smyd5 controls heterochromatin and chromosome integrity ... · erties of the h3k9 methyltransferase...

18
Integrated Systems and Technologies SMYD5 Controls Heterochromatin and Chromosome Integrity during Embryonic Stem Cell Differentiation Benjamin L. Kidder 1,2 , Runsheng He 1,2 , Darawalee Wangsa 3 , Hesed M. Padilla-Nash 3 , M. Margarida Bernardo 2,4 , Shijie Sheng 2,4 , Thomas Ried 3 , and Keji Zhao 5 Abstract Epigenetic regulation of chromatin states is thought to control gene expression programs during lineage specication. However, the roles of repressive histone modications, such as trimethylated histone lysine 20 (H4K20me3), in develop- ment and genome stability are largely unknown. Here, we show that depletion of SET and MYND domaincontaining protein 5 (SMYD5), which mediates H4K20me3, leads to genome-wide decreases in H4K20me3 and H3K9me3 levels and derepression of endogenous LTR- and LINE-repetitive DNA elements during differentiation of mouse embryonic stem cells. SMYD5 depletion resulted in chromosomal aberra- tions and the formation of transformed cells that exhibited decreased H4K20me3 and H3K9me3 levels and an expression signature consistent with multiple human cancers. Moreover, dysregulated gene expression in SMYD5 cancer cells was asso- ciated with LTR and endogenous retrovirus elements and decreased H4K20me3. In addition, depletion of SMYD5 in human colon and lung cancer cells results in increased tumor growth and upregulation of genes overexpressed in colon and lung cancers, respectively. These ndings implicate an impor- tant role for SMYD5 in maintaining chromosome integrity by regulating heterochromatin and repressing endogenous repet- itive DNA elements during differentiation. Cancer Res; 77(23); 672945. Ó2017 AACR. Introduction Mammalian DNA is packaged into two classes of chromatin: euchromatin, which is open and transcriptionally active, and heterochromatin, a densely packed chromatin structure that is largely refractory to transcription factor binding and transcription (1). Heterochromatin is located at specic chromosomal features such as centromeres and telomeres and interspersed throughout the genome (2). Heterochromatin is an integral player in the regulation of gene expression (3), and it promotes genome integrity by stabilizing DNA repeats by inhibiting recombination between homologous DNA repeats (4). Chromatin compaction is regulated in part by histone mod- ications, such as H4K20 and H3K9 methylation, which are enriched at heterochromatin regions. H4K20 methylation is involved in several cellular functions such as heterochromatin formation and chromosome condensation (5), transcriptional activation and repression (6), genome stability (7), DNA repli- cation (8), and DNA repair (9). The sequential methylation of H4K20me1 and H4K20me2, by Suv420h1 or Suv420h2, which catalyze H4K20me3, supports the formation of pericentric heter- eochromatin (7). H4K20me3 marks also repress transcription of repetitive elements (10). SMYD5 (SET and MYND domain-containing protein 5) med- iates H4K20me3 marks at LINE/LTRrepetitive DNA sequences (11). SMYD5 promotes mouse embryonic stem (ES) cell self- renewal by silencing lineage-specic gene expression: SMYD5 is recruited by LINE- and LTR-repetitive DNA elements to the vicinity of differentiation genes and keeps them silenced by depositing H4K20me3 marks. SMYD5 regulates heterochromatin formation and silencing of endogenous retroviruses (ERV) in ES cells by mediating H4K20me3 marks and interacting with chro- matin repressors heterochromatin protein 1 (HP1) and the H3K9 methyltransferase G9a, respectively (11). The ERV-silencing prop- erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes in hetero- chromatin formation and silencing of repetitive DNA elements. However, it is unclear how SMYD5 regulates heterochromatin formation, and how it contributes to silencing of repetitive DNA elements and genome stability during differentiation. Here, we demonstrate that depletion of SMYD5 results in decreased H4K20me3 levels and decreased H3K9me3 chromatin immunoprecipitation sequencing (ChIP-Seq) levels during ES cell differentiation. Depletion of SMYD5 causes chromosome aberrations and is accompanied by cell transformation during ES cell differentiation. Finally, resulting cancer cells display 1 Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan. 2 Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan. 3 Cancer Genomics Section, National Cancer Institute, NIH, Bethesda, Maryland. 4 Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan. 5 Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Authors: Benjamin L. Kidder, Wayne State University School of Medicine, 4100 John R St, HWCRC Building, Room 624, Detroit, MI 48201. Phone: 313-576-8302; Fax: 313-576-8306; E-mail: [email protected]; and Keji Zhao, NIH, 9000 Rockville Pike, Bethesda, MD 20892. Phone: 301-496- 2098; Fax: 301-402-0971; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-17-0828 Ó2017 American Association for Cancer Research. Cancer Research www.aacrjournals.org 6729 on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Upload: others

Post on 25-Aug-2020

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Integrated Systems and Technologies

SMYD5 Controls Heterochromatin andChromosome Integrity during EmbryonicStem Cell DifferentiationBenjamin L. Kidder1,2, Runsheng He1,2, Darawalee Wangsa3, Hesed M. Padilla-Nash3,M. Margarida Bernardo2,4, Shijie Sheng2,4, Thomas Ried3, and Keji Zhao5

Abstract

Epigenetic regulation of chromatin states is thought tocontrol gene expression programs during lineage specification.However, the roles of repressive histone modifications, suchas trimethylated histone lysine 20 (H4K20me3), in develop-ment and genome stability are largely unknown. Here, weshow that depletion of SET and MYND domain–containingprotein 5 (SMYD5), which mediates H4K20me3, leads togenome-wide decreases in H4K20me3 and H3K9me3 levelsand derepression of endogenous LTR- and LINE-repetitiveDNA elements during differentiation of mouse embryonicstem cells. SMYD5 depletion resulted in chromosomal aberra-tions and the formation of transformed cells that exhibited

decreased H4K20me3 and H3K9me3 levels and an expressionsignature consistent with multiple human cancers. Moreover,dysregulated gene expression in SMYD5 cancer cells was asso-ciated with LTR and endogenous retrovirus elements anddecreased H4K20me3. In addition, depletion of SMYD5 inhuman colon and lung cancer cells results in increased tumorgrowth and upregulation of genes overexpressed in colon andlung cancers, respectively. These findings implicate an impor-tant role for SMYD5 in maintaining chromosome integrity byregulating heterochromatin and repressing endogenous repet-itive DNA elements during differentiation. Cancer Res; 77(23);6729–45. �2017 AACR.

IntroductionMammalian DNA is packaged into two classes of chromatin:

euchromatin, which is open and transcriptionally active, andheterochromatin, a densely packed chromatin structure that islargely refractory to transcription factor binding and transcription(1). Heterochromatin is located at specific chromosomal featuressuch as centromeres and telomeres and interspersed throughoutthe genome (2). Heterochromatin is an integral player in theregulation of gene expression (3), and it promotes genomeintegrity by stabilizing DNA repeats by inhibiting recombinationbetween homologous DNA repeats (4).

Chromatin compaction is regulated in part by histone mod-ifications, such as H4K20 and H3K9 methylation, which areenriched at heterochromatin regions. H4K20 methylation is

involved in several cellular functions such as heterochromatinformation and chromosome condensation (5), transcriptionalactivation and repression (6), genome stability (7), DNA repli-cation (8), and DNA repair (9). The sequential methylation ofH4K20me1 and H4K20me2, by Suv420h1 or Suv420h2, whichcatalyze H4K20me3, supports the formation of pericentric heter-eochromatin (7). H4K20me3 marks also repress transcription ofrepetitive elements (10).

SMYD5 (SET and MYND domain-containing protein 5) med-iates H4K20me3 marks at LINE/LTR–repetitive DNA sequences(11). SMYD5 promotes mouse embryonic stem (ES) cell self-renewal by silencing lineage-specific gene expression: SMYD5 isrecruited by LINE- and LTR-repetitive DNA elements to thevicinity of differentiation genes and keeps them silenced bydepositing H4K20me3marks. SMYD5 regulates heterochromatinformation and silencing of endogenous retroviruses (ERV) in EScells by mediating H4K20me3 marks and interacting with chro-matin repressors heterochromatin protein 1 (HP1) and the H3K9methyltransferase G9a, respectively (11). The ERV-silencing prop-erties of the H3K9 methyltransferase ESET during differentiation(12) implicates a role for histone-modifying enzymes in hetero-chromatin formation and silencing of repetitive DNA elements.However, it is unclear how SMYD5 regulates heterochromatinformation, and how it contributes to silencing of repetitive DNAelements and genome stability during differentiation.

Here, we demonstrate that depletion of SMYD5 results indecreased H4K20me3 levels and decreased H3K9me3 chromatinimmunoprecipitation sequencing (ChIP-Seq) levels during EScell differentiation. Depletion of SMYD5 causes chromosomeaberrations and is accompanied by cell transformation duringES cell differentiation. Finally, resulting cancer cells display

1Department of Oncology, Wayne State University School of Medicine, Detroit,Michigan. 2Barbara Ann Karmanos Cancer Institute, Wayne State UniversitySchool of Medicine, Detroit, Michigan. 3Cancer Genomics Section, NationalCancer Institute, NIH, Bethesda, Maryland. 4Department of Pathology, WayneState University School of Medicine, Detroit, Michigan. 5Systems Biology Center,National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland.

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

Corresponding Authors: Benjamin L. Kidder, Wayne State University School ofMedicine, 4100 JohnRSt, HWCRCBuilding, Room624, Detroit, MI 48201. Phone:313-576-8302; Fax: 313-576-8306; E-mail: [email protected]; andKeji Zhao, NIH, 9000 Rockville Pike, Bethesda, MD 20892. Phone: 301-496-2098; Fax: 301-402-0971; E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-17-0828

�2017 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 6729

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 2: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

decreased H4K20me3 and H3K9me3 levels and the expressionsignature of SMYD5-depleted transformed cells is correlated witha number of human cancers and can predict patient survivaloutcome.

Materials and MethodsMouse ES cell culture

Mouse R1 ES cells were obtained fromATCC (2010) and storedin liquid nitrogen. R1 ES cells were cultured as previouslydescribed with minor modifications and used in this study frompassage 30 to passage 42 (13). Briefly, R1 ES cells were cultured onirradiated mouse embryonic fibroblasts (MEF) in DMEM, 15%FBSmedia containing leukemia inhibitory factor (LIF; ESGRO) at37�C with 5% CO2. For ChIP experiments, ES cells were culturedon gelatin-coated dishes in ES cell media containing 1.5 mmol/LCHIR9901 (GSK3 inhibitor) for several passages to remove feedercells. R1 ES cells have been tested for mycoplasma using Myco-Fluor Mycoplasma Detection Kit (Thermo Fisher Scientific). Forembryoid body (EB) formation, ES cells were cultured in lowattachment binding dishes to promote 3D formation in ES cellmedia without LIF. shSmyd5 cancer cells were cultured in DMEMcontaining 15% FBS, glutamine, 2-mercaptoethanol, and nones-sential amino acids.

Human ES cell cultureHuman ES cells (H1) were obtained from Dr. Guokai Chen

(NIH, Bethesda, MD) and stored in liquid nitrogen. H1 hESCswere cultured as previously described with minor modifications(14), and used in this study from passage 30 to 40. Briefly, H1hESCs were cultured on Matrigel-coated dishes in serum-freedefined E8media and passaged using EDTA (14). H1 hESCs weretested for mycoplasma using MycoFluor Mycoplasma DetectionKit (Thermo Fisher Scientific). For EB formation, hESCs werecultured on low attachment binding dishes to promote 3Dformation in E8 media for 2–3 days, and subsequently culturedin media containing high-glucose DMEM supplemented with15% FBS, L-glutamine, nonessential amino acids, and b-mercap-toethanol. Human shSmyd5 cancer cells were cultured in DMEMcontaining 15% FBS, glutamine, b-mercaptoethanol, and nones-sential amino acids.

Human cancer and nontumorigenic cell line cultureHCT-116, A549, and MCF7 cells were obtained from the NCI

and stored in liquid nitrogen. HCT-116, A549, and MCF7 cellswere cultured in media containing RPMI1640, 5% FBS, and 1mmol/L L-glutamine. A549 cells were used in this study frompassage 4–15, and HCT-116 and MCF7 cells were passaged nomore than 10–15 times. MCF10A cells, which were passaged nomore than 10–15 times, were cultured in media containingDMEM/F12, 5% FBS, 20 ng/mL EGF, 1 mg/mL hydrocortisone,200 ng/mL cholera toxin, and 10 mg/mL insulin.

Lentiviral transductionMouse R1 ES cells were transduced with lentiviral particles

encoding shRNAs as described previously (11). Briefly, shRNAswere cloned into the pSIH1-H1-puro Vector (System Biosciences)according to the manufacturer's protocol. To generate lentiviralparticles, HEK 293T cells were cotransfected with an envelopeplasmid (pLP/VSVG), packaging vector (psPAX2), and shRNAexpression vector using Lipofectamine 2000. Twenty-four to 48

hours posttransfection, themediumcontaining lentiviral particleswas harvested and used to infect mouse ES cells. Twenty-fourhours posttransduction, ES cells were stably selected in the pres-ence of 1 mg/mL puromycin to generate a heteroclonal popula-tion. For human ES cells (H1), the medium containing lentiviralparticles was harvested and used to transduce hESCs for 4–6hours. The E8 media were subsequently changed and posttrans-duction hESCs were stably selected and maintained in the pres-ence of 0.5 mg/mL puromycin to generate a heteroclonal popu-lation. For HCT-116, A549, and MCF7 cells, the medium(RPMI1640, glutamine, and 5% FBS) containing lentiviral parti-cles was used to transduce human cancer cells overnight. Themedia were subsequently changed and posttransduction HCT-116, A549, andMCF7 cells were stably selected in the presence of1 mg/mL puromycin to generate a heteroclonal population. ForMCF10A cells, themedium (DMEM/F12, 5% FBS, 20 ng/mL EGF,1 mg/mL hydrocortisone, 200 ng/mL cholera toxin, and 10 mg/mLinsulin) containing lentiviral particles was used to transduceMCF10A cells overnight. The media was subsequently changedand posttransduction MCF10A cells were stably selected in thepresence of 1 mg/mL puromycin to generate a heteroclonalpopulation.

Teratoma and tumor formationMouse ES cells were cultured on gelatin-coated dishes to

remove feeder cells, dissociated into single cells, and 106 ES cellswere injected subcutaneously into SCID-beigemice. After three tofourweeks,micewere euthanized and teratomaswerewashed andfixed in 10% buffered formalin. Teratomas were then embeddedin paraffin. Thin sections were cut and stained with hematoxylinand eosin using standard techniques. shSmyd5 cancer cell tumorformation was performed as described above for ES cells. Tensections were cut from three different tumors (30 sections total).All animals were treated according to Institutional Animal Careand Use Committee guidelines approved for these studies at theNational Heart, Lung, and Blood Institute (Bethesda, MD) and atWayne State University (Detroit, MI).

Xenograft tumor model. Human colon and lung adenocarcinomacell lines HCT-116 and A549, respectively, were dissociated intosingle cells and 106 cells were injected subcutaneously into femaleSCID-beige mice, aged 6–8 weeks, in accordance with InstitutionAnimal Care and Use Committee guidelines under currentapproved protocols atWayne State University. After several weeks(4–6), when the tumors grew to approximatelym 1 cm in diam-eter, mice were euthanized and tumors were washed in PBS andweighed.

DQ-collagen IV–based proteolytic activity assayMouse and human shSmyd5 cancer cells and control shLuc

cells were serial diluted in the respective maintenance media,seeded in 350 mL/well of cold Matrigel and plated in a 24-wellplate. Matrigel polymerization occurred after 30-minute incuba-tion at 37�C; 1 mL/well of maintenance medium (DMEM con-taining 15% FBS, glutamine, 2-mercaptoethanol, and nonessen-tial amino acids) was added and replenished every other day. Theproteolytic activity of the Matrigel-embedded cells was ascer-tained by first washing the cells with warm PBS and then incu-bating overnight in the presence or absence of DQ-collagen IV(10 mg/mL) in serum-free, phenol red–free DMEM at 37�C. DQ-collagen IV is a heavily fluorescein-labeled collagen IV substrate

Kidder et al.

Cancer Res; 77(23) December 1, 2017 Cancer Research6730

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 3: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

that yields green fluorescence upon cleavage by matrix metal-loproteinases (MMP). The fluorescence resulting from theDQ-collagen IV cleavage was visualized using a Leica DMIRBfluorescence microscope equipped with the SPOT RT3 cameraand the SPOT advanced software. The cell nuclei were counter-stained with Hoechst or DAPI dye.

qRT-PCR expression analysisRNA isolation and qRT-PCR were performed as previously

described with minor modifications (11). Briefly, total RNA washarvested from ES cells using an RNeasy Mini Kit or miRNeasyMini Kit (Qiagen) and DNase treated using Turbo DNA-free(Ambion). Reverse transcription was performed using a Super-script III kit (Invitrogen). Primers used for qRT-PCR (Supplemen-tary Table S1) were designed using the Universal Probe LibraryAssay design Center (Roche) or Primer 3 (http://bioinfo.ut.ee/primer3-0.4.0/).

RNA-Seq analysisRNA was harvested from shSmyd5 cancer cells as described

previously for ES cells (11). mRNA was purified using a Dyna-beads mRNA purification kit (Invitrogen). Double-strandedcDNA was generated using a SuperScript Double-StrandedcDNA Synthesis Kit (Invitrogen). cDNA was end-repaired usingthe End-It DNA End-Repair Kit (Epicentre), followed by addi-tion of a single A nucleotide, and ligation of PE adapters(Illumina) or custom-indexed adapters. PCR was performedusing Phusion High Fidelity PCR Master Mix. RNA-Seq librarieswere sequenced on Illumina GAIIX or HiSeq platforms accord-ing to the manufacturer's protocol. At least two biologicalreplicates were performed for shSmyd5 cancer cells RNA-Seqexperiments.

The "read per kilo bases of exon model per million reads"(RPKM)measure, as definedpreviously (15),was used to quantifythe mRNA expression level of a gene from RNA-Seq datasets.Differentially expressed genes were identified using edgeR (16)with a false discovery rate (FDR)<0.001 and fold-change (FC)>2.Genes with RPKM < 3 in both conditions in comparison wereexcluded from this analysis. The RPBM measure was used toquantify RNA expression levels of LINE and LTR repeats fromRNA-Seq datasets.

OncomineDifferentially expressed genes between shSmyd5 cancer cells

and shLuc EBs were evaluated using Oncomine software.shSmyd5 cancer–repressed genes are underexpressed in shSmyd5cancer cells and underexpressed in human lung adenocarcinomaversus normal lung (within the top 5% underexpressed; P ¼3.44E�10, ref. 17; P ¼ 1.12E�7, ref. 18), and underexpressed inadditional human cancers including colon adenocarcinomas andinvasive breast carcinomas versus normal colon and breast.

ChIP-SeqChIP-Seq experiments were performed as previously described

with minor modifications (11). The polyclonal H4K20me3 anti-body (07-463) was obtained from Millipore. The polyclonalH3K9me3 (ab8898) antibody was obtained from Abcam. Briefly,ES cells were harvested and chemically crosslinked with 1%formaldehyde (Sigma) for 10 minutes at 37�C and subsequentlysonicated using a Misonix XL2020 sonifier and setting #5 (18

cycles: 30-second pulse time, 1-minute rest). Sonicated cellextracts were used for ChIP assays. ChIP-enriched DNA wasend-repaired using the End-It DNA End-Repair Kit (Epicentre),followed by addition of a single A nucleotide, and ligation of anindexed adapter. PCRwas performed using PhusionHigh FidelityPCR master mix. ChIP libraries were sequenced on an IlluminaHiSeq platform according to the manufacturer's protocol. At leasttwo biological replicates were performed for the H4K20me3 andH3K9me3 ChIP-Seq experiments.

Sequence reads were mapped to the mouse genome usingbowtie2. To allow mapping to repetitive elements we used thedefault mode of bowtie2, which searches for multiple align-ments, and reports the best one based on the alignment score(MAPQ; http://bowtie-bio.sourceforge.net/bowtie2/manual.shtml).

ChIP-Seq read enriched regions were identified relative toInput DNA (sonicated chromatin) as previously described withminor modifications (11). ChIP-Seq read enriched regions(peaks) were identified relative to Input DNA using "SpatialClustering for Identification of ChIP-Enriched Regions"(SICER) software (19) with a window size setting of 200 bp,a gap setting of 400 bp and a FDR setting of 0.001. For acomparison of ChIP-enrichment between samples a fold-change threshold of 1.5 and an FDR setting of 0.001 wereused. The RPBMmeasure (reads per base per million reads) wasused to quantify the density of histone modification andSMYD5 binding at genomic regions from ChIP-Seq datasets.The Kolmogorov–Smirnov test was also performed to obtain Pvalue statistics and compare densities at genomic regions.

DNA sequencingGenomic DNA (gDNA) was harvested from shLuc ES cells and

shSmyd5 cancer cells using the Promega Wizard Genomic DNAPurification Kit. gDNA was sonicated using a Diagenode Bior-uptor and end-repaired using the End-It DNA End-Repair Kit(Epicentre) and sequenced on an Illumina HiSeq platformaccording to the manufacturer's protocol as described above.DNA sequencing (DNA-Seq) was performed using a read lengthof 75 bp. A total of 2.75� 108 and 2.53� 108 readswere obtainedfor shLuc ES cells and shSmyd5 cancer cells, respectively. At leasttwo biological replicates were performed for the shSmyd5 cancercell DNA-Seq experiments.

Input-SeqshLuc and shSmyd5 ES cells and shSmyd5 cancer cells were

harvested and chemically crosslinked with 1% formaldehyde(Sigma) for 10minutes at 37�C and subsequently sonicated usinga Misonix XL2020 sonifier and setting #5 (18 cycles: 30-secondpulse time, 1-minute rest). Sonicated cell extracts were used forInput-Seq assays. Input DNA was end-repaired using the End-ItDNA End-Repair Kit (Epicentre) and sequenced on an IlluminaHiSeq platform according to the manufacturer's protocol asdescribed above. At least two biological replicates were performedfor the shSmyd5 cancer cell Input-Seq experiments.

Copy number variation sequencingCopy number variation sequencing (CNV-Seq) software

(20) with default settings (P < 0.001) was used to identifyregions of copy number alteration. For CNV-Seq analysis ofInput-Seq data, regions that increased at least 2-fold were usedfor these analyses, while for CNV-Seq analysis of DNA-Seq

SMYD5 Regulates Genome Stability

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6731

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 4: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

data, regions that decreased at least 2-fold were used for theseanalyses.

Annotation of repetitive DNA sequencesRepetitive DNA sequence classes (e.g., LINE, LTR), families (L1,

ERVK), and names (e.g., L1Md_T, IAPLTR1) were defined accord-ing to the annotations provided by the UCSC Genome Browserand RepeatMasker (http://www.repeatmasker.org), which usescurated libraries of repeats such as Repbase (http://www.girinst.org/repbase/).

Spectral karyotypingPreparation of mouse metaphase chromosome suspension,

Spectral karyotyping (SKY) probes, slide pretreatment, slide dena-turation, detection, and imaging have been described previously(21). Numerical aberrations and structural aberrations weredescribed according to nomenclature rules from Jackson Labora-tories (http://www.informatics.jax.org/mgihome/nomen/gene.shtml). Ploidy designations for chromosome numbers used inthis study have been presented previously (22). Loss of chromo-somes (relative to cell ploidy) is classified as clonal when theidentical chromosome is lost in three ormore cells, and the gain ofchromosomes as clonal when being present in two or more cells.Structural rearrangementsmust be detected in twoormore cells tobe classified as clonal changes.

FISHWhole chromosome paints for chromosomes X, 3, 6, 14, and

19were used to further define several structural aberrations foundby SKY. Protocols used in the preparation of mouse metaphasechromosome suspension, FISH probes, slide pretreatment, slidedenaturation, detection, and imaging are found at http://www.riedlab.nci.nih.gov/index.php/protocols

Statistical analysisWe have applied the Kolmogorov–Smirnov test using R

(https://cran.r-project.org/) to obtain P value statistics and com-pare densities at genomic regions using ChIP-Seq data. Survivalanalysis was determined by log-rank (Mantel–Cox) analysis usingPrism software. P < 0.05 was considered significant.

ChIP-Seq. ChIP-Seq peaks were identified relative to Input DNAusing SICER software (19) with a window size setting of 200 bp, agap setting of 400bp and a FDR setting of 0.001. For a comparisonof ChIP-enrichment between samples a fold-change threshold of1.5 and an FDR setting of 0.001 were used.

RNA-Seq. Differentially expressed genes were identified usingEdgeR (FDR < 0.001 and FC > 2; ref. 23).

CNV-Seq.CNV-Seq software (20)with default settings (P< 0.001)was used to identify regions of copy number alteration. For CNV-Seq analysis of Input-Seq data, regions that increased at least2-fold were considered significant, while for CNV-Seq analysis ofDNA-Seq data, regions that decreased at least 2-fold were con-sidered significant.

Availability of dataThe sequencing data from this study havebeen submitted to the

NCBI Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo) under accession no. GSE94955.

ResultsSMYD5 safeguards genome integrity during ES celldifferentiation

To test whether SMYD5 plays a role in safeguarding genomeintegrity, we evaluated the differentiation of SMYD5-depleted(short hairpin Smyd5; shSmyd5) ES cells relative to shLuc EScells. RNAi knockdown resulted in decreased SMYD5 proteinlevels in shSmyd5 ES cells relative to shLuc ES cells (Fig. 1A).Previously, we showed that EBdifferentiation of SMYD5-depletedES cells leads to complex structures containing bulges lined with aprimitive endoderm (PE) layer (11). Interestingly, by extendingour EB assays from 14 to 21 days, we observed the formation oftransformed-like cells during differentiation in the absence ofSMYD5 (Fig. 1B, i). These transformed colonies continued toemerge from the EBs and proliferate during the remainder of theculture period, and outnumbered the EBs after 21 days of culture.

To further characterize these transformed cells, we isolatedsmall clumps of homogenously translucent andmorphologicallysimilar cells (Fig. 1B, ii and iii) and plated them on tissue culturedishes (Fig. 1B, iv and vi). In addition to their ability to proliferatein suspension, the transformed shSmyd5 cancer cells are capableof proliferating as amonolayer for an extended period of time (>2months), and display a high proliferative rate (17.7-hour celldoubling time; Fig. 1C). Likewise, the shSmyd5 cancer cells arealso capable of proliferating for an extended period of time (>2months) in suspension in an anchorage-independent manner asclusters of cells, which is indicative of transformation (Fig. 1B, iiand iii). Moreover, the shSmyd5 cancer cells proliferated in 3DMatrigel concentrically away from the original embedded cellsprotruding the matrix in all directions (Supplementary Fig. S1A,top). Concomitantly, matrix degradation occurred as evidencedby progressive degradation of the Matrigel layer, and green fluo-rescence was detected following incubation with DQ-collagen IV(Fig. 1D; Supplementary Fig. S1A and S1B). The observed pro-teolytic activity of shSmyd5 cancer cells in 3D Matrigel (Fig. 1D;Supplementary Fig. S1A, bottom and S1B) is most likely derivedfrom cleavage of DQ-collagen IV by the membrane-tethered 1-MMP (MMP14)/MMP2 axis, which has been extensively charac-terized (24) and has been shown to play a role in cell invasion(25). In contrast, control (shLuc) ES cells, which were embeddedin3DMatrigel and cultured in LIF-independentmedia, formedEBstructures, but they did not protrude the 3D Matrigel, and fluo-rescencewas not detected following incubationwithDQ-collagenIV (Supplementary Fig. S1C). Altogether, these results suggest thatshSmyd5 cancer cells are transformed.

To evaluate the tumorigenicity of shSmyd5 cancer cells in vivo,we injected them subcutaneously into SCID-beige mice andobserved the formation of tumors containing mainly adenocar-cinoma-like cells (Fig. 1E). In contrast, control shLuc ES cells formteratomas following subcutaneous injection into SCID-beigemice, which consist of a heterogeneous mixture of cells of thethree germ layers, including ectoderm (keratinized epithelium,epidermis), mesoderm (muscle, adipocytes), and endoderm(glandular epithelium; Fig. 1F). To rule out the possibility thatcell transformation was caused by an integration event thatdisrupted a tumor suppressor gene or by an off target effect ofthe shSmyd5 construct, we tested three different sequences toknock down Smyd5 (11) and all of them led to the formation oftransformed cells (Supplementary Fig. S1D–S1G). As describedabove, while shLuc ES cells formed spherical EB structures con-

Kidder et al.

Cancer Res; 77(23) December 1, 2017 Cancer Research6732

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 5: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Figure 1.

Stochastic transformation of SMYD5-depleted ES cells during differentiation.A,Western blot analysis of SMYD5 in shLuc and shSmyd5 ES cells. HSC70was used as aloading control. B, Schematic of experimental design. (i) Formation of atypical cellular aggregates, which originated from cells budding off from the primitiveendoderm (PE) layer of shSmyd5 EBs. Stereo- (ii) and bright field (iii) microscopy of shSmyd5-EB induced aggregates. (iv–vi) Passaged shSmyd5-EB–derivedcancer cells. C, Proliferation rate of shSmyd5 cancer cells was determined by passaging several times. D, shSmyd5 cancer cells were embedded in Matrigel,and proteolytic activity was ascertained after incubating with DQ-collagen IV (see Materials and Methods). Nuclei were stained with Hoechst dye. E, Hematoxylinand eosin-sectioned tumors generated from shSmyd5 cancer cells. F, Hematoxylin and eosin histologic sections of teratomas generated from shLuc ES cells.G and H, Representative karyotype of a control (shLuc) ES cell (G) and shSmyd5 hypo diploid (top) and hypo tetraploid (bottom) cancer cell analyzed by SKY (H).RGB color displaymetaphase (i), inverted-DAPI stainedmetaphase (ii), pseudo-colored classificationmetaphase (iii), and karyotype of the samemetaphase spread(iv). I, Whole chromosome paint analysis was used to define structural and numerical aberrations of chromosomes X, 3, 14, and 19 in shSmyd5 4n cancer cells.

SMYD5 Regulates Genome Stability

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6733

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 6: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Kidder et al.

Cancer Res; 77(23) December 1, 2017 Cancer Research6734

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 7: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

taining a PE layer during early differentiation (day 6; Supplemen-tary Fig. S1E, left; ref. 11), shSmyd5 ES cells formed structurescontaining bulges lined with a PE layer (Supplementary Fig. S1E,right). The clusters of transformed cells emerged from shSmyd5-1,shSmyd5-2, and shSmyd5-3 EBs (Supplementary Fig. S1F), butnot shLuc EBs. Moreover, the transformed shSmyd5 cancer cellsare capable of proliferating as a monolayer (Supplementary Fig.S1G). In addition, shSmyd5-3 cancer cells developed tumorscontaining adenocarcinoma-like cells in vivo following injectioninto SCID-beige mice (Supplementary Fig. S1H).

To investigate whether the transformed shSmyd5 cells areassociatedwith any chromosomal aberrations, we performed SKYanalysis, using previously defined nomenclature rules (22). Six-teen control (shLuc) ES cell metaphase spreads analyzed by SKYrevealed a diploid population (Fig. 1G), while 14 shSmyd5 cancercell metaphase spreads analyzed by SKY revealed a polyclonalpopulation of 50% near-diploid cells (2n ¼ 40; chromosomenumbers ranged from 39–49; Fig. 1H, top) and 50% near-tetra-ploid cells (chromosome number ranged from 70 to 83; Fig. 1H,bottom). The shSmyd5 cells are of male origin, and in both cellpopulations, the Y chromosome was lost. In the diploid cellpopulation, chromosomes that were clonally gained are X, 1, 2,4, 12, and19.Clonal structural aberrations involved chromosomes12, 14, and 19 (Supplementary Table S2). Structural aberrationsinvolving chromosomes 14 and 19 were found to contain homo-geneously staining regions, which are typically indicative of geneamplifications. Chromosome 19 also was found by SKY to bedeleted at the distal end of the chromosome (19D1).

In the tetraploid shSmyd5 cancer cells, more prevalent chro-mosome losses include chromosomes 10, 11, 13, 17, and 18, anda gain of chromosome 8 was found in 3 of 7 cells. The samestructural aberrations involving chromosomes 14 and 19 werealso found in the tetraploid cell population (Supplementary TableS2). Themain differences between the 2n and 4n shSmyd5 cancercell populations is the increase of chromosome instability (CIN)in the 4n cells, which includes the presence of several novelunbalanced translocations and dicentric chromosomes in the4n population. The dicentric chromosomes were complex in thatthey not only had amplifications of regions from chromosome 19but were also fused to different chromosomes (2, 6, 8, and 12;Supplementary Table S2). In summary, all of the structuralaberrations involving chromosomes 12, 14, and 19, resulted inan imbalance (gains and losses) of these chromosome sequences(Supplementary Table S2).

Whole chromosome paints (WCP) for chromosomes X, 3, 6,14, and 19 were used to further define several clonal aberrationsfound by SKY (Fig. 1I). These FISH results confirmed the deletionsand several translocations observed in the SKY analysis.

Copynumber alterations in shSmyd5 cancer cells are associatedwith decreased H4K20me3/H3K9me3 and enriched withrepetitive elements

Copy number alterations (CNA), which are a structural varia-tion that is a source of genetic variation and disease susceptibility,are commonly found in cancer cells with compromised genomeintegrity (23). To identify regions of CNA between shSmyd5cancer cells and control (shLuc) ES cells, we performed whole-genome DNA-Seq. Using DNA-Seq, we obtained 7.75� and 7.13coverage of the mouse genome for shLuc ES cells and shSmyd5cancer cells, respectively. We then used CNV-Seq software (20) toidentify CNA regions. Using this approach, we found 3,427 CNAregions (size range of 7 kb–2.26 Mb; average size of 235 kb;median size of 15.9 kb; Fig. 2A; red and green). A number of themajor deletions identified using SKY analysis involving chromo-somes 9, 12, 14, 19 were also identified using DNA-Seq.

To evaluate open chromatin regions, we also sequenced ChIPinput chromatin from shLuc ES cells, shSmyd5 ES cells, andshSmyd5 cancer cells (Input-Seq). While we did not observesignificant alterations in open chromatin between shLuc EScells and shSmyd5 ES cells using CNV-Seq software, we found2,088 regions (size range of 54 kb–8.6 Mb; average size of682 kb, median size of 202.2 kb) between shSmyd5 cancercells and shLuc ES cells (77% of altered Input-Seq regionsincreased in shSmyd5 cancer cells; Fig. 2A). These results sug-gest that shSmyd5 cancer cells may exhibit more decondens-ed chromatin relative to control (shLuc) ES cells. Because wedid not identify any significant alterations in open chromatinbetween shLuc and shSmyd5 ES cells, we used shLuc ES cells asa control for shSmyd5 cancer cells.

We then investigated whether regions of altered Input-Seqchromatin compaction identified between shSmyd5 cancer cellsand shLuc ES cells are correlated with SMYD5 binding andchanges in H4K20me3, H3K9me3/2, and HP1 levels in shSmyd5ES cells relative to shLuc ES cells at CNA regions (Fig. 2B). UsingChIP-Seqdata,we generated for shSmyd5ES cells relative to shLucES cells (11), we found that H4K20me3, H3K9me3/2, and HP1levels decrease at CNA regions and regions of altered Input-Seqopen chromatin (Fig. 2B). We also observed SMYD5 enrichmentat regions of altered Input-Seq chromatin compaction (Fig. 2B).Moreover, we also investigated whether there is an associationbetween CNA regions or regions of altered Input-Seq open chro-matin compaction and enrichment of H4K20me3 and SMYD5 byevaluating the percentage of H4K20me3 or SMYD5 islands thatoverlap the CNA regions or the regions of altered Input-Seq openchromatin compaction, respectively. These results show that 33%of H4K20me3 islands and 23% of SMYD5 islands overlap withaltered Input-Seq regions (Fig. 2C, top), and 2% of H4K20me3

Figure 2.Chromosomal aberrations in shSmyd5 cancer cells are enriched at DNA repeats and are associated with an altered epigenomic landscape. A, CNA-Seq analysis ofshSmyd5 cancer cells relative to shLuc ES cells. Circular genome map showing distributions of DNA copy number alterations (DNA-Seq; red and green,outer two histograms) and regionswith open chromatin determined using CNV-Seq software (Input-Seq; orange and blue, inner two histograms). For DNA-Seq, redindicates copy number loss, while green indicates copy number gain. For Input-Seq, blue indicates decreased enrichment of Input, while orange indicatesincreased enrichment.B, Empirical cumulative distribution for the fold-change of density of H4K20me3, H3K9me3, H3K9me2, andHP1 at CNA regions in shSmyd5 EScells relative to shLuc ES cells, and SMYD5 density at CNA regions in ES cells.C,Percentage of H4K20me3, SMYD5, or randomgenomic sequences of comparable sizeand frequency that overlap CNA regions obtained from Input-Seq (top) or DNA-Seq (bottom). CNA regions that increased 10-fold (Input-Seq; n ¼ 4,048) ordecreased 8-fold (DNA-Seq; n ¼ 1,279) in shSmyd5 cancer cells relative to shLuc ES cells were used for these analyses. D and E, LINE and LTR repetitive DNAsequences are enriched in CNA regions. Comparison of Input-Seq (D) and DNA-Seq CNA enriched sequences (red) and annotated repetitive sequencesbetween shSmyd5 cancer cells and shLuc ES cells relative to random regions (black; E). The percentage of CNA regions with at least 30% repeat length is shown forInput-Seq (D) and DNA-Seq (E) results. Note the predominance of LTR- and LINE-repetitive DNA sequences in the CNA-enriched islands.

SMYD5 Regulates Genome Stability

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6735

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 8: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

islands and 2.4% of SMYD5 islands overlap with DNA-Seq CNAregions (Fig. 2C, bottom). SMYD5 and H4K20me3 occupancywas markedly higher at CNA (DNA-Seq) and altered open chro-matin regions (Input-Seq) relative to random genomic sequencesof comparable size and frequency (Fig. 2C), which were used ascontrols, demonstrating that SMYD5 and H4K20me3 areenriched at CNA and altered Input-Seq regions in shSmyd5 cancercells.

BecauseH4K20me3and SMYD5are enriched at LINE- andLTR-repetitiveDNAsequences (11),we characterizedwhether theCNAor Input-Seq regions contain known repetitive sequences. To thisend,we evaluated the percentage ofCNAor Input-Seq regions thatcontain at least 30% coverage of a repeat element. Indeed, theenrichment of LINE and LTR elements was significantly higher atthese regions than at random genomic sequences, which wereused as controls (Fig. 2D and E). We also evaluated the percentcoverage of LTR and LINE sequences for all CNA or Input-Seqregions, and observed enrichment of LTR and LINE sequences inInput-Seq (Supplementary Fig. S2A and S2B) and DNA-Seq(Supplementary Fig. S2C and S2D) CNA regions relative torandom genomic regions. We also evaluated the percent coverageof ERVK and L1 sequences for all CNA or Input-Seq regions, andobserved enrichment of ERVK and L1 sequences in altered Input-Seq regions (Supplementary Fig. S2E and S2F) andDNA-SeqCNAregions (Supplementary Fig. S2G and S2H) relative to randomgenomic regions.

Depletion of SMYD5 leads to decreased H4K20me3 anddecreased H3K9me3 levels during differentiation

Next, we tested whether H4K20me3 and H3K9me3 is reducedin shSmyd5 cancer cells or shSmyd5 EBs relative to control EBs orES cells using ChIP-Seq. These results revealed a global decrease ofH4K20me3 (Fig. 3A) and H3K9me3 (Fig. 3B) in shSmyd5 EBsrelative to shLuc EBs. Likewise, we observed a global decrease inH4K20me3 (Fig. 3C) and H3K9me3 (Fig. 3D) levels in shSmyd5cancer cells relative to day 14 shLuc EBs. Average profiles (Fig. 3Eand F) or boxplots (Fig. 3G and H) around H4K20me3 orH3K9me3 peaks also revealed global decreases in H4K20me3(Fig. 3E and G) and H3K9me3 (Fig. 3F and H) levels during EBdifferentiation and in shSmyd5 cancer cells. Browser views showdecreased levels of H4K20me3 and H3K9me3 (Fig. 3I). Theseresults demonstrate that depletion of SMYD5 leads to morepronounced decreases in H4K20me3 and H3K9me3 levels in EBsand shSmyd5 cancer cells relative to ES cells. However, we foundthat the protein levels of the H3K9 methyltransferase, G9a, weresimilar between shLuc and shSmyd5 ES cells (Supplementary Fig.S3A), and between shLuc and shSmyd5 EBs (Supplementary Fig.S3B), suggesting that changes in H3K9me3 levels in SMYD5-depleted cells is not due to altered expression of G9a.

Our data further show that H4K20me3 and H3K9me3 levelsexhibit even greater decreases in shSmyd5 cancer cells relative toEBs at CNA regions (Fig. 3J and K). These results suggest that thedecrease in H4K20me3 during differentiation in the absence ofSMYD5 is correlated with the formation of CNA regions inshSmyd5 cancer cells.

Depletion of SMYD5 leads to elevated expression of repetitiveDNA elements during differentiation

While we previously demonstrated that SMYD5 mediatessilencing of LTR and LINE repeat classes in ES cells (11), it is

unknown whether SMYD5 silences repetitive DNA elementsduring differentiation. Therefore, we investigated whetherdecreased H4K20me3 leads to increased expression of LINE andLTR repeats in SMYD5-depleted EBs and shSmyd5 cancer cells.Our results show that SMYD5-depleted EBs and shSmyd5 cancercells exhibit increased expression of L1 (Fig. 4A) and ERVK repeatfamilies (Fig. 4B) and subfamilies (Fig. 4C and D). Moreover, weobserved similar expression profiles of ERVK (Supplementary Fig.S4A) and L1 repeats (Supplementary Fig. S4B) between differentshSmyd5 cancer cell samples.

To investigatewhether decreasedH4K20me3 leads to increasedexpression of full-length, or intact, LTR retrotransposons andERVs in SMYD5-depleted EBs and shSmyd5 cancer cells, we firstperformed a de novo search for full-length LTR retrotransposonsandERVs in themouse genomeusing LTRharvest software (http://genometools.org/), and annotated internal features of these LTRregions using LTRdigest software. We then evaluated the expres-sion of LTR internal annotated features in differentiated controlEBs (shLuc) and SMYD5-depleted (shSmyd5), and in shSmyd5cancer cells. These results reveal an increase in expression of LTRretrotransposons/ERV features including sequences encodingviral proteins such as gag and pol during EB differentiation ofSMYD5-depleted cells (Fig. 4E, left), and even higher expressionlevels of these elements in shSmyd5 cancer cells (Fig. 4E, right).We also observed increased expression of IAP family of LTRretrotransposons in shSmyd5 cancer cells relative to ES cells (Fig.4F). These findings suggest that depletion of SMYD5 leads todecreased H4K20me3 levels and increased expression of theunderlying LTR retrotransposon/ERV sequences.

To investigate a relationship between the derepression of LTRretrotransposons/ERV regions, occupancy of SMYD5/H4K20me3,and cancer in humans, we evaluated the overlap between LTRregions and SMYD5/H4K20me3 occupancy. Using this methodwe found 955 regions that were occupied by SMYD5/H4K20me3and contained LTR retrotransposons/ERV sequences (11). Wethen used GREAT gene ontology software (26) to functionallyannotate these regions, and found that genes within these regionswere identified in a copy number alterations study of 191 breasttumor samples, and genes coamplified within MYCN in primaryneuroblastoma tumors (Fig. 4G), suggesting that SMYD5-mediat-ed repression of LTR retrotransposons/ERV regions, marked byH4K20me3, represses expression of nearby genes in mouse cellswhose expression and copy number is positively correlated withhuman cancers.

To investigate whether loss of SMYD5-dependent silencing ofLTR/ERV elements leads to upregulated expression of nearbygenes, we evaluated the number of upregulated genes in shLucday 14 EBs (control) and shSmyd5 cancer cells that contain LTR/ERV sequences within 10 kb of their transcriptional start site(TSS; Fig. 5A and B). Of the 3,715 genes that were upregulatedin shSmyd5 cancer cells relative to control EBs (1.5 fold-change,FDR<0.05), 880 genes (24%) contained LTR/ERV sequences (Fig.5A, left) and 645 genes (17%) contained LINE/L1 sequences (Fig.5A, right) within 10 kb of their TSS. Annotation of the LTR/ERV(Fig. 5C, left) and LINE/L1 (Fig. 5C, right) elements revealed thatthey mainly reside in intronic and intergenic regions. We thenevaluated the expression state of LTR/ERV elements nearby dif-ferentially expressed genes. These results revealed an increase inthe expression of LTR/ERV (Fig. 5D, left) and LINE/L1 (Fig. 5D,right) elements in cancer cells relative to control EBs (Fig. 5D).

Kidder et al.

Cancer Res; 77(23) December 1, 2017 Cancer Research6736

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 9: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Figure 3.

Decreased H4K20me3 and H3K9me3 levels in SMYD5 knockdown EBs and shSmyd5 cancer cells. A and B, Changes in the global distribution of H4K20me3 (A) andH3K9me3 (B) in shSmyd5 EBs relative to shLuc EBs, and H4K20me3 (C) and H3K9me3 (D) in shSmyd5 cancer cells relative to shLuc EBs. Average profile ofH4K20me3 (E) and H3K9me3 (F) in shLuc and shSmyd5 EBs, and shSmyd5 cancer cells. Density of H4K20me3 (G) and H3K9me3 (H) in shLuc and shSmyd5ES cells, shLuc and shSmyd5 EBs, and shSmyd5 cancer cells. P value for all <2.2e-16. I, Browser view of H4K20me3 and H3K9me3 levels in shLuc andshSmyd5 d14 EBs, and shSmyd5 cancer cells. J and K, Density of H4K20me3 and H3K9me3 levels at Input-Seq (J) and DNA-Seq CNA (K) regions in shLuc EB d14,shSmyd5 EB d14, and shSmyd5 cancer cells.

SMYD5 Regulates Genome Stability

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6737

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 10: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Figure 4.

Elevated expression of repetitive LINE/LTR-repetitive DNA elements in SMYD5 knockdown EBs and cancer cells. Fold-change expression of L1 (A) and ERVK(B) repeat families in day 14 differentiated shLuc and shSmyd5 EBs, and in shSmyd5 cancer cells relative to day 14 differentiated control (shLuc) EBs.Fold-change expression of L1 (C) and ERVK (D) repeat subfamilies in day 14 shSmyd5 EBs (left), and in shSmyd5 cancer cells (right) relative to day 14 shLuc EBs.E, Fold-change expression of full-length LTR internal features in day 14 shSmyd5 EBs (left) and in shSmyd5 cancer cells (right) relative to day 14 shLuc EBs.F, qRT-PCR analysis of IAP expression in control (shLuc) ES cells and shSmyd5 cancer cells. G, GREAT (26) gene ontology analysis of regions occupied bySMYD5/H4K20me3 in ES cells overlapping with LTR regions annotated by performing a de novo search for LTR retrotransposons/ERVs in the mouse genomeusing LTRharvest software, and annotating using LTRdigest software.

Kidder et al.

Cancer Res; 77(23) December 1, 2017 Cancer Research6738

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 11: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Figure 5.

Upregulated genes in shSmyd5 cancer cells are associated with LTR/ERV elements and decreased H4K20me3. Loss of SMYD5-dependent silencing ofLTR/ERV elements influences the expression of nearby genes. A, Number of differentially expressed (DE) genes between shLuc EB d14 andshSmyd5 cancer cells (fold change > 1.5; P < 0.05). B, Expression of upregulated genes between shLuc EB d14 and shSmyd5 cancer cells. C, Annotationof LTR/ERV elements nearby differentially expressed genes in shLuc and shSmyd5 ES cells, and shLuc EB d14 and shSmyd5 cancer cells usingHOMER software. D, Fold-change expression of LTR/ERV elements nearby differentially expressed genes relative to total mRNA. E, Density ofH4K20me3 marks nearby LTR/ERV element and within 10 kb of TSS of differentially expressed genes. F and G, OMIM diseases expression analysis.Network2Canvas was used to evaluate upregulated genes (shSmyd5 cancer cells vs. shLuc EB d14) nearby H4K20me3 marks and LTR/LINE (F)and LINE/L1 (G) elements. Each node (square) represents a gene list associated with a gene-set library (OMIM diseases). The brightness (white) ofeach node is determined by its P value.

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6739

SMYD5 Regulates Genome Stability

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 12: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Cancer Res; 77(23) December 1, 2017 Cancer Research6740

Kidder et al.

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 13: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Moreover, we also observed decreased H4K20me3 levels atnearby LTR/ERVK (Fig. 5E, left) and LINE/L1 (Fig. 5E, right) inshSmyd5 cancer cells (Fig. 5E), suggesting that SMYD5-dependentcontrol of H4K20me3 supports the repression of LTR/ERV ele-ments of nearby genes. Overall, these results demonstrate thatSMYD5 influences gene expression of nearby genes by silencingLTR/ERV elements.

Interestingly, the upregulated genes in shSmyd5 cancer cells,which contain H4K20me3 marks and LTR/LINE elements, areenriched in multiple human cancers (lung and breast cancer; Fig.5F and G). These results suggest that SMYD5-dependent silencingof LTR/LINE elements represses the expression of a cancer tran-scriptional program during differentiation.

shSmyd5 cancer cell signature genes predict outcome of patientsurvival

To investigate whether shSmyd5-induced cell transformation isrelated to human cancers, we first identified differentiallyexpressed genes between shSmyd5 cancer cells and shLuc andshSmyd5 ES cells and day 14 EB using RNA-Seq. K-means clus-tering followed by hierarchical clustering identified major pat-terns of gene expression variability (Fig. 6A). Notably, shSmyd5cancer cells expressed a number of MMPs, namely MMP23,MMP2, MMP14, MMP19, and MMP24. The observed proteolyticactivity of shSmyd5 cancer cells in 3D Matrigel (Fig. 1D; Supple-mentary Fig. S1A) is most likely due to the membrane tethered 1-MMP (MMP14)/MMP2 axis, which has been shown to play a rolein cell invasion (25).

Principal component analysis (PCA) was used to determine thethree-dimensional proximity of shSmyd5 cancer cells to shLuc andshSmyd5 ES cells and EBs (Fig. 6B). PCA revealed a skewedtrajectory of shSmyd5 EB differentiation and shSmyd5 cancer cellformation compared with control EB differentiation (Fig. 6B).Moreover, by comparing shSmyd5 cancer cell and shLuc day 14EBDEgeneswith gene expressiondata fromprimitive cells (ES cells)and differentiated cells (day 14 EB), using gene set enrichmentanalysis (GSEA; ref. 27), we found that differentially expressed genesare enriched in EBs (Fig. 6C), suggesting that differentiation genesare dysregulated in shSmyd5 cancer cells. DAVID Gene ontology(GO) analysis further confirmed that developmental GO terms,including cell differentiation, system development, cell develop-ment, gene expression, and lung development, were overrepresent-ed between shSmyd5 cancer cells and shLuc day 14 EB (Fig. 6D).

Next, we identified differentially expressed genes betweenshSmyd5 cancer cells and shLuc EB day 14 cells (Fig. 6E). Our

results show that shSmyd5 cancer cells overexpress lineage-spe-cific genes such as Nog, Snai, Sox7, Thbd, and Spink2 and under-express genes such as Col1a2, Erbb2ip, Gata3, Smyd5, and Sox11(Fig. 6E). Interrogation of the overexpressed and underexpressedsignature genes with a compendium of cancer expression datasetsusing Oncomine (28) revealed a correlation between genes thatwere underexpressed in shSmyd5 cancer cells and underexpressedin human lung adenocarcinoma versus normal lung with knownclinical outcomes (within the top 5% underexpressed; P ¼3.44E�10, ref. 17; P ¼ 1.12E�7, ref. 18; Fig. 6F). We divided thelung adenocarcinoma datasets into two groups: those with high(top 10%) and those with low (bottom 10%) expression ofshSmyd5 cancer cell–repressed genes. A Kaplan–Meier analysiswas then performed on the datasets to investigate an associationbetween the shSmyd5 cancer cell expression signature and patientoutcome (survival; Fig. 6G). We found that lung adenocarcinomapatients with a low expression of shSmyd5 cancer cell–repressedgenes have a decreased rate of survival relative to patients with ahigh expression profile of shSmyd5 cancer cell–repressed genes. Asimilar correlation was observed between the underexpressedshSmyd5 cancer cell repressed genes and additional humancancers including colon adenocarcinomas (Fig. 6H) and invasivebreast carcinomas (Fig. 6I).

We also investigated copy number variation (CNV) and themutational profile of SMYD5 in human cancers using CONAN(29; Fig. 6J). These results show that a loss of heterozygosity ofSMYD5 is present in many human cancers including lung andbreast cancer (Fig. 6J). Altogether, these results suggest that a lossof SMYD5 is associated with cancer formation.

To investigate whether knockdown of SMYD5 may poten-tiate tumor initiation in human cells, we depleted SMYD5 mRNAin human embryonic stem cells (H1 hESCs) using RNAi (seeMaterials and Methods; Fig. 7A and B; Supplementary Fig. S5A).qRT-PCR confirmed that SMYD5 mRNA levels were reducedin shSmyd5 hESCs relative to control (shLuc) hESCs (Fig. 7C).We observed altered differentiation of SMYD5-depleted hESCs,including a greater frequency of cavitated/cystic EBs and adecreased frequency of solid EBs relative to shLuc hESCs at day13 of differentiation (Fig. 7D and E; Supplementary Fig. S5B).Interestingly, by extending the EB assay, we also observed theformation of transformed-like cells during EB differentiation ofshSmyd5 hESCs (Fig. 7F andG; Supplementary Fig. S5C and S5D).Similar to mouse shSmyd5 cancer cells, human shSmyd5 cancercells are capable of proliferating in suspension in an anchorage-independent manner as clusters of cells on low binding dishes

Figure 6.Integrative transcriptomic analysis reveals that a loss of SMYD5 is associatedwith a poor cancer prognosis.A,Analysis of RNA-Seqdata. Differentially expressed (DE)genes (FDR <0.001; fold change >1.5) were clustered according to k-means. B, PCA analysis of differentially expressed genes. C, GSEA of differentiallyexpressed genes between shSmyd5 cancer cells and shLuc d14 EB.D,DAVIDgene ontology (GO) functional annotation of differentially expressed genes.E,Heatmapof differentially expressed genes (FDR < 0.001; fold change >1.5) between shSmyd5 cancer cells and shLuc EB day 14 (green, decrease; red, increase). F,Representative dataset from a meta-analysis of public mRNA expression data from human lung adenocarcinoma tumors with known clinical outcomes usingOncomine. Differentially expressed genes between shSmyd5 cancer cells and shLuc EBd14 (>1.5 fold-change; FDR<0.001)were evaluated usingOncomine software(see Materials and Methods for details). Heatmap: shSmyd5 cancer cell repressed genes were ordered by hierarchical clustering and tumors were sorted byaverage expressionof shSmyd5 cancer cell–repressed genes from low (blue) to high (red).G, The lung adenocarcinomadatasetswere divided into twogroups, thosewith high (top 10%) and those with low (bottom 10%) expression of shSmyd5 cancer cell repressed genes. Kaplan–Meier curves for the overall survival of patientswith lung adenocarcinoma tumors exhibiting high (top 10%, "High shSmyd5 cancer cell repressed," black) or low (bottom 10%, "Low shSmyd5 cancer cell repressed,"expressed) expression of shSmyd5 cancer cell repressed genes are shown. H and I, Representative datasets from meta-analyses of The Cancer Genome Atlas(TCGA) mRNA expression datasets derived from human colon adenocarcinoma tumors and normal colon tissue (H), and human invasive breast carcinomatumors and normal breast tissue (I). shSmyd5 cancer cell–repressed genes and tumors were ordered as described above. J, CNV analysis performed using CONANsoftware of human cancers revealed that SMYD5 is associated with a loss of heterozygosity (LOH). The heatmap shows the percentage of human cancers withmutated SMYD5.

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6741

SMYD5 Regulates Genome Stability

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 14: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Figure 7.

Depletion of SMYD5 in human ES cells and human cancer cells. A, Schematic of experimental design. B, hESCs transduced with shLuc or shSmyd5 lentiviral particles(see Materials and Methods). C, qRT-PCR expression of SMYD5 in shLuc and shSmyd5 hESCs. D, EB formation shows enhanced cavitation of shSmyd5 hEBsrelative to control (shLuc) hEBs at day 13. Inset images in D show solid shLuc EB and cavitated shSmyd5 EB. E, The percentage of solid or cavitated shLucand shSmyd5 EBs. F, EB differentiation at day 15 reveals the formation of atypical cellular aggregates of human shSmyd5 EBs, but not shLuc EBs. shSmyd5 cancer cellclusters at day 15 (arrows show clusters; G), day 18, and day 22 (H). I, Passaged shSmyd5 human cancer cells. J, Bright-field and fluorescence microscopyof shLuc EBs (left) and shSmyd5 human cancer cells (right) cultured in 3DMatrigel with DQ-collagen IV.K and L, qRT-PCR expression analysis of shLuc and shSmyd5HCT-116 colon cancer (K) and A549 lung cancer cells (L). M and O, Tumor growth assay. A total of 106 shLuc or shSmyd5 HCT-116 cells, or shLuc or shSmyd5A549 cells, were subcutaneously injected into SCID-beigemice.N andP,Weight of shLuc and shSmyd5HCT-116 and shLuc and shSmyd5A549 tumors. Tumorweightplots are presented as mean � SEM.

Cancer Res; 77(23) December 1, 2017 Cancer Research6742

Kidder et al.

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 15: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

(Fig. 7G and H; Supplementary Fig. S5E) or as a monolayer (Fig.7I). Moreover, human shSmyd5 cancer cells proliferated in3D Matrigel (Fig. 7J, right), and incubation with DQ-collagenIV revealed proteolytic activity of the embedded cells (Fig. 7J,right). In contrast, control (shLuc) EBs did not proliferate in3D Matrigel, and fluorescence was not detected followingincubation with DQ-collagen IV (Fig. 7J, left).

To evaluate whether depletion of SMYD5 in a nontumorigenicepithelial cell line leads to transformation we knocked downSMYD5 in MCF10A breast epithelial cells and assayed for 3Dgrowth in Matrigel. Our results demonstrate that shSmyd5MCF10A cells exhibit altered growth characteristics relative tocontrol (shLuc) MCF10A cells. Specifically, while shLuc MCF10Acells formed normal round acini (Supplementary Fig. S6A, left),shSmyd5 MCF10A cells formed irregularly shaped acini thatexpanded into the matrix (Supplementary Fig. S6A, right). Inaddition, Phalloidin and DAPI staining showed that shLucMCF10A cells formed normal acini with a lumen (SupplementaryFig. S6B, left), while shSmyd5 MCF10A cells formed irregularacini without a lumen structure (Supplementary Fig. S6B, right).Moreover, 3D culture in Collagen I showed that shSmyd5MCF10A cells exhibit a highly branched structure relative to shLucMCF10A cells (Supplementary Fig. S6C). Taken together, theseresults suggest that depletionof SMYD5 inMCF10A cells leads to apartially transformed phenotype.

To evaluate whether SMYD5 regulates expression of genes inhuman cancer cells, we knocked down SMYD5 in HCT-116colon cancer, A549 lung cancer, and MCF7 breast cancer cellsusing RNAi (see Materials and Methods). qRT-PCR demonstrat-ed that SMYD5 mRNA levels decreased by 86% in shSmyd5HCT-116 cells (Fig. 7K), 82% in shSmyd5 A549 cells (Fig. 7L),and 90% in shSmyd5 MCF7 cells (Supplementary Fig. S7A)relative to shLuc HCT-116, shLuc A549, or shLuc MCF7 cells,respectively. Moreover, we observed upregulation of genesoverexpressed in colon cancer such as CDX2, HNF4A, NAV2,and HOXB9 (30–33), and downregulation of genes under-expressed in colon cancer including SMAD4 and ALDH1 (34,35) in shSmyd5 HCT-116 cells relative to shLuc HCT-116 cells(Fig. 7K). In addition, we observed upregulation of genesoverexpressed in lung cancer such as FOXA3, SOX2, FGFR1,EGFR, RHOV, CCND1, TBX2, and USP7 (36–42) in shSmyd5A549 cells relative to shLuc A549 cells (Fig. 7L). We alsoobserved upregulation of genes overexpressed in breast cancersuch as PGR, MMP11, CCNB1, STK15 (43–46), and down-regulation of SCUBE2 (47) in shSmyd5 MCF7 cells relative toshLuc MCF7 cells (Supplementary Fig. S7A).

We also investigated the in vivo consequence of depletingSMYD5 in human colon and lung cancer cells. To this end, shLucor shSmyd5 HCT116 cells, or shLuc or shSmyd5 A549 cells wereinjected subcutaneously into SCID-beigemice.Mice injectedwithshSmyd5 HCT-116 cells (Fig. 7M and N) or shSmyd5 A549 cells(Fig. 7O and P) exhibited significantly increased tumor growthrelative to tumors generated from shLuc HCT116 shLuc A549cells, respectively.

DiscussionResults presented in this study implicate a role for SMYD5 in

maintaining genome stability of ES cells during differentiation.The formation of transformed cells during differentiation inSMYD5-depleted ES cells is likely attributed to decreased

H4K20 methylation, resulting in decreased levels of heterochro-matin marks (H3K9 methylation/HP1), and transcriptional dys-regulation of the underlying repetitive DNA elements. H4K20methylation has been linked tomultiple cellular processes includ-ing heterochromatin formation, transcriptional regulation andrepression (6), and genome stability (7). Moreover, our resultsshow that loss of SMYD5-dependent silencing of LTR/ERV ele-ments leads to upregulated expression of nearby genes. Ourresults, which implicate a role for SMYD5 in regulating genomestability, are in alignment with the known function of otherH4K20 histone methyltransferases, Suv420h1 and Suv420h2(48). In addition, decreased H4K20me3 levels are a commonhallmark of cancer, where decreasedH4K20me3 levels occur earlyduring transformation, and are progressively lost through themost malignant stages (49). Also, decreased H4K20me3 is cor-related with preneoplasia and squamous cell lung cancer, and thelevel of H4K20me3 was found to decrease with disease progres-sion (50). Concomitantly, decreased H4K20me3 is associatedwith poor prognosis in breast cancer and tumor progression (51).H3K9 methylation also plays a critical role in protecting genomestability, where a loss of Suv39h H3K9 HMTases leads to chro-mosomal instability, and decreased levels of H3K9 methylationhave been found in cancer cells (52). Our results are consistentwith these findings, where we observed decreased levels of therepressive histone modifications H4K20me3 and H3K9me3 inshSmyd5 cancer cells relative to control cells, suggesting thatdecreased levels of H4K20me3 and H3K9me3 is associated withgenome instability, tumorigenesis, and cancer progression. More-over, our results also suggest that SMYD5may potentially act as atumor suppressor in human cells, where depletion of SMYD5 inhESCs resulted in the formation of transformed cells duringdifferentiation, and depletion of SMYD5 in human cancer cellslead to increased tumor growth and changes in expression ofgenes associated with cancer progression or prognosis.

In this study, we found that shSmyd5 cancer cells exhibitchromosomal aberrations, including copy number alterations.Our results revealed a positive correlation between SMYD5 bind-ing, altered levels of repressive histone modifications and hetero-chromatin proteins (H4K20me3, H3K9me3, HP1) in SMYD5-depleted cells, and the occurrence of copy number alterations inshSmyd5 cancer cells. On the basis of these findings, we proposethat SMYD5-dependent H4K20me3 is important for maintaininga heterochromatic structure that is important to protect genomestability. Absence of SMYD5 may favor a more relaxed hetero-chromatic state, whichmay be prone to genome instability duringcellular state transitions from self-renewal to lineage commit-ment. In this case, inappropriate chromosome conformationalchanges may ensue from the induction of specific transcriptionalprograms during differentiation in the absence of adequate levelsof heterochromatin, thus resulting in genome instability. It is alsopossible that derepression of ERVs during differentiation in theabsence of SMYD5 may promote genome instability throughinsertional and postinsertion-based mutagenesis of activatedERVs (53, 54).

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

Authors' ContributionsConception and design: B.L. Kidder, S. Sheng, K. ZhaoDevelopment of methodology: B.L. Kidder, M.M. Bernardo, S. Sheng, K. Zhao

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6743

SMYD5 Regulates Genome Stability

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 16: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

Acquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): B.L. Kidder, R. He, D. Wangsa, M.M. Bernardo,S. ShengAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): B.L. Kidder, R. He, D. Wangsa, H.M. Padilla-Nash,M.M. Bernardo, S. Sheng, T. Ried, K. ZhaoWriting, review, and/or revision of the manuscript: B.L. Kidder, D. Wangsa,M.M. Bernardo, S. Sheng, K. ZhaoStudy supervision: B.L. Kidder, S. Sheng, K. ZhaoOther (analysis of spectral karyotyping, cytogenetics): H.M. Padilla-Nash

AcknowledgmentsWe thankDrs. ZhiyongDing,Wenfei Jin,GangqingHu, AnaRobles, andCurt

Harris for helpful discussions. This work utilized the Wayne State University(WSU) High Performance Computing Grid and the NIH HPC Biowulf cluster.The DNA Sequencing and Genomics Core, Light Microcopy Core, TransgenicCore, and Pathology Core facilities of National Heart, Lung and Blood Institute

assisted with this work. This work was supported by the Division of IntramuralResearch of the National Heart, Lung and Blood Institute and Wayne StateUniversity.

Grant SupportThis work was supported by Wayne State University, Karmanos Cancer

Institute, the Division of Intramural Research of the National Heart, Lung andBlood Institute, and a grant from the National Heart, Lung and Blood Institute(1K22HL126842-01A1) awarded to B.L. Kidder.

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 March 24, 2017; revised August 10, 2017; accepted September 21,2017; published OnlineFirst September 26, 2017.

References1. Grewal SI, Moazed D. Heterochromatin and epigenetic control of gene

expression. Science 2003;301:798–802.2. Grewal SI, Elgin SC. Heterochromatin: new possibilities for the inheritance

of structure. Curr Opin Genet Dev 2002;12:178–87.3. Avner P, Heard E. X-chromosome inactivation: counting, choice and

initiation. Nat Rev Genet 2001;2:59–67.4. Guarente L. Sir2 links chromatin silencing, metabolism, and aging. Genes

Dev 2000;14:1021–6.5. Beck DB, Oda H, Shen SS, Reinberg D. PR-Set7 and H4K20me1: at the

crossroads of genome integrity, cell cycle, chromosome condensation, andtranscription. Genes Dev 2012;26:325–37.

6. Karachentsev D, Sarma K, Reinberg D, Steward R. PR-Set7-dependentmethylation of histone H4 Lys 20 functions in repression of gene expres-sion and is essential for mitosis. Genes Dev 2005;19:431–5.

7. Schotta G, Sengupta R, Kubicek S, Malin S, Kauer M, Callen E, et al. Achromatin-wide transition to H4K20 monomethylation impairs genomeintegrity and programmed DNA rearrangements in the mouse. Genes Dev2008;22:2048–61.

8. Vermeulen M, Eberl HC, Matarese F, Marks H, Denissov S, Butter F, et al.Quantitative interaction proteomics and genome-wide profiling of epige-netic histone marks and their readers. Cell 2010;142:967–80.

9. BotuyanMV, Lee J, Ward IM, Kim JE, Thompson JR, Chen J, et al. Structuralbasis for the methylation state-specific recognition of histone H4-K20 by53BP1 and Crb2 in DNA repair. Cell 2006;127:1361–73.

10. Schotta G, Lachner M, Sarma K, Ebert A, Sengupta R, Reuter G, et al. Asilencing pathway to induce H3-K9 and H4-K20 trimethylation at consti-tutive heterochromatin. Genes Dev 2004;18:1251–62.

11. Kidder BL, Hu G, Cui K, Zhao K. SMYD5 regulates H4K20me3-markedheterochromatin to safeguard ES cell self-renewal and prevent spuriousdifferentiation. Epigenetics Chromatin 2017;10:8.

12. Matsui T, LeungD,MiyashitaH,Maksakova IA,MiyachiH, KimuraH, et al.Proviral silencing in embryonic stem cells requires the histone methyl-transferase ESET. Nature 2010;464:927–31.

13. Kidder BL, Hu G, Yu ZX, Liu C, Zhao K. Extended self-renewal andaccelerated reprogramming in the absence of Kdm5b. Mol Cell Biol2013;33:4793–810.

14. Beers J, GulbransonDR,GeorgeN, Siniscalchi LI, Jones J, Thomson JA, et al.Passaging and colony expansion of human pluripotent stem cells byenzyme-free dissociation in chemically defined culture conditions. NatProtoc 2012;7:2029–40.

15. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B. Mapping andquantifying mammalian transcriptomes by RNA-Seq. Nat Methods 2008;5:621–8.

16. RobinsonMD,McCarthyDJ, SmythGK. edgeR: a Bioconductor package fordifferential expression analysis of digital gene expression data. Bioinfor-matics 2009;26:139–40.

17. Bhattacharjee A, Richards WG, Staunton J, Li C, Monti S, Vasa P, et al.Classification of human lung carcinomas by mRNA expression profilingreveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci U S A2001;98:13790–5.

18. Beer DG, Kardia SL, Huang CC, Giordano TJ, Levin AM, Misek DE, et al.Gene-expression profiles predict survival of patients with lung adenocar-cinoma. Nat Med 2002;8:816–24.

19. Zang C, Schones DE, Zeng C, Cui K, Zhao K, PengW. A clustering approachfor identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 2009;25:1952–8.

20. Xie C, TammiMT.CNV-seq, a newmethod todetect copy number variationusing high-throughput sequencing. BMC Bioinformatics 2009;10:80.

21. Liyanage M, Coleman A, du Manoir S, Veldman T, McCormack S, DicksonRB, et al. Multicolour spectral karyotyping of mouse chromosomes. NatGenet 1996;14:312–5.

22. Padilla-Nash HM, Hathcock K, McNeil NE, Mack D, Hoeppner D, Ravin R,et al. Spontaneous transformation of murine epithelial cells requires theearly acquisition of specific chromosomal aneuploidies and genomicimbalances. Genes Chromosomes Cancer 2012;51:353–74.

23. Girirajan S, Campbell CD, Eichler EE. Human copy number variation andcomplex genetic disease. Annu Rev Genet 2011;45:203–26.

24. Hernandez-Barrantes S, Bernardo M, Toth M, Fridman R. Regulation ofmembrane type-matrix metalloproteinases. Semin Cancer Biol 2002;12:131–8.

25. Friedl P, Wolf K. Tube travel: the role of proteases in individual andcollective cancer cell invasion. Cancer Res 2008;68:7247–9.

26. McLeanCY, BristorD,HillerM,Clarke SL, Schaar BT, LoweCB, et al. GREATimproves functional interpretation of cis-regulatory regions. Nat Biotech-nol 2010;28:495–501.

27. 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.

28. Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, et al.ONCOMINE: a cancer microarray database and integrated data-miningplatform. Neoplasia 2004;6:1–6.

29. Forer L, Schonherr S,WeissensteinerH,Haider F, Kluckner T,Gieger C, et al.CONAN: copy number variation analysis software for genome-wide asso-ciation studies. BMC Bioinformatics 2010;11:318.

30. Chellappa K, Robertson GR, Sladek FM. HNF4alpha: a new biomarker incolon cancer? Biomark Med 2012;6:297–300.

31. Tan F, Zhu H, Tao Y, Yu N, Pei Q, Liu H, et al. Neuron navigator 2overexpression indicates poor prognosis of colorectal cancer and promotesinvasion through the SSH1L/cofilin-1 pathway. J Exp Clin Cancer Res2015;34:117.

32. Dalerba P, Sahoo D, Paik S, Guo X, Yothers G, Song N, et al. CDX2 as aprognostic biomarker in stage II and stage III colon cancer. N Engl J Med2016;374:211–22.

33. Huang K, Yuan R, Wang K, Hu J, Huang Z, Yan C, et al. Overexpression ofHOXB9 promotesmetastasis and indicates poor prognosis in colon cancer.Chin J Cancer Res 2014;26:72–80.

34. Woodford-Richens KL, Rowan AJ, Gorman P, Halford S, Bicknell DC,Wasan HS, et al. SMAD4 mutations in colorectal cancer probablyoccur before chromosomal instability, but after divergence of the

Cancer Res; 77(23) December 1, 2017 Cancer Research6744

Kidder et al.

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 17: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

microsatellite instability pathway. Proc Natl Acad Sci U S A 2001;98:9719–23.

35. Alazzouzi H, Alhopuro P, Salovaara R, Sammalkorpi H, Jarvinen H,Mecklin JP, et al. SMAD4 as a prognostic marker in colorectal cancer. ClinCancer Res 2005;11:2606–11.

36. Clinical Lung Cancer Genome Project; Network Genomic Medicine. Agenomics-based classification of human lung tumors. Sci Transl Med2013;5:209ra153.

37. Rudin CM, Durinck S, Stawiski EW, Poirier JT, Modrusan Z, Shames DS,et al. Comprehensive genomic analysis identifies SOX2 as a frequentlyamplified gene in small-cell lung cancer. Nat Genet 2012;44:1111–6.

38. Guo M, Tomoshige K, Meister M, Muley T, Fukazawa T, Tsuchiya T, et al.Gene signature driving invasive mucinous adenocarcinoma of the lung.EMBO Mol Med 2017;9:462–81.

39. ShepelevMV, Korobko IV. TheRHOVgene is overexpressed in humannon-small cell lung cancer. Cancer Genet 2013;206:393–7.

40. Gautschi O, Ratschiller D, Gugger M, Betticher DC, Heighway J. Cyclin D1in non-small cell lung cancer: a key driver of malignant transformation.Lung Cancer 2007;55:1–14.

41. Zhang Z, Guo Y. High TBX2 expression predicts poor prognosis in non-small cell lung cancer. Neoplasma 2014;61:476–80.

42. Zhao GY, Lin ZW, Lu CL, Gu J, Yuan YF, Xu FK, et al. USP7 overexpressionpredicts a poor prognosis in lung squamous cell carcinoma and large cellcarcinoma. Tumour Biol 2015;36:1721–9.

43. Horwitz KB, McGuire WL. Estrogen control of progesterone receptor inhuman breast cancer. Correlation with nuclear processing of estrogenreceptor. J Biol Chem 1978;253:2223–8.

44. Basset P, Okada A, Chenard MP, Kannan R, Stoll I, Anglard P, et al. Matrixmetalloproteinases as stromal effectors of human carcinoma progression:therapeutic implications. Matrix Biol 1997;15:535–41.

45. Ding K, Li W, Zou Z, Zou X, Wang C. CCNB1 is a prognostic biomarker forERþ breast cancer. Med Hypotheses 2014;83:359–64.

46. Dai Q, Cai QY, Shu XO, Ewart-Toland A, Wen WQ, Balmain A, et al.Synergistic effects of STK15 gene polymorphisms and endogenous estro-gen exposure in the risk of breast cancer. Cancer Epidemiol Biomarkers Prev2004;13:2065–70.

47. Lin YC, Lee YC, Li LH, Cheng CJ, Yang RB. Tumor suppressorSCUBE2 inhibits breast-cancer cell migration and invasion throughthe reversal of epithelial-mesenchymal transition. J Cell Sci 2014;127:85–100.

48. Jorgensen S, Schotta G, Sorensen CS. Histone H4 lysine 20 methylation:key player in epigenetic regulation of genomic integrity. Nucleic Acids Res2013;41:2797–806.

49. Fraga MF, Ballestar E, Villar-Garea A, Boix-Chornet M, Espada J, SchottaG, et al. Loss of acetylation at Lys16 and trimethylation at Lys20 ofhistone H4 is a common hallmark of human cancer. Nat Genet2005;37:391–400.

50. VanDen Broeck A, Brambilla E,Moro-Sibilot D, Lantuejoul S, Brambilla C,Eymin B, et al. Loss of histone H4K20 trimethylation occurs in preneo-plasia and influences prognosis of non-small cell lung cancer. Clin CancerRes 2008;14:7237–45.

51. Yokoyama Y, Matsumoto A, Hieda M, Shinchi Y, Ogihara E, Hamada M,et al. Loss of histone H4K20 trimethylation predicts poor prognosis inbreast cancer and is associated with invasive activity. Breast Cancer Res2014;16:R66.

52. Feinberg AP, KoldobskiyMA, Gondor A. Epigenetic modulators, modifiersand mediators in cancer aetiology and progression. Nat Rev Genet2016;17:284–99.

53. Konkel MK, Batzer MA. A mobile threat to genome stability: the impact ofnon-LTR retrotransposons upon the human genome. Semin Cancer Biol2010;20:211–21.

54. Hedges DJ, Deininger PL. Inviting instability: transposable elements,double-strand breaks, and the maintenance of genome integrity. MutatRes 2007;616:46–59.

www.aacrjournals.org Cancer Res; 77(23) December 1, 2017 6745

SMYD5 Regulates Genome Stability

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828

Page 18: SMYD5 Controls Heterochromatin and Chromosome Integrity ... · erties of the H3K9 methyltransferase ESET during differentiation (12) implicates a role for histone-modifying enzymes

2017;77:6729-6745. Published OnlineFirst September 26, 2017.Cancer Res   Benjamin L. Kidder, Runsheng He, Darawalee Wangsa, et al.   during Embryonic Stem Cell DifferentiationSMYD5 Controls Heterochromatin and Chromosome Integrity

  Updated version

  10.1158/0008-5472.CAN-17-0828doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2017/09/26/0008-5472.CAN-17-0828.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/77/23/6729.full#ref-list-1

This article cites 54 articles, 18 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/77/23/6729.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://cancerres.aacrjournals.org/content/77/23/6729To request permission to re-use all or part of this article, use this link

on December 15, 2020. © 2017 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-0828