M-box2 TSS

    Luciferase

    Luciferase

    Luciferase

    Luciferase

    0 5 10 15

    Control

    Control

    % In

    put

    0.03

    0.02

    0.01

    0.00

    n.s.

    GPNMBA

    E

    G H

    F

    B C D

    25 mm 25 mm

    25 mm25 mm

    Figure 3.

    Gpnmb is a direct transcriptional target of PRCC-TFE3. A, ICC using anti-HA antibody on HEK293-derived stable cell lines, which express HA-tagged wild-typeTFE3 and PRCC-TFE3 in a doxycycline-dependent manner. B,Western blotting with anti-TFE3, anti-HA, and anti-b actin on HEK293-derived doxycycline-inducible cell lines cultured without and with doxycycline. C, GPNMB expression was quantified by qRT-PCR on HEK293-derived doxycycline-inducible cell lines,which were cultured without (open bar) and with doxycycline (solid bar). Data represent means� SD (triplicate, unpaired t test: WT-TFE3 �� , P¼ 0.0084, PRCC-TFE3 �� , P¼ 0.0092). Representative data from at least three independent experiments are shown. D,Western blotting with anti-GPNMB and anti-histone H3 onHEK293-derived doxycycline-inducible cell lines cultured without and with doxycycline. E, Scheme of Luciferase reporter constructs with human GPNMBpromoter. Putative TFE3 consensus sequences are listed as M-box1 (CACATGA) and M-box2 (TCACATGA).Wt, wild-typeGPNMB promoter construct; Mt1:M-box1 is mutated to CTCGAGA; Mt2: M-box2 is mutated to TCTCGAGA; M-box1/2: both M-box1 and M-box2 are mutated to CTCGAGA and TCTCGAGA,respectively. F, Each Luciferase reporter construct and pGL4 as a negative control were transfected into PRCC-TFE3 doxycycline-inducible HEK 293 cell line withphRL internal control. Twelve hours after transfection, mediumwas changed to newmediumwith or without doxycycline, followed by additional 24-hourincubation and harvest. The x-axis displays relative Luciferase activity. GPNMB promoter activity is upregulated by PRCC-TFE3 induction in an M-Box–dependentmanner. Data represent means� SD (triplicate, unpaired t test: n.s., not significant; �� , P

  • RCC heterogeneity and facilitate the development of effectivetargeted therapeutics. This PRCC-TFE3 mouse model can beutilized in future studies to evaluate potential driver genemutations by crossing with genetically engineered mice forselective gene deletion.

    Because the chimeric TFE3 proteins responsible for TFE3-RCCdevelopment act as oncogenic transcription factors, transcription-ally upregulated direct targets of chimeric TFE3 could be prom-ising candidates for TFE3-RCC diagnostic markers. Gpnmb wasone of the most significantly upregulated genes in PRCC-TFE3–expressing kidneys, and, with luciferase reporter assays and ChIP-qPCR, we have shown that GPNMB is a direct transcriptionaltarget of PRCC-TFE3. GPNMB expression was significantly higherin human TFE3-RCCs than in clear cell or papillary RCCs, therebydemonstrating high sensitivity and specificity as a biomarker forTFE3-RCC.

    In pathologic diagnosis of TFE3-RCCs using FFPE tissues,strong TFE3 nuclear immunostaining is considered the diag-nostic gold standard (19), but TFE3 IHC can be problematic

    because the antigenicity of TFE3 can be easily altered byimproper fixation and sample storage conditions. Therefore,pathologists often face difficulties to evaluate TFE3 staining (24,26). The break-apart FISH assay is recommended to augmentthe histopathologic diagnosis of TFE3-RCC (25, 27). However,the FISH result may be judged as negative in cases in which thepartner localizes to the vicinity of TFE3 on the short arm ofchromosome X (54). Hence, a surrogate marker that can beused for routine pathologic diagnosis in FFPE tissues is desired.We found that GPNMB immunostaining using FFPE tissues waspositive in all 9 human TFE3-RCCs in this study. Furthermore,our study showed that 55 of 56 (98.2%) sporadic ccRCCs and17 of 20 (85.0%) sporadic papillary RCCs were negativelystained for GPNMB (Table 3). Although sporadic chromo-phobe RCCs and hybrid oncocytic tumors associated with BHDsyndrome also stain for GPNMB (40), these tumors are histo-logically distinctive and rarely need to be distinguished fromTFE3-RCCs. Pathologists may find GPNMB immunostaininguseful in diagnosing TFE3-RCC when tumors have papillary

    GPNMB

    β-Actin

    GPNMB CTSK

    Xp11.2tRCC ccRCC

    UO

    K12

    0

    UO

    K14

    6

    UO

    K12

    4

    UO

    K10

    9

    UO

    K11

    1

    UO

    K11

    5

    UO

    K14

    0

    20

    15

    10

    5

    0

    20

    15

    10

    5

    0

    Rel

    ativ

    e ex

    pres

    sion

    Rel

    ativ

    e ex

    pres

    sion

    GPNMB CTSK

    KIRPKIRC

    n.s.

    n.s.n.s.

    n.s.P < 0.0001

    P < 0.0001

    P < 0.0001

    P < 0.0001

    ccRCC ccRCCTFE3-RCC TFE3-RCCTo

    tal-p

    RC

    C

    Tota

    l-pR

    CC

    Type

    1-pR

    CC

    Type

    1-pR

    CC

    Type

    2-pR

    CC

    Type

    2-pR

    CC

    TFE

    3-R

    CC

    TFE

    3-R

    CC

    Cell line Age Sex Cytogenetics FusionA

    C D

    B

    Figure 4.

    GPNMB overexpression differentiates TFE3-RCC from clear cell RCC and papillary RCC. A, Detailed information of four TFE3-RCC cell lines. B,Western blottinganalysis demonstrates significantly higher GPNMB expression in TFE3-RCC cell lines compared with ccRCC cell lines. C, Comparison of GPNMB and Cathepsin K(CTSK) expression in TFE3-RCC (n¼ 4) and ccRCC (n¼ 465). The gene expression database for ccRCC (KIRC) from TCGA project, which contained 4 cases ofTFE3-RCC was utilized for analysis.D, The gene expression database for papillary RCC (KIRP) from TCGA containing 6 cases of TFE3-RCC was utilized tocompare GPNMB and Cathepsin K (CTSK) expression in TFE3-RCC and papillary RCC. Total papillary RCC (n¼ 283 without TFE3-RCC) includes 161 cases of type Ipapillary RCC and 79 cases of type II papillary RCC. Data are represented as a box-and-whisker plot. (unpaired t test with or withoutWelch correction, based on Pvalue of the F test: n.s., not significant).

    TFE3-RCC Mouse: A Model for Human Disease

    www.aacrjournals.org Mol Cancer Res; 2019 OF11

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • and/or clear cell morphologies with indefinite TFE3 staining.This histopathologic study was based on a limited number ofhuman TFE3-RCC cases. Amassing additional clinicopathologicdata will be necessary for a better understanding of the clin-icopathologic signature of TFE3-RCC, which will contribute tothe design of potential therapeutic agents for treating advancedcases of TFE3-RCC.

    Our study provides a valuable model for the development oftargeted therapies for advanced TFE3-RCC. PRCC-TFE3 mousemodel kidneys and human TFE3-RCC samples demonstrate sig-nificant elevation of RET expression. Moreover, the RET inhibitorvandetanib significantly suppressed TFE3-RCC growth in mice.The receptor tyrosine kinase inhibitor vandetanib is FDAapproved for the treatment of advanced cases of medullarythyroid carcinoma (55). Because there is no established treatmentfor advanced TFE3-RCCwith poor prognosis, vandetanib or othermultiple tyrosine kinase inhibitors that target RET could bepromising candidates as effective therapeutic agents for advancedTFE3-RCC. In addition, GPNMB itself could be a therapeutic

    target for advanced TFE3-RCC. An antibody–drug conjugate(ADC) targeting GPNMB, glembatumumab vedotin, comprisedof an anti-GPNMB antibody conjugated to a cellular toxin,monomethylauristatin E (MMAE), causes death of GPNMB-pos-itive cells. Indeed, glembatumumab vedotin is being evaluated inclinical trials for several cancers, including advanced melanomaand breast cancer (56). It would be of interest to test this ADC in apreclinical study in the PRCC-TFE3mousemodel. GPNMB is alsoknown to promote tumor growth and invasion throughintegrin signaling, VEGFR activation, or EGFR activation (45, 57).Targeting GPNMB signaling may also be a promising strategy fortreatment of advanced TFE3-RCC. Our current work has providedthe basis for the development of effective therapies againstadvanced TFE3-RCC that eventually could lead to clinicaltrials that may benefit patients with this very aggressive form ofRCC.

    Disclosure of Potential Conflicts of InterestM. Baba reports receiving commercial research grant from Ono Pharmaceu-

    tical Co. Ltd and Bristol-Myers Squibb K.K. No potential conflicts of interestwere disclosed by the other authors.

    DisclaimerThe content of this publication does not necessarily reflect the views or

    policies of the Department of Health andHuman Services, nor doesmention oftrade names, commercial products, or organizations imply endorsement by theUS Government. All the funding sources had no involvement in study design,data collection, data interpretation, report writing, or decision to submit thepaper for publication. NCI-Frederick at the Frederick National Laboratory forCancer Research is accredited by AAALAC International and follows the PublicHealth Service Policy for the Care and Use of Laboratory Animals. Animal carewas provided in accordance with the procedures outlined in the "Guide for Careand Use of Laboratory Animals (National Research Council; 1996; NationalAcademy Press; Washington D.C.).

    H&E TFE3 GPNMB Cathepsin K

    Case 1 male

    Case 4 female

    Case 8 female

    ASPL-TFE3

    PRCC-TFE3

    SFPQ-TFE3

    A

    B

    C F I L

    E H K

    D G J

    100 μm

    50 μm

    50 μm

    Figure 5.

    Immunostaining of TFE3, GPNMB, and Cathepsin K in human TFE3-RCCs. A–L, Histology, IHC (TFE3, GPNMB, and Cathepsin K), and TFE3 gene break-apart FISHon representative cases of human TFE3-RCC with different TFE3 fusion genes (Cases #1, 4, 8 in Table 1).A–C, H&E staining displays characteristic histologyresembling clear cell RCC and papillary RCCs composed of epithelioid clear cells and eosinophilic cells. Occasional psammoma bodies are seen. D–F, IHC showsnuclear staining for TFE3. Representative images of TFE3 gene break-apart FISH are inserted. All cases show TFE3 translocations represented by separation ofgreen and red probes. The male case has a split (D) and the female cases have split and fused signals (E and F). G–I, Immunostaining for GPNMB demonstratessignificant cytoplasmic staining. J–L, IHC of Cathepsin K demonstrates either negative staining (J and K) or positive staining (L).

    Table 2. Summary of IHC findings in patients with TFE3-RCC

    Negative Positive 1þ 2þTFE3 0%(0/9) 100%(9/9) 1 8GPNMB 0%(0/9) 100%(9/9) 2 7Cathepsin K 33%(3/9) 67%(6/9) 4 2

    Table 3. Summary of GPNMB staining on human RCC samples

    Negative Positive 1þ 2þTFE3-RCC 0%(0/9) 100%(9/9) 2 7Clear cell RCC 98.2%(55/56) 1.8%(1/56) 1 0Papillary RCC 85.0%(17/20) 15.0%(3/20) 3 0

    Baba et al.

    Mol Cancer Res; 2019 Molecular Cancer ResearchOF12

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • Authors' ContributionsConception and design: M. Baba, M. Furuya, H. Hasumi, Y. Huang,Y. Nagashima, M. Yao, L.S. Schmidt, W.M. LinehanDevelopment of methodology: M. Baba, M. Furuya, H. Hasumi, Y. Huang,I. Kato, J.D. Kalen, W.M. LinehanAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): M. Baba, M. Furuya, T. Motoshima, M. Lang,W. Ma, H. Hasumi, T. Kadomatsu, L. Ileva, J.D. Kalen, L.A.W. Krisna,Y. Hasumi, R. Kurahashi, M. Oyama, Y. Nagashima, K. Araki, M. Eto,M. Yao, T. Kamba, Y. Oike, L.S. Schmidt, W.M. LinehanAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):M. Baba, M. Furuya, M. Lang, S. Funasaki, W. Ma, H.-W. Sun,H.Hasumi, I. Kato, Y. Satou, A. Sugiyama, K. Nishimoto, Y. Nagashima,T. Kamba, L.S. Schmidt, W.M. LinehanWriting, review, and/or revision of the manuscript: M. Baba, M. Furuya,M. Lang, H. Hasumi, I. Kato, K. Nishimoto, Y. Nagashima, N. Kuroda,M. Yao, L.S. Schmidt, W.M. LinehanAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): M. Baba, M. Furuya, H. Hasumi, N. Morris,T. Suda, L.S. Schmidt, W.M. LinehanStudy supervision: M. Baba, M. Furuya, H. Hasumi, Y. Oike, W.M. LinehanOthers (assistance of the development of mouse model in this manuscript):H. HasumiOthers (pathology services): B.O. Karim

    AcknowledgmentsWe thankMariaMerino for informative in-depth discussions of comparative

    human/mouse normal and abnormal histopathology, Nobuko Irie for excellenttechnical support and acknowledge the support provided to us by theUniversityof Texas Southwestern O'Brien Kidney Research Core Center and Peter Igarashifor Cadherin 16 (KSP)-Cre transgenic mice. pRosa26-DEST was a gift from NickHastie and Peter Hohenstein (Addgene plasmid # 21189). M. Baba wassupported, in part, by a JSPS KAKENHI Grant-in-Aid for Scientific Research(S), (#18H05284, #26221309), Grant-in-Aid for Scientific Research (B; #15H04975, # 18H02938), Grant-in-Aid for Challenging Research (Exploratory;

    # 18K19619, #18K19553), Grant-in -Aid for Scientific Research on InnovativeAreas (#16H06276), Grant-in-Aid for Scientific Research (C; #18K09140),Novartis Research Grant, a Research grant from Ono Pharmaceutical Co. Ltdand Bristol-Myers Squibb K.K. Grant, the Joint Usage/Research Center Programof the Advanced Medical Research Center, Yokohama City University (Yoko-hama, Japan), and the program of the Joint Usage/Research Center for Devel-opmental Medicine, Institute of Molecular Embryology and Genetics, Kuma-moto University (Kumamoto, Japan). M. Furuya was supported by JSPSKAKENHI, Grant-in-Aid for Scientific Research (C; #17K08745). T. Motoshimawas supported by JSPS KAKENHI, Grant-in-Aid for Scientific Research (C;#16K11013) and AKUA (Asahi Kasei pharma Urological Academy) ResearchGrant. T. Kadomatsu was supported by JSPS KAKENHI, Grant-in-Aid forScientific Research (C; #18K07236). H. Hasumi was supported by JSPSKAKENHI, Grant-in-Aid for Scientific Research (C; #16K11020). Y. Nagashimawas supported by JSPS KAKENHI, Grant-in-Aid for Scientific Research (C;#17K11162). T. Kamba was supported by JSPS KAKENHI, Grant-in-Aid forScientific Research (C; #18K09140). T. Suda was supported by a JSPS KAKENHIGrant-in-Aid for Scientific Research (S), (#18H05284, #26221309), and theNational Medical Research Council grant of Singapore TranslationalResearch Investigator Award (NMRC/STaR/0019/2014). Y. Oike was sup-ported in part by a JSPS KAKENHI Grant-in-Aid for Challenging Research(Exploratory; # 18K19519). This research was supported, in part, by theIntramural Research Program of NIH, Frederick National Laboratory, Centerfor Cancer Research. This project has been funded, in part, with Federal fundsfrom the Frederick National Laboratory for Cancer Research, NIH, undercontract HHSN261200800001E.

    The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

    Received November 19, 2018; revised March 12, 2019; accepted April 26,2019; published first May 1, 2019.

    References1. de Jong B, Molenaar IM, Leeuw JA, Idenberg VJ, Oosterhuis JW. Cytoge-

    netics of a renal adenocarcinoma in a 2-year-old child. Cancer GenetCytogenet 1986;21:165–9.

    2. Shipley JM, Birdsall S, Clark J, Crew J, Gill S, Linehan WM, et al. Mappingthe X chromosome breakpoint in two papillary renal cell carcinoma celllines with a t(X;1)(p11.2;q21.2) and the first report of a female case.Cytogenet Cell Genet 1995;71:280–4.

    3. Sidhar SK, Clark J, Gill S, Hamoudi R, Crew J, Gwilliam R, et al. The t(X;1)(p11.2;q21.2) translocation in papillary renal cell carcinoma fuses a novelgene PRCC to the TFE3 transcription factor gene. HumMol Genet 1996;5:1333–8.

    4. Ross H,Argani P. Xp11 translocation renal cell carcinoma. Pathology 2010;42:369–73.

    5. WetermanMA,WilbrinkM,Dijkhuizen T, vandenBerg E,Geurts vanKesselA. Fine mapping of the 1q21 breakpoint of the papillary renal cellcarcinoma-associated (X;1) translocation. Hum Genet 1996;98:16–21.

    6. Ladanyi M, Lui MY, Antonescu CR, Krause-Boehm A, Meindl A, Argani P,et al. The der(17)t(X;17)(p11;q25) of human alveolar soft part sarcomafuses the TFE3 transcription factor gene to ASPL, a novel gene at 17q25.Oncogene 2001;20:48–57.

    7. Argani P, AntonescuCR, Couturier J, Fournet JC, Sciot R,Debiec-RychterM,et al. PRCC-TFE3 renal carcinomas: morphologic, immunohistochemical,ultrastructural, and molecular analysis of an entity associated with the t(X;1)(p11.2;q21). Am J Surg Pathol 2002;26:1553–66.

    8. WetermanMA,WilbrinkM,Geurts vanKessel A. Fusion of the transcriptionfactor TFE3 gene to a novel gene, PRCC, in t(X;1)(p11;q21)-positivepapillary renal cell carcinomas. Proc Natl Acad Sci U S A 1996;93:15294–8.

    9. Argani P, Antonescu CR, Illei PB, Lui MY, Timmons CF, Newbury R, et al.Primary renal neoplasms with the ASPL-TFE3 gene fusion of alveolar softpart sarcoma. Am J Pathol 2001;159:179–92.

    10. Huang W, Goldfischer M, Babayeva S, Mao Y, Volyanskyy K, Dimitrova N,et al. Identification of a novel PARP14-TFE3 gene fusion from 10-year-oldFFPE tissue by RNA-seq. Genes Chromosomes Cancer 2015;54:500–5.

    11. Argani P, Lui MY, Couturier J, Bouvier R, Fournet JC, Ladanyi M. A novelCLTC-TFE3 gene fusion in pediatric renal adenocarcinoma with t(X;17)(p11.2;q23). Oncogene 2003;22:5374–8.

    12. CreightonCJ,MorganM,GunaratnePH,WheelerDA,GibbsRA, RobertsonA, et al. Comprehensive molecular characterization of clear cell renal cellcarcinoma. Nature 2013;499:43–9.

    13. Malouf GG, Su X, Yao H, Gao J, Xiong L, He Q, et al. Next-generationsequencing of translocation renal cell carcinoma reveals novel RNA splic-ing partners and frequent mutations of chromatin-remodeling genes.Clin Cancer Res 2014;20:4129–40.

    14. Cancer Genome Atlas Research Network, Linehan WM, Spellman PT,Ricketts CJ, Creighton CJ, Fei SS, et al. Comprehensive molecular charac-terization of papillary renal-cell carcinoma. N Engl J Med 2016;374:135–45.

    15. Clark J, Lu YJ, Sidhar SK, Parker C,Gill S, SmedleyD, et al. Fusion of splicingfactor genes PSF and NonO (p54nrb) to the TFE3 gene in papillary renalcell carcinoma. Oncogene 1997;15:2233–9.

    16. Kauffman EC, Ricketts CJ, Rais-Bahrami S, Yang Y, Merino MJ, Bottaro DP,et al. Molecular genetics and cellular features of TFE3 and TFEB fusionkidney cancers. Nat Rev Urol 2014;11:465–75.

    17. Argani P, Olgac S, Tickoo SK, GoldfischerM,MochH, ChanDY, et al. Xp11translocation renal cell carcinoma in adults: expanded clinical, pathologic,and genetic spectrum. Am J Surg Pathol 2007;31:1149–60.

    18. Hemesath TJ, Steingrimsson E, McGill G, Hansen MJ, Vaught J,Hodgkinson CA, et al. Microphthalmia, a critical factor in melanocytedevelopment, defines a discrete transcription factor family. Genes Dev1994;8:2770–80.

    TFE3-RCC Mouse: A Model for Human Disease

    www.aacrjournals.org Mol Cancer Res; 2019 OF13

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • 19. Argani P, Lal P, Hutchinson B, Lui MY, Reuter VE, Ladanyi M. Aberrantnuclear immunoreactivity for TFE3 in neoplasmswith TFE3 gene fusions: asensitive and specific immunohistochemical assay. Am J Surg Pathol 2003;27:750–61.

    20. Kuroda N, Mikami S, Pan CC, Cohen RJ, Hes O, Michal M, et al. Review ofrenal carcinoma associated with Xp11.2 translocations/TFE3 gene fusionswith focus onpathobiological aspect.HistolHistopathol 2012;27:133–40.

    21. Komai Y, Fujiwara M, Fujii Y, Mukai H, Yonese J, Kawakami S, et al. AdultXp11 translocation renal cell carcinoma diagnosed by cytogenetics andimmunohistochemistry. Clin Cancer Res 2009;15:1170–6.

    22. Zhong M, De Angelo P, Osborne L, Paniz-Mondolfi AE, Geller M, Yang Y,et al. Translocation renal cell carcinomas in adults: a single-institutionexperience. Am J Surg Pathol 2012;36:654–62.

    23. CajaibaMM,Dyer LM,Geller JI, Jennings LJ, GeorgeD, KirschmannD, et al.The classification of pediatric and young adult renal cell carcinomasregistered on the children's oncology group (COG) protocol AREN03B2after focused genetic testing. Cancer 2018;124:3381–9.

    24. Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, et al. TheInternational Society of Urological Pathology (ISUP) Vancouver Classifi-cation of Renal Neoplasia. Am J Surg Pathol 2013;37:1469–89.

    25. Green WM, Yonescu R, Morsberger L, Morris K, Netto GJ, Epstein JI, et al.Utilization of a TFE3 break-apart FISH assay in a renal tumor consultationservice. Am J Surg Pathol 2013;37:1150–63.

    26. Kuroda N, Tanaka A, Ohe C, Nagashima Y. Recent advances of immu-nohistochemistry for diagnosis of renal tumors. Pathol Int 2013;63:381–90.

    27. Rao Q, Williamson SR, Zhang S, Eble JN, Grignon DJ, Wang M, et al.TFE3 break-apart FISH has a higher sensitivity for Xp11.2 translocation-associated renal cell carcinoma compared with TFE3 or cathepsin Kimmunohistochemical staining alone: expanding the morphologicspectrum. Am J Surg Pathol 2013;37:804–15.

    28. Hohenstein P, Slight J, Ozdemir DD, Burn SF, Berry R, Hastie ND. High-efficiency Rosa26 knock-in vector construction for Cre-regulated over-expression and RNAi. Pathogenetics 2008;1:3.

    29. Shao X, Somlo S, Igarashi P. Epithelial-specific Cre/lox recombination inthe developing kidney and genitourinary tract. J Am Soc Nephrol 2002;13:1837–46.

    30. Baba M, Furihata M, Hong SB, Tessarollo L, Haines DC, Southon E, et al.Kidney-targeted Birt-Hogg-Dube gene inactivation in a mouse model:Erk1/2 and Akt-mTOR activation, cell hyperproliferation, and polycystickidneys. J Natl Cancer Inst 2008;100:140–54.

    31. Hasumi H, Baba M, Hasumi Y, Lang M, Huang Y, Oh HF, et al. Folliculin-interacting proteins Fnip1 and Fnip2 play critical roles in kidney tumorsuppression in cooperation with Flcn. Proc Natl Acad Sci U S A 2015;112:E1624–31.

    32. SubramanianA, TamayoP,Mootha VK,Mukherjee S, Ebert BL,GilletteMA,et al. Gene set enrichment analysis: a knowledge-based approach forinterpreting genome-wide expression profiles. Proc Natl Acad Sci U S A2005;102:15545–50.

    33. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J,et al. PGC-1alpha-responsive genes involved in oxidative phosphorylationare coordinately downregulated in human diabetes. Nat Genet 2003;34:267–73.

    34. BabaM,Hong SB, SharmaN,WarrenMB, NickersonML, Iwamatsu A, et al.Folliculin encoded by the BHD gene interacts with a binding protein,FNIP1, and AMPK, and is involved in AMPK and mTOR signaling.Proc Natl Acad Sci U S A 2006;103:15552–7.

    35. Anglard P, Trahan E, Liu S, Latif F, Merino MJ, Lerman M, et al. Molecularand cellular characterization of human renal cell carcinoma cell lines.Cancer Res 1992;52:348–56.

    36. Sourbier C, Srivastava G, Ghosh MC, Ghosh S, Yang Y, Gupta G, et al.Targeting HIF2alpha translation with Tempol in VHL-deficient clear cellrenal cell carcinoma. Oncotarget 2012;3:11.

    37. Baba M, Endoh M, Ma W, Toyama H, Hirayama A, Nishikawa K, et al.Folliculin regulates osteoclastogenesis through metabolic regulation.J Bone Miner Res 2018;33:1785–98.

    38. Ricketts CJ, De Cubas AA, Fan H, Smith CC, Lang M, Reznik E, et al. TheCancer Genome Atlas comprehensive molecular characterization of renalcell carcinoma. Cell Rep 2018;23:313–26.

    39. Cerami E,Gao J,DogrusozU,Gross BE, Sumer SO, Aksoy BA, et al. The cBiocancer genomics portal: an open platform for exploring multidimensionalcancer genomics data. Cancer Discov 2012;2:401–4.

    40. FuruyaM, Hong SB, Tanaka R, Kuroda N, Nagashima Y, Nagahama K, et al.Distinctive expression patterns of glycoprotein non-metastatic B andfolliculin in renal tumors in patients with Birt-Hogg-Dube syndrome.Cancer Sci 2015;106:315–23.

    41. Kuroda N, Katto K, Tanaka Y, Yamaguchi T, Inoue K, Ohara M, et al.Diagnostic pitfall on the histological spectrum of adult-onset renal carci-noma associated with Xp11.2 translocations/TFE3 gene fusions. Med MolMorphol 2010;43:86–90.

    42. Calcagni A, Kors L, Verschuren E, De Cegli R, Zampelli N, Nusco E, et al.Modelling TFE renal cell carcinoma in mice reveals a critical role of WNTsignaling. Elife 2016;5:e17047.

    43. Weterman MA, Wilbrink M, Janssen I, Janssen HA, van den Berg E, FisherSE, et al. Molecular cloning of the papillary renal cell carcinoma-associatedtranslocation (X;1)(p11;q21) breakpoint. Cytogenet Cell Genet 1996;75:2–6.

    44. Lopez-Beltran A, Scarpelli M, Montironi R, Kirkali Z. 2004 WHO classifi-cation of the renal tumors of the adults. Eur Urol 2006;49:798–805.

    45. Tanaka M, Homme M, Yamazaki Y, Shimizu R, Takazawa Y, Nakamura T.Modeling alveolar soft part sarcoma unveils novel mechanisms of metas-tasis. Cancer Res 2017;77:897–907.

    46. Kobos R, Nagai M, Tsuda M, Merl MY, Saito T, Lae M, et al. Combiningintegrated genomics and functional genomics to dissect the biology of acancer-associated, aberrant transcription factor, the ASPSCR1-TFE3 fusiononcoprotein. J Pathol 2013;229:743–54.

    47. Davis IJ, Hsi BL, Arroyo JD, Vargas SO, Yeh YA,Motyckova G, et al. Cloningof an Alpha-TFEB fusion in renal tumors harboring the t(6;11)(p21;q13)chromosome translocation. Proc Natl Acad Sci U S A 2003;100:6051–6.

    48. Kuiper RP, SchepensM, Thijssen J, van AsseldonkM, van den Berg E, BridgeJ, et al. Upregulation of the transcription factor TFEB in t(6;11)(p21;q13)-positive renal cell carcinomas due to promoter substitution. Hum MolGenet 2003;12:1661–9.

    49. Hong SB, OhH, Valera VA, BabaM, Schmidt LS, LinehanWM. Inactivationof the FLCN tumor suppressor gene induces TFE3 transcriptional activity byincreasing its nuclear localization. PLoS One 2010;5:e15793.

    50. Sardiello M, Palmieri M, di Ronza A, Medina DL, Valenza M, GennarinoVA, et al. A gene network regulating lysosomal biogenesis and function.Science 2009;325:473–7.

    51. Roczniak-Ferguson A, Petit CS, Froehlich F, Qian S, Ky J, Angarola B,et al. The transcription factor TFEB links mTORC1 signaling totranscriptional control of lysosome homeostasis. Sci Signal 2012;5:ra42.

    52. Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, et al.Genomic architecture and evolution of clear cell renal cell carcinomasdefined by multiregion sequencing. Nat Genet 2014;46:225–33.

    53. Turajlic S, Xu H, Litchfield K, Rowan A, Horswell S, Chambers T, et al.Deterministic evolutionary trajectories influence primary tumor growth:TRACERx renal. Cell 2018;173:595–610.

    54. Argani P, Zhang L, Reuter VE, Tickoo SK, Antonescu CR. RBM10-TFE3 renalcell carcinoma a potential diagnostic pitfall due to cryptic intrachromo-somal Xp11.2 inversion resulting in false-negative TFE3 FISH. Am J SurgPathol 2017;41:655–62.

    55. Valerio L, Pieruzzi L, Giani C, Agate L, Bottici V, Lorusso L, et al.Targeted therapy in thyroid cancer: state of the art. Clin Oncol 2017;29:316–24.

    56. Rose AAN, Biondini M, Curiel R, Siegel PM. Targeting GPNMB withglembatumumab vedotin: current developments and future opportunitiesfor the treatment of cancer. Pharmacol Ther 2017;179:127–41.

    57. Taya M, Hammes SR. Glycoprotein non-metastatic melanoma protein B(GPNMB) and cancer: a novel potential therapeutic target. Steroids 2018;133:102–7.

    Mol Cancer Res; 2019 Molecular Cancer ResearchOF14

    Baba et al.

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/

  • Published OnlineFirst May 1, 2019.Mol Cancer Res Masaya Baba, Mitsuko Furuya, Takanobu Motoshima, et al. GPNMB as a Diagnostic Marker for Human DiseaseModel Reveals Novel Therapeutic Targets and Identifies TFE3 Xp11.2 Translocation Renal Cell Carcinoma Mouse

    Updated version

    10.1158/1541-7786.MCR-18-1235doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mcr.aacrjournals.org/content/suppl/2019/05/01/1541-7786.MCR-18-1235.DC1

    Access the most recent supplemental material at:

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

    Subscriptions

    Reprints and

    [email protected] at

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

    Permissions

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

    .http://mcr.aacrjournals.org/content/early/2019/05/22/1541-7786.MCR-18-1235To request permission to re-use all or part of this article, use this link

    on June 24, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst May 1, 2019; DOI: 10.1158/1541-7786.MCR-18-1235

    http://mcr.aacrjournals.org/lookup/doi/10.1158/1541-7786.MCR-18-1235http://mcr.aacrjournals.org/content/suppl/2019/05/01/1541-7786.MCR-18-1235.DC1http://mcr.aacrjournals.org/cgi/alertsmailto:[email protected]://mcr.aacrjournals.org/content/early/2019/05/22/1541-7786.MCR-18-1235http://mcr.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice