9
Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Parafn-embedded tissue blocks of 72 primary tumors were collected from the archives of the De- partment of Pathology, Helsinki University Hospital (Finland) after approval of the Ethics Committee. The clinicopathological data have been published (1). Four-micrometer sections were subjected to dual colorimetric immunohistochemistry (IHC) (Envision G/2 Doublestain; Dako) using antibodies SP2 (Neo- markers) for PR and CK5 (Leica). Tumors with 1% 3, 3- diaminobenzidine (DAB)-stained nuclei or 1% Perm Red- stained cytoplasm were considered positive. Cell Lines and Reagents. T47D cells were from Iafa Keydar (Tel Aviv University, Tel Aviv, Israel); MCF-7 breast cancer cells were from Sam Brooks (Michigan Cancer Foundation, Detroit, MI); MCF-10A, BT-474, MDA231, SKBR3, and BT-20 were from the ATCC. SUM149 and SUM159 were from Asterand. Cell lines were maintained in the media suggested by the suppliers. BJ-3Z were derived as previously reported (2, 3). Cells were tagged with ZsGreen uor by retroviral infection as previously described (4). All parental and derived cell lines have been authenticated and deemed pure by short tandem repeat analysis. For hormone and drug inhibitor treatments, or hormone withdrawal, cells were cultured in phenol red-free MEM and 5% FBS depleted of hormones by 30-min incubation at 45 °C with dextran-coated charcoal pellets [0.25% Norit A, 0.0025% dextran in 0.01 M Tris·HCl (pH 8.0); 1 mL charcoal per 2 mL serum] followed by ltration. A complete list of reagents and antibodies can be found in Table S2. Xenografts and Tumor-Derived Cell Lines. All animal procedures were performed according to a protocol approved by the Uni- versity of Colorado Institutional Animal Care and Use Com- mittee. Tumors were grown by injecting 10 6 ZsGreen-tagged cells with Matrigel into the mammary fat pad of ovariectomized (ovxd) athymic nu/nu (Taconic) or nonobese diabetic/SCID (National Cancer Institute) mice. Hormones were delivered by s.c. silastic pellets containing cellulose (placebo), or 2 mg es- tradiol-17β (E) with cellulose. Tumors were harvested at one to four in vivo passage generations. At necropsy, tumors were re- sected aseptically, mechanically dissociated, and enzymatically digested for 2 to 3 h at 37 °C using a solution of HBSS containing collagenase IV, hyaluronidase, and DNase I (Sigma). Dissoci- ated tumor cells were washed extensively before both stromal and tumor cells were plated into media containing neomycin (0.1 mg/mL) to select against the stromal cells and to select for tumor cells expressing the Neo resistance marker cloned into the ZsGreen plasmid. A list of tumor-derived cell lines grown from the parental T47D line is provided in Table S1. Numbers denote cell lines derived from the same tumor. In vitro, tumor cells were divided into steroid-depleted media supplemented with 1 nM E (denoted E-#) or no E (denoted EWD-#). Some lines were supplemented with EWD plus γ-secretase inhibitor (GSI) [de- noted EWD-#+DAPT, compound E (CpdE), or XIX (dened in Table S1 legend)], or E plus tamoxifen (Tam) or fulvestrant (Fulv) (Table S1). E lines grew at a rate comparable to parental cells. Proliferation of EWD cells slowed initially, entered crisis, then resumed after 3040 d, with GSI having no overt inhibitory or stimulatory effects. EWD-8 did not undergo crisis; it was derived from a placebo-treated mouse and had adapted to EWD in vivo. 3D Culture, Immunocytochemistry (ICC), IHC, and Western Blotting. IHC of 3D Matrigel colonies was reported previously (5). Briey, cells were plated into eight-well chambered slides (BD) precoated with 60 μL growth-factor reduced Matrigel and in- cubated overnight before treatments. For assessment of cell proliferation by BrdU incorporation, the 3D colonies were treated with 0.25 mg/mL BrdU for 3 h before harvest. After treatments, the Matrigel and colonies were embedded in His- togel (Richard-Allan Scientic), formalin xed overnight, and then switched into 70% vol/vol ethanol. The histogel and Ma- trigel blocks containing the xed colonies were then parafn embedded and cut on a microtome (Leica) into 4-μm sections. Sections were stained by standard IHC procedures. Antibody incubations used Tris-buffered saline (pH 7.4) containing 1% Triton-X. Nuclei were stained with 1 μg/mL DAPI dissolved in methanol, and cells were mounted in Fluoromount (Sigma) for uorescence or Permount (Fisher) for colorimetric stains. Dual colorimetric staining with DAB and Perm Red used Envision G/2 Doublestain Kit (Dako). Images were taken on a Nikon Eclipse E600 microscope equipped with a CoolSnap fx camera (Photometrics) operated by ImagePro software (Media Cyber- netics). For Western blotting, cells were solubilized in RIPA buffer [50 mM Tris·HCl (pH 7.4), 150 mM NaCl, 1% Triton X- 100, 1% sodium deoxycholate, and 0.1% wt/vol SDS] amended with protease and phosphatase inhibitors (20 mM β-glycer- ophosphate, 0.1 mM sodium orthovanadate, 10 mM sodium uoride, 1 mM PMSF, and protease inhibitor mixture; Roche). Protein concentrations were determined by Bradford assay (Bi- orad). Fifty micrograms of total protein were resolved on 10% SDS/PAGE gels, transferred, and probed. A list of antibodies is provided in Table S2. Gene Expression Proling and Analysis. All microarray procedures including total cellular RNA isolation, amplication, and Cy3/Cy5 labeling of 1.5 μg of experimental and reference RNA were done as previously described (6, 7). Microarray hybridizations were performed using Agilent human oligonucleotide 4x44K custom designed gene chips. These data have been made available for download from the Gene Expression Omnibus (GEO) database, accession no. GSE31870. The gene probes were initially ltered by requiring the lowest normalized intensity values in both sample and reference to be >10. Probes mapping to the same gene were averaged, and of 33,257 probes, a total of 16,937 genes were examined. The expression data were analyzed using a two-class unpaired signicance analysis of microarrays, and all 1,298 genes comprising the luminobasalsignature referenced in the article were considered differentially expressed with a false discovery rate of 0. Clustering analyses were performed using Cluster v2.12 (8). Colored squares in gures containing cluster analyses indicate the relative mean transcript abundance (in log 10 scale), with higher expression in red and lower expression in green. For comparisons with the 52 breast cancer cell line dataset generated by Neve et al. (9), Affymetrix Human Exon 1.0 ST Arrays were performed in triplicate from RNA isolated from the E-3 and EWD-8 cell lines. A total of 12,108 common genes were identied, and the raw data were normalized using robust multiarray average. An array platform/cell line correction value was generated by normalizing the gene expression values from the E-3 line in this study with the T47D line used in the Neve dataset. This correction value was then applied to the EWD-8 arrays before further analyses. Haughian et al. www.pnas.org/cgi/content/short/1106509108 1 of 9

Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

Supporting InformationHaughian et al. 10.1073/pnas.1106509108SI Materials and MethodsPrimary Breast Cancers. Paraffin-embedded tissue blocks of 72primary tumors were collected from the archives of the De-partment of Pathology, Helsinki University Hospital (Finland)after approval of the Ethics Committee. The clinicopathologicaldata have been published (1). Four-micrometer sections weresubjected to dual colorimetric immunohistochemistry (IHC)(Envision G/2 Doublestain; Dako) using antibodies SP2 (Neo-markers) for PR and CK5 (Leica). Tumors with ≥1% 3, 3′-diaminobenzidine (DAB)-stained nuclei or ≥1% Perm Red-stained cytoplasm were considered positive.

Cell Lines and Reagents. T47D cells were from Iafa Keydar (TelAviv University, Tel Aviv, Israel); MCF-7 breast cancer cells werefrom Sam Brooks (Michigan Cancer Foundation, Detroit, MI);MCF-10A, BT-474, MDA231, SKBR3, and BT-20 were from theATCC. SUM149 and SUM159 were from Asterand. Cell lineswere maintained in the media suggested by the suppliers. BJ-3Zwere derived as previously reported (2, 3). Cells were tagged withZsGreen fluor by retroviral infection as previously described (4).All parental and derived cell lines have been authenticated anddeemed pure by short tandem repeat analysis. For hormone anddrug inhibitor treatments, or hormone withdrawal, cells werecultured in phenol red-free MEM and 5% FBS depleted ofhormones by 30-min incubation at 45 °C with dextran-coatedcharcoal pellets [0.25% Norit A, 0.0025% dextran in 0.01 MTris·HCl (pH 8.0); 1 mL charcoal per 2 mL serum] followed byfiltration. A complete list of reagents and antibodies can befound in Table S2.

Xenografts and Tumor-Derived Cell Lines. All animal procedureswere performed according to a protocol approved by the Uni-versity of Colorado Institutional Animal Care and Use Com-mittee. Tumors were grown by injecting 106 ZsGreen-tagged cellswith Matrigel into the mammary fat pad of ovariectomized(ovx’d) athymic nu/nu (Taconic) or nonobese diabetic/SCID(National Cancer Institute) mice. Hormones were delivered bys.c. silastic pellets containing cellulose (placebo), or 2 mg es-tradiol-17β (E) with cellulose. Tumors were harvested at one tofour in vivo passage generations. At necropsy, tumors were re-sected aseptically, mechanically dissociated, and enzymaticallydigested for 2 to 3 h at 37 °C using a solution of HBSS containingcollagenase IV, hyaluronidase, and DNase I (Sigma). Dissoci-ated tumor cells were washed extensively before both stromaland tumor cells were plated into media containing neomycin (0.1mg/mL) to select against the stromal cells and to select for tumorcells expressing the Neo resistance marker cloned into theZsGreen plasmid. A list of tumor-derived cell lines grown fromthe parental T47D line is provided in Table S1. Numbers denotecell lines derived from the same tumor. In vitro, tumor cells weredivided into steroid-depleted media supplemented with 1 nM E(denoted E-#) or no E (denoted EWD-#). Some lines weresupplemented with EWD plus γ-secretase inhibitor (GSI) [de-noted EWD-#+DAPT, compound E (CpdE), or XIX (definedin Table S1 legend)], or E plus tamoxifen (Tam) or fulvestrant(Fulv) (Table S1). E lines grew at a rate comparable to parentalcells. Proliferation of EWD cells slowed initially, entered crisis,then resumed after 30–40 d, with GSI having no overt inhibitoryor stimulatory effects. EWD-8 did not undergo crisis; it wasderived from a placebo-treated mouse and had adapted to EWDin vivo.

3D Culture, Immunocytochemistry (ICC), IHC, and Western Blotting.IHC of 3D Matrigel colonies was reported previously (5).Briefly, cells were plated into eight-well chambered slides (BD)precoated with 60 μL growth-factor reduced Matrigel and in-cubated overnight before treatments. For assessment of cellproliferation by BrdU incorporation, the 3D colonies weretreated with 0.25 mg/mL BrdU for 3 h before harvest. Aftertreatments, the Matrigel and colonies were embedded in His-togel (Richard-Allan Scientific), formalin fixed overnight, andthen switched into 70% vol/vol ethanol. The histogel and Ma-trigel blocks containing the fixed colonies were then paraffinembedded and cut on a microtome (Leica) into 4-μm sections.Sections were stained by standard IHC procedures. Antibodyincubations used Tris-buffered saline (pH 7.4) containing 1%Triton-X. Nuclei were stained with 1 μg/mL DAPI dissolved inmethanol, and cells were mounted in Fluoromount (Sigma) forfluorescence or Permount (Fisher) for colorimetric stains. Dualcolorimetric staining with DAB and Perm Red used EnvisionG/2 Doublestain Kit (Dako). Images were taken on a NikonEclipse E600 microscope equipped with a CoolSnap fx camera(Photometrics) operated by ImagePro software (Media Cyber-netics). For Western blotting, cells were solubilized in RIPAbuffer [50 mM Tris·HCl (pH 7.4), 150 mM NaCl, 1% Triton X-100, 1% sodium deoxycholate, and 0.1% wt/vol SDS] amendedwith protease and phosphatase inhibitors (20 mM β-glycer-ophosphate, 0.1 mM sodium orthovanadate, 10 mM sodiumfluoride, 1 mM PMSF, and protease inhibitor mixture; Roche).Protein concentrations were determined by Bradford assay (Bi-orad). Fifty micrograms of total protein were resolved on 10%SDS/PAGE gels, transferred, and probed. A list of antibodies isprovided in Table S2.

Gene Expression Profiling and Analysis. All microarray proceduresincluding total cellular RNA isolation, amplification, and Cy3/Cy5labeling of 1.5 μg of experimental and reference RNA were doneas previously described (6, 7). Microarray hybridizations wereperformed using Agilent human oligonucleotide 4x44K customdesigned gene chips. These data have been made available fordownload from the Gene Expression Omnibus (GEO) database,accession no. GSE31870. The gene probes were initially filteredby requiring the lowest normalized intensity values in bothsample and reference to be >10. Probes mapping to the samegene were averaged, and of 33,257 probes, a total of 16,937genes were examined. The expression data were analyzed usinga two-class unpaired significance analysis of microarrays, and all1,298 genes comprising the “luminobasal” signature referencedin the article were considered differentially expressed with a falsediscovery rate of 0. Clustering analyses were performed usingCluster v2.12 (8). Colored squares in figures containing clusteranalyses indicate the relative mean transcript abundance (inlog10 scale), with higher expression in red and lower expressionin green. For comparisons with the 52 breast cancer cell linedataset generated by Neve et al. (9), Affymetrix Human Exon 1.0ST Arrays were performed in triplicate from RNA isolated fromthe E-3 and EWD-8 cell lines. A total of 12,108 common geneswere identified, and the raw data were normalized using robustmultiarray average. An array platform/cell line correction valuewas generated by normalizing the gene expression values fromthe E-3 line in this study with the T47D line used in the Nevedataset. This correction value was then applied to the EWD-8arrays before further analyses.

Haughian et al. www.pnas.org/cgi/content/short/1106509108 1 of 9

Page 2: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

Two separate human primary tumor microarray datasets wereused in these studies. The 516 tumor dataset was generated bycombining three publicly available datasets available in the GEOrepository: GSE2034 (10), GSE12276 (11), and GSE2603 (12).These tumor data were combined using distance weighted dis-crimination (13) and then subtyped using the PAM50 and clau-din-low predictors (14, 15) with 159 luminal A, 112 luminal B, 79Her2, 101 basal-like, and 65 claudin-low subtype tumors definedin the dataset. The NKI265 tumor dataset (http://microarray-pubs.stanford.edu/wound_NKI) contains 84 luminal A, 69 lumi-nal B, 49 Her2, 42 basal-like, and 21 claudin-low tumors aspreviously described (15). Cluster (v2.12) and Java Treeview(v1.1.3) were used to assess how the 1,298 gene luminobasalsignature clustered tumors within the 516 and NKI265 datasets.

Karyotype Analysis. Karyotype verification of clonal origin froma single cell line was performed as previously reported (16), withminor modifications. Briefly, cells were arrested with colcemid(0.05 μg/μL; 2.5 h), harvested in hypotonic KCl, and air dried toprecleaned slides. Giemsa/Trypsin/Leishman banding was per-formed 4 d later by standard methods. Images were acquired andanalyzed using the CytoVision (version 3.1) system (AppliedImaging). At least five metaphases per line were analyzed, andkaryotyping designation followed the International System forHuman Cytogenetic Nomenclature 2009 (17). Of 16 structuralabnormalities previously reported in the T47D cell line (18), theparental T47D, E-3, EWD-3, and EWD-8 lines shared a total of13 structural abnormalities, consistent with their clonal deriva-tion from the T47D breast cancer cell line. This analysis failed toidentify gain or loss of broad chromosomal abnormalities thatwould distinguish luminal and luminobasal cells.

Genotype Array. Genotyping of the parental T47D, E-3, EWD-3,and EWD-8 cell lines was performed on the Illumina InfiniumGenotyping platform. Genomic DNA (gDNA) was isolated usingthe QIAamp DNA Mini Kit (Qiagen) and quantitated usingPicoGreen (Invitrogen). The gDNA was processed according toIllumina’s established protocols and DNA hybridized to theHumanOmni2.5-Quad BeadChip and imaged by an iScanReader (Illumina). Changes to SNPs between the lines wereassessed using the reproducibility and heritability function inGenome Studio Software (Illumina). A total of ≈2.43 × 106

SNPs in the genome were evaluated. The number of variantsbetween the parental T47D and E-3, EWD-3, and EWD-8 lineswas 191, 62, and 380, respectively. For reference, differences

between the genotype of an unrelated human female comparedwith the parental T47D line was 12,681 variant SNPs. The lim-ited number of variant SNPs confirms the T47D origin of theE-3, EWD-3, and EWD-8 sublines and does not suggest a highdegree of genetic instability at the level of the DNA base pairs.

Antibody Labeling and Flow Cytometry. Cells were harvested usingHBSS supplemented with 1 mM EDTA and blocked with 10%normal human serum (Millipore) and 0.5%BSA for 15min at 4 °C.For intracellular markers, cells were fixed using the Cytofix/Cy-toperm Fixation/Permeabilization kit (BD Biosciences) for 20 minat 4 °C. Cells were stained for 30 min and then washed withmultiple wash buffer changes for at least 30 min. When necessary,primary antibodies were conjugated using the Zenon AntibodyLabeling Kit (Invitrogen) per the manufacturer’s instructions.

Notch Promoter Reporter and shRNA Constructs. Cellular Notchactivity was assessed using the RBP-Јκ-Luc Cignal Reporter (SABiosciences). Breast cancer cell lines were transfected with Lip-oD293 (SignaGen Laboratories) and cotransfected with SV-40promoter-renilla luciferase to control for transfection efficiency.Lentiviral particles encoding nonspecific (scrambled) or Notch1targeted shRNAs were obtained from the University of ColoradoFunctional Genomics core laboratory. The Notch1 shRNA con-struct is identified within the Mission shRNA library (Sigma) asTRCN0000003360. The scrambled control vector was the Mis-sion Non-Target shRNA Control Vector (SHC002; Sigma).Early-passage cells from the tumor-derived E-3 line were in-fected by mixing lentivirus with media containing 8 μg/mL pol-ybrene. Stably infected cells were selected using 2 μg/mLpuromycin for 1 wk and then maintained in media containing lowdose (0.2 μg/mL) puromycin. The cells were switched out ofE-containing media and maintained under these EWD/low-dosepuromycin conditions for ≈8 wk to enable expansion of the lu-minobasal population.

Statistical Analysis. All reported values are mean ± SEM. Sig-nificance levels for comparisons between treatment groups, orfor comparisons of gene expression values, were analyzed usingan unpaired Student t test or ANOVA. The microarray clusteranalyses were derived using Java Treeview version 1.1.3. Hier-archical clustering showing average linkage was performed usingCluster v2.12 (8). The differences were considered significantwhen the P value was <0.05.

1. Joensuu K, Heikkilä P, Andersson LC (2008) Tumor dormancy: Elevated expression ofstanniocalcins in late relapsing breast cancer. Cancer Lett 265:76e83.

2. Jacobsen BM, et al. (2006) Spontaneous fusion with, and transformation of mousestroma by, malignant human breast cancer epithelium. Cancer Res 66:8274e8279.

3. Sartorius CA, et al. (1994) New T47D breast cancer cell lines for the independent studyof progesterone B- and A-receptors: Only antiprogestin-occupied B-receptors areswitched to transcriptional agonists by cAMP. Cancer Res 54:3868e3877.

4. Harrell JC, et al. (2006) Estrogen receptor positive breast cancer metastasis: Alteredhormonal sensitivity and tumor aggressiveness in lymphatic vessels and lymph nodes.Cancer Res 66:9308e9315.

5. Pinto MP, et al. (2010) Vascular endothelial growth factor secreted by activatedstroma enhances angiogenesis and hormone-independent growth of estrogenreceptor-positive breast cancer. Cancer Res 70:2655e2664.

6. Hu Z, et al. (2006) The molecular portraits of breast tumors are conserved acrossmicroarray platforms. BMC Genomics 7:96.

7. Harrell JC, et al. (2011) Genomic analysis identifies unique signatures predictive ofbrain, lung, and liver relapse. Breast Cancer Res Treat, 10.1007/s10549-011-1619-7.

8. Eisen MB, Spellman PT, Brown PO, Botstein D (1998) Cluster analysis and display ofgenome-wide expression patterns. Proc Natl Acad Sci USA 95:14863e14868.

9. Neve RM, et al. (2006) A collection of breast cancer cell lines for the study offunctionally distinct cancer subtypes. Cancer Cell 10:515e527.

10. Wang Y, et al. (2005) Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 365:671e679.

11. Bos PD, et al. (2009) Genes that mediate breast cancer metastasis to the brain. Nature459:1005e1009.

12. Minn AJ, et al. (2005) Genes that mediate breast cancer metastasis to lung. Nature436:518e524.

13. Benito M, et al. (2004) Adjustment of systematic microarray data biases.Bioinformatics 20:105e114.

14. Parker JS, et al. (2009) Supervised risk predictor of breast cancer based on intrinsicsubtypes. J Clin Oncol 27:1160e1167.

15. Prat A, et al. (2010) Phenotypic and molecular characterization of the claudin-lowintrinsic subtype of breast cancer. Breast Cancer Res 12:R68.

16. Varella-Garcia M, Chen L, Zheng X, Yu L, Dixon D (2006) Karyotypic characteristics ofhuman uterine leiomyoma and myometrial cell lines following telomerase induction.Cancer Genet Cytogenet 170:71e75.

17. Brothman AR, Persons DL, Shaffer LG (2009) Nomenclature evolution: Changes in theISCN from the 2005 to the 2009 edition. Cytogenet Genome Res 127:1e4.

18. Lu YJ, Morris JS, Edwards PA, Shipley J (2000) Evaluation of 24-color multifluor-fluorescence in-situ hybridization (M-FISH) karyotyping by comparison with reversechromosome painting of the human breast cancer cell line T-47D. Chromosome Res 8:127e132.

Haughian et al. www.pnas.org/cgi/content/short/1106509108 2 of 9

Page 3: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

Plastic T47D Cells (Not Passaged In Vivo)

E EWD

CK

5D

AP

IP

R

PR

ERα

CK5

β-actin

E EWD BT20

0.0% 0.0%

B

D

A

E (60d) EWD (60d)

CK

5D

AP

IP

R

0 .74 ± 0.3% 2.4 ± 0.4%*

a) b)

Tumor-derived BT-474Plastic BT-474No Trt

0.0%

a) b)

PR

ERα

CK5

β-actin

Line

2

E-2 switchto EWD

EWD-2 switchto E

E-2 switchto EWD

EWD-2 switchto E

45d 90dTime:

3.6 ±1.1%

C

3.8 ±0.7%

E (60d) EWD (60d)Tumor-derived MCF-7Plastic MCF-7

No Trt

a) b)

ER

C

K5

4.2 ±1.0%

CK

5D

AP

IP

R

ER

C

K5

Fig. S1. Luminobasal cells are rarely present in luminal breast cancer cell lines cultured on plastic. (A) Wild-type T47D cells that had never been xenograftedwere cultured 60 d in steroid-depleted media (EWD) or the same medium supplemented with 10 nM E. (a) Cells were analyzed by ICC for expression of lu-minobasal CK5 (red), and luminal PR (green) and DAPI (blue). EWD failed to expand luminobasal cells as observed in tumor-derived populations. PercentageCK5+ content is indicated. (Scale bars, 50 μm.) (b) Extracts from E and EWD cells were immunoblotted for CK5, estrogen receptor (ER), and progesteronereceptor (PR). Note that EWD increased ER (reversing ligand-dependent ER down-regulation) and deinduced PR, but CK5 was not expressed. Lysate from thetriple-negative BT-20 cell line was included as a positive control for CK5 protein; β-actin is protein loading control. (B and C) Immunocytochemistry of the basalmarker CK5 and luminal markers ER or PR with DAPI counterstain to identify the luminobasal population within cultures of plastic or tumor-derived MCF-7 (B)and BT474 (C) breast cancer cell lines. The tumor-derived cells were grown as tumors in E-treated mice before being returned to in vitro cultures for 60 d insteroid-depleted media without (EWD) or with 1 nM E. *P < 0.01. (Scale bars, 50 μm.) (D) Estrogen regulates expansion or contraction of the luminobasalpopulation in tumor-isolated T47D cells. Line 2 growing in E or EWD was left unchanged or switched to the opposite condition for 45 d or 90 d. Lysates wereimmunoblotted for CK5, ER, and PR to show E-dependent reversal of luminobasal cell expansion.

Haughian et al. www.pnas.org/cgi/content/short/1106509108 3 of 9

Page 4: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

MCF-10A

BJ-3Z

Her

2

BT474

Vim

entin

MDA231

A

GA

TA3

MU

C1

CK

8/18

alp

ha s

moo

th

mus

cle

actin

CK

14C

K5

CK

5C

K5

CK

5C

K5

CK

5FO

XA

1

EWD-3 E-3 EWD-8

B EWD-3 E-3 EWD-8

Fig. S2. Luminobasal cells do not express luminal or myoepithelial marker proteins or Her2 receptor. Cell lines E-3, EWD-3, and EWD-8 derived from mouse-passaged T47D xenografts (Table S1) were grown as 3D colonies in Matrigel. (A) Parrafin sections of 3D colonies were dual stained for the basal marker CK5(red) with various luminal markers, including MUC1, GATA3, CK8/18, and FOXA1 (green) and counterstained with DAPI (blue). (Scale bars, 20 μm.) (B) Paraffin-embedded colonies were sectioned and stained by IHC using the indicated fluors for myoepithelial markers (α-smooth muscle actin, CK14, vimentin) and Her2.Counterstain is DAPI (blue). BJ-3Z (transformed mouse mammary gland stroma), MCF-10A (“normal” human breast), MDA231 (triple-negative breast cancer),and BT474 (Her2+/luminal breast cancer) served as positive controls. (Scale bars, 50 μm.)

Haughian et al. www.pnas.org/cgi/content/short/1106509108 4 of 9

Page 5: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

SK

BR

3A

U56

5H

CC

2185

SU

M18

5PE

MD

AM

B45

3E-3(T47D)

MD

AM

B13

4VI

LY2

MC

F7C

AM

A1

ZR75

BZR

751

MD

AM

B17

5VII

UA

CC

812

SU

M44

PE

BT4

74M

DA

MB

361

BT4

83H

CC

1428

ZR75

3060

0MP

EM

DA

MB

415

HC

C20

2H

CC

1008

SU

M22

5CW

NS

UM

52P

EB

T20

EWD-8

MD

AM

B43

5M

DA

MB

231

HB

L100

SU

M15

9PT

SU

M13

15M

O2

HS

578T

MD

AM

B15

7B

T549

MD

AM

B43

6H

CC

38H

CC

1143

HC

C70

HC

C19

54S

UM

190P

TH

CC

3153

HC

C19

37S

UM

149P

TH

CC

1500

MC

F10A

MC

F12A

MD

AM

B46

8H

CC

1569

HC

C11

87H

CC

2157

-2.0

-1.3

-0.70.0

0.7

1.3

2.0

B

C

EWD-8 MDA231 SUM159 SUM149 SKBR3 MCF7 T47D E-3

A

1298 gene Luminobasal signature

-2.0

-1.3

-0.70.0

0.7

1.3

2.0

EWD-8SUM149 MDA231 SUM159 T47D E-3 SKBR3 MCF7

-1.0

-0.7

-0.30.0

0.3

0.7

1.0

All 16934 genes

Luminal/Her2Triple-Negative

1298 gene Luminobasal signatureLuminal/Her2 Triple-Negative

Luminal/Her2Triple-Negative

D0.80.6

0.40.20.0

-0.2-0.4-0.6

Basal -Like

Claudin-Low

Her-2LuminalA B

***0.80.6

0.40.20.0

-0.2-0.4-0.6

Basal -Like

Claudin-Low

Her-2LuminalA B

*****

-0.8-0.8

NK

I265

Tum

or D

ata

Set

Aver

age

expr

essi

on

Luminobasal Up genes Luminobasal Down genes

NK

I265

Tum

or D

ata

Set

Aver

age

expr

essi

on

Fig. S3. Clustering on the luminobasal signature demonstrates an association with triple-negative breast cancer cell lines, whereas clustering across all genesconfirms the luminal T47D origin of the luminobasal cells. (A) Clustering analysis based on all 16,934 genes present in the arrays. Gene expression wascompared across the pure luminobasal EWD-8, pure luminal E-3, triple-negative (SUM149, SUM159, MDA231), Her2-positive/luminal (SKBR3), and luminal(MCF-7, T47D) cell lines. Note how the pure luminobasal EWD-8 line remains associated with the parental luminal T47D cell line. All lines were analyzed on thesame microarray platform. (B) Clustering analysis of same breast cancer cell lines shown in A based on all 1,298 genes defining the luminobasal signature. Notethat the pure luminobasal EWD-8 line now associates with triple-negative cell lines. (C) Clustering of the E-3 and EWD-8 lines with a large panel of breastcancer cell lines from Neve et al. (1) based on 1,298 genes in the luminobasal signature. (D) Luminobasal signature genes are associated with the triple-negativebasal-like and claudin-low breast cancers in the NKI265 tumor dataset. ANOVA box plots of the average expression values for the (Left) 576 up and (Right) 526down genes present in both the luminobasal signature and NKI265 tumors. The average was calculated across tumors within the indicated subtype. Sixty-sixpercent of tumors fall within the interquartile range (IQR) (colored box), where the bar indicates the median value; whiskers show the range within subtypeand are 1.5*IQR. **P < 0.001, ***P < 0.0001.

1. Neve RM (2006) A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10:515e527.

Haughian et al. www.pnas.org/cgi/content/short/1106509108 5 of 9

Page 6: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

-2.0

-1

.0

0.0

1

.0

2.0

3

.0

-3.0

Luminal A Luminal B Her2 Claudin-low Basal-like

516 tumor data set

NKI 265 tumor data set

A

B

Fig. S4. Luminobasal gene signature clusters luminal and triple-negative breast cancers. The 516 tumor dataset (A) and the NKI265 tumor dataset (B) wereclustered using the 1,298 gene luminobasal signature. Gray boxes indicate regions of tight clustering among tumors identified as triple-negative, being eitherbasal-like or claudin-low subtype.

A Merge

E-3

, Pla

cebo

Tum

orM

CF-

7 Tu

mor

Merge

GA

TA3

CK

5

B

EG

FR C

K5

EG

FR C

K5

GA

TA3

CK

5 Merge

Merge

Fig. S5. Luminobasal cell clusters in luminal tumors express basal markers and contain rare cells with dual luminal and basal features. Parrafin sections oftumors from the E-3, placebo group (A), or MCF-7 tumors (B) were stained by immunofluorescence for the basal markers CK5 (red) and EGFR (green, Upper) orCK5 (red) and the luminal marker GATA-3 (green, Lower). Arrows show rare cells that coexpress both luminal GATA-3 and basal CK5. (Scale bars, 20 μm.)

Haughian et al. www.pnas.org/cgi/content/short/1106509108 6 of 9

Page 7: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

B

0

200

400

600

800

Rel

ativ

e Li

ght U

nits

E EWD E+Tam

E+Fulv

Line 1

no trt

P < 0.01

DAPT CpdE XIX

EWD-8

RBP-Jκ-Luc

Merge

EWD-3 Merge

Merge

A

Notch Ligands

Canonical: Delta-Like 1* ( 6.4 fold) Jagged-1* (13.3 fold) Jagged-2 ( 7.5 fold)

Non-canonical: DNER (19.2 fold)

Notch1 Receptor * ( 9.5 fold)

ADAM10 ( 5.7 fold)

Deltex 3-Like ( 6.3 fold)

MAML-2* (10.3 fold)

Notch Pathway Genes Upregulated in Luminobasal Cells

Transcription

HEY1 (8.4 fold)

Not

ch1

CK

5

C

EWD-8

Not

ch1

CK

5E-3

Not

ch1

CK

5

D

Notch1 ESR1 4.0.

2.0

0.0

-2.0

-4.0

1.0

3.0

-1.0

-3.0

Aver

age

Exp

ress

ion 4.0.

2.0

0.0

-2.0

-4.0

*1.0

3.0

-1.0

-3.0

Basal -Like

Claudin-Low

Her-2 LuminalA B Basal

-Like Claudin-Low

Her-2 LuminalA B

*

P < 0.01

Fig. S6. Activated Notch signaling pathway in luminobasal cells. (A) Schematic based on KEGG pathway diagram (http://www.genome.jp/kegg/pathway.html)of Notch pathway genes up-regulated in luminobasal cells. Numbers indicate the fold change in gene expression between luminobasal (EWD-8 line) andluminal cells (E-3 line) in the Affymetrix platform arrays (SI Materials and Methods). All genes listed are differentially expressed with a false discovery rate of 0.Asterisk (*) denotes Notch genes that were also up-regulated in residual breast disease after letrozole (an aromatase inhibitor) therapy in a microarray dataset(GSE7515) published by Creighton et al. (1). (B) Paraffin sections of Lines EWD-3 and EWD-8 grown as 3D colonies were analyzed for coexpression of Notch1(green) and CK5 (red) with DAPI counterstain (blue). Note that only the core luminobasal cells of EWD-3 express Notch1 and CK5, and E-3 lacks both markers.(Scale bars, 20 μm.) (C) Average gene expression values for Notch1 receptor and ER (ESR1) across 516 breast tumors categorized by subtype (seeMethods). Sixty-six percent of tumors fall within the colored region where the bar indicates the median value; whiskers show range. (D) Notch-dependent transcription isincreased by E deprivation and blocked by GSIs. Left: The pure luminobasal EWD-8 line (Fig. 1E, main text) was transfected with the RBP-Jκ Luc and treated 48 hwith EWD plus DAPT (2 μM), plus CpdE (5 nM), or plus XIX (5 nM), before luciferase quantitation. Right: Xenograft 1 cells were propagated in the absence of E(EWD) or in the presence of E, E plus Tam, or E plus Fulv (Fig. 1D, main text). These lines were transfected with the RBP-Jκ Luc Notch reporter for 48 h beforequantitation of luciferase activity.

1. Creighton CJ, et al. (2009) Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA 106:13820e13825.

Haughian et al. www.pnas.org/cgi/content/short/1106509108 7 of 9

Page 8: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

EG

FR

CK

5 C

K5

FOX

A1

EWD-3

CK

5G

ATA

-3

EWD-3+DAPT EWD-3+CpdE EWD-3+XIX

CK

5N

otch

1

EWD-7E-7 EWD-7 + DAPT EWD-7 + CpdE EWD-7 + XIX

Line

727.5 ± 2.9% 0.44 ± 0.8% 0.47 ± 0.9% 0.7 ± 1.4% 0.9 ± 1.5%

CK

5 E

R

A

C

No Trt DAPT

CK

5 E

R

B

Tumor-derived MCF-7

Fig. S7. Notch inhibition preserves the luminal cell state. (A) Dual ICC staining for the luminobasal marker CK5 (red) and luminal marker ER (green) in tumor-derived MCF-7 cells treated 2 wk with 2 μM DAPT. The percent luminobasal content was determined by flow cytometry (Fig. 4C, main text). (Scale bars, 50 μm.)(B) Cells dissociated from xenograft 7 were propagated >45 d in media with 10 nM E, EWD, or EWD plue 2 μM DAPT, 5 nM CpdE, or 5 nM XIX. Cultures werestained as in A, and numbers indicate the proportion of luminobasal cells. (Scale bars, 50 μm.) (C) Cells from T47D xenograft 3 (Fig. 4D, main text) werepropagated ≈45 d under EWD conditions (EWD-3) or under EWD in the presence of the GSIs DAPT, CpdE, or XIX. Cells were then grown as 3D Matrigel colonies,paraffin embedded, sectioned, and stained by IHC for expression of luminal (GATA-3, FOXA1) and luminobasal (CK5, EGFR) markers as described in SI Materialsand Methods. (Scale bars, 20 μm.)

Table S1. Tumor-derived cell lines: Proportion luminobasal cells in 45- to 60-d culture

Line Xenograft ovx’d mice In vitro treatments E (%) EWD (%)

1 E E, EWD, E + Tam, E + Fulv <1 30–402 E + P E, EWD <1 40–503 E + P E, EWD, EWD + DAPT, EWD + CpdE, lentiviral shRNA vectors <1 30–404 E E, EWD <1 20–305 E + P E, EWD <1 30–406 E + P E, EWD <1 10–207 E + P E, EWD, EWD + DAPT, EWD + CpdE, EWD + XIX <1 20–308 Placebo EWD 90–100 90–100

Parental line is ER+PR+ luminal T47D cells. E, 17β-estradiol; EWD, estrogen withdrawal; Tam, 4-OH tamoxifen; Fulv, fulvestrant(ICI-182780); P, medroxyprogesterone acetate; DAPT, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester; CpdE,compound E [(2S)-2-[(3,5-Difluorophenyl)acetyl]amino-N-[(3S)-1-methyl-2-oxo-5-phenyl-2,3-dihydro-1H-1,4-benzodiazepin-3-yl]propa-namide] ; XIX, (2S,3R)-3-(3,4-Difluorophenyl)-2-(4-fluorophenyl)-4-hydroxy-N-((3S)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diaze-pin-3-yl)-butyramide.

Haughian et al. www.pnas.org/cgi/content/short/1106509108 8 of 9

Page 9: Supporting Information - PNAS€¦ · Supporting Information Haughian et al. 10.1073/pnas.1106509108 SI Materials and Methods Primary Breast Cancers. Paraffin-embedded tissue blocks

Table S2. Reagents and antibodies

Chemical/antibody (ID) Target/application Dose/dilution Source

17β-estradiol ER 1 or 10 nM SigmaMedroxyprogesterone Acetate PR 10 or 100 nM Sigma4-OH Tamoxifen ER 100 nM SigmaICI-182780 (Fulvestrant) ER 100 nM Tocris BioscienceBromodeoxyuridine (BrdU) DNA 0.25 mg/mL SigmaDAPT (GSI IX) Notch, GSI 2 μM Calbiochem/EMD BiosciencesCompound E (GSI XXI) Notch, GSI 5 nM Calbiochem/EMD BiosciencesGSI XIX Notch, GSI 5 nM Calbiochem/EMD BiosciencesCK5 (NCL-L-CK5) IHC, IF, ICC, WB 1:100; 1:1,000 Leica MicrosystemsCK5 (2290-1) IHC, IF, ICC 1:200 EpitomicsPR (Clone 1294) WB 1: 1,000 DAKOPR (SP2) IHC, ICC 1:200 NeomarkersER (Ab-15) WB 1:500 NeomarkersER-α (SP1) IHC, IF 1:100 Neomarkersβ-actin (AC74) WB 1:20,000 SigmaGATA3 (HG3-31) IF 1:200 Santa Cruz BiotechnologyMucin 1 (VU4H5) IF 1:100 Santa Cruz BiotechnologyFOXA1 (ab23738) IF 1:600 AbcamEGFR (06847) IF, IHC 1:400 MilliporeNotch1 (D1E11XP) IF 1:100 Cell SignalingSLUG (C19G7) IF 1:50 Cell SignalingCK8/18 (NCL-L-503) IHC, IF 1:100 Leica MicrosystemsCK14 (RB-9020) IF 1:200 Thermo ScientificSmooth Muscle Actin (E184) IF 1:400 Epitomicsp63 (Ab-4) IF 1:50 EpitomicsHer2 (SP3) IF 1:50 NeomarkersVimentin (NCL-Vim-V9) IF 1:100 Leica MicrosystemsBrdU (347580) IHC 1:50 BD Bioscience

GSI, γ-secretase inhibitor; IHC, immunohistochemistry; IF, immunofluorescence; ICC, immunocytochemistry;WB, Western blot.

Other Supporting Information Files

Dataset S1 (XLSX)

Haughian et al. www.pnas.org/cgi/content/short/1106509108 9 of 9