An integrated genomic approach identifies persistent tumor suppressive effects of transforming growth factor-β in human breast cancer

Misako Sato, Mitsutaka Kadota, Binwu Tang, Howard H Yang, Yu-an Yang, Mengge Shan, Jia Weng, Michael A Welsh, Kathleen C Flanders, Yoshiko Nagano, Aleksandra M Michalowski, Robert J Clifford, Maxwell P Lee, Lalage M Wakefield, Misako Sato, Mitsutaka Kadota, Binwu Tang, Howard H Yang, Yu-an Yang, Mengge Shan, Jia Weng, Michael A Welsh, Kathleen C Flanders, Yoshiko Nagano, Aleksandra M Michalowski, Robert J Clifford, Maxwell P Lee, Lalage M Wakefield

Abstract

Introduction: Transforming growth factor-βs (TGF-βs) play a dual role in breast cancer, with context-dependent tumor-suppressive or pro-oncogenic effects. TGF-β antagonists are showing promise in early-phase clinical oncology trials to neutralize the pro-oncogenic effects. However, there is currently no way to determine whether the tumor-suppressive effects of TGF-β are still active in human breast tumors at the time of surgery and treatment, a situation that could lead to adverse therapeutic responses.

Methods: Using a breast cancer progression model that exemplifies the dual role of TGF-β, promoter-wide chromatin immunoprecipitation and transcriptomic approaches were applied to identify a core set of TGF-β-regulated genes that specifically reflect only the tumor-suppressor arm of the pathway. The clinical significance of this signature and the underlying biology were investigated using bioinformatic analyses in clinical breast cancer datasets, and knockdown validation approaches in tumor xenografts.

Results: TGF-β-driven tumor suppression was highly dependent on Smad3, and Smad3 target genes that were specifically enriched for involvement in tumor suppression were identified. Patterns of Smad3 binding reflected the preexisting active chromatin landscape, and target genes were frequently regulated in opposite directions in vitro and in vivo, highlighting the strong contextuality of TGF-β action. An in vivo-weighted TGF-β/Smad3 tumor-suppressor signature was associated with good outcome in estrogen receptor-positive breast cancer cohorts. TGF-β/Smad3 effects on cell proliferation, differentiation and ephrin signaling contributed to the observed tumor suppression.

Conclusions: Tumor-suppressive effects of TGF-β persist in some breast cancer patients at the time of surgery and affect clinical outcome. Carefully tailored in vitro/in vivo genomic approaches can identify such patients for exclusion from treatment with TGF-β antagonists.

Trial registration: ClinicalTrials.gov NCT01401062.

Figures

Figure 1
Figure 1
Smad3 mediates tumor suppression by TGF-β in the MCF10A model of breast cancer progression. (A) Schematic illustration of the MCF10A-based xenograft model of breast cancer progression. TGF-β has tumor-suppressor activity in M3 cells but this effect is lost in M4 cells and instead TGF-β promotes metastasis. (B) Knockdown of Smad2 and Smad3 protein in M3 and M4 cells was verified by Western blot, quantitated relative to the β-actin loading control and normalized relative to the shGFP condition for each cell line. (C) Relative contributions of Smad2 and Smad3 to the tumor-suppressive effect of TGF-β. Mice were orthotopically implanted with M3 cells or M4 cells, genetically modified to stably express shSmad2, shSmad3 or the control shGFP, and tumor volumes were assessed after seven weeks (M3) or four weeks (M4). Bars indicate median +/−interquartile range; P <0.05 was statistically significant, Mann-Whitney U test. ns, not significant. (D) Kinetics of Smad3 phosphorylation. Western blot of total Smad3 protein and C-terminal phosphorylated Smad3 (Smad3-CP) levels at various time points after TGF-β treatment in M1 to 4 cells. Total Smad3 is shown for t = 0 h. (E) Western blot showing linker phosphorylated Smad3 (Smad3-LP) at 1 hour after treatment with 2 ng/ml TGF-β. TGF-β, transforming growth factor beta.
Figure 2
Figure 2
Identification of Smad3 target genes by ChIP-chip in the MCF10A progression series. (A) The anti-Smad3 antibody recognizes a unique band in wild type and Smad2 null but not Smad3 null IMECs by Western blot. (B) ChIP-QPCR showing ability of Smad3 antibody (αS3) to immunoprecipitate Smad3 bound to the Smad7 promoter in Smad3 wild-type but not Smad3 knockout mouse embryo fibroblasts. CON, isotype-matched control antibody. (C) Time course of Smad3 occupancy at promoters of three previously characterized Smad3 target genes assessed by ChIP-QPCR following treatment of M3 cells with TGF-β. (D) Genome browser view (hg18) of Smad3 binding in the promoter of IFNK in M1 to M4 cells. The signal represents the difference between the TGF-β-treated and untreated conditions. The threshold represents signal intensity corresponding to FDR = 0.15. Black rectangles represent regions of significant Smad3 binding. (E) ChIP-QPCR validation of Smad3 occupancy at the IFNK locus. Results are mean +/−SD (n = 3) normalized to no TGF-β condition. *P <0.05 for enrichment >2-fold. (F) Enrichment of the canonical Smad binding element (SBE) in SBRs. The black line represents 190 high confidence SBRs and the grey line represents 190 random promoter regions with no Smad3 binding. The generic SMAD binding motif is shown. (G) Top 10 enriched transcription factor (TF) matrices within +/−250 bp of the center of 190 high confidence SBRs. (H) Schematic showing co-occurrence for the most enriched TF motifs. Pairwise analysis of each enriched motif was performed using the Fisher’s exact test with Bonferroni correction. The adjusted P values for co-occurrence of pairs of TFs are represented by the connecting lines: P <1e-7 (purple), P <1e-5 (pink), P <1e-2 (black). ChIP, chromatin immunoprecipitation; FDR, false discovery rate; IMEC, immortalized mouse mammary epithelial cells; QPCR, quantitative polymerase chain reaction; SBR, Smad binding region; TGF-β, transforming growth factor beta.
Figure 3
Figure 3
Smad3 binding differs widely across the progression series. (A) Petal plot showing the overlap in Smad3 target genes between the different cell lines. The genes involved are given in Supporting Information Table S1 in Additional file 4. (B) Representative genes with distinct Smad3 occupancy patterns as confirmed by ChIP-QPCR. Closed circles indicate TGF-β-induced Smad3 occupancy. (C) DNA methylation status at promoter regions of each target gene in (B) as determined by MeDIP-QPCR. Relative enrichment in the bound (MeDIP) vs. unbound fractions is shown. PPIA and MyoD were controls for unmethylated and highly methylated DNAs respectively. (D) QPCR quantitation of target enrichment following ChIP using anti-H3AcK9/14 to identify active chromatin. Enrichment at the SBR was calculated relative to input DNA. PPIA and MyoD were controls for active and inactive promoters respectively. Active chromatin has an enrichment value >1.00 (indicated by threshold line). ChIP, chromatin immunoprecipitation; H3AcK9/14, histone H3 acetylated on lysine 9 or 14; MeDIP, methylated DNA immunoprecipitation; QPCR, quantitative polymerase chain reaction; SBR, Smad binding region; TGF-β, transforming growth factor beta.
Figure 4
Figure 4
Strategy for integration of ChIP-chip and gene expression datasets to generate a core TGF-β/Smad3 tumor suppressor signature. The experimental strategy for identification of the TGF-β/Smad3 tumor suppressor signature (TSTSS) is shown. ChIP, chromatin immunoprecipitation; TGF-β, transforming growth factor beta.
Figure 5
Figure 5
Generation of core TGF-β/Smad3 tumor suppressor signature. (A) Unsupervised hierarchical clustering of differentially expressed Smad3 target genes in M3 and M4 cells treated with TGF-β in vitro for 6 hours. The 38 genes induced by TGF-β in vitro in M3 only were taken forward for further analysis. (B) RTQ-PCR validation of the 26 genes out of the original 38 genes that microarray analysis showed to be also regulated by TGF-β in M3 tumors in vivo. M3 tumors transduced with the dnTβRII represents the low TGF-β signal condition in vivo, while M3 tumors transduced with control lentivirus represents the high TGF-β signal condition in vivo. Expression was normalized to the low signal condition for each gene. Results are the mean +/−SEM for three tumors/experimental group. The difference between the high and low TGF-β signaling conditions is statistically significant (P <0.05; unpaired t test) for all genes shown. Note the six genes, marked by arrows, which are downregulated by TGF-β in vivo whereas they were upregulated in vitro. (C) Smad3 dependence of TGF-β regulation of select target genes in vivo. Further RT-QPCR quantitation was performed for six representative genes under the following conditions: (i) M3 cells treated with 5 ng/ml TGF-β (high TGF-β signal condition) or vehicle (low signal condition) in vitro; (ii) M3 tumors in vivo following transduction with a dnTβRII to block all TGF-β responses (low signal condition) or LacZ control lentivirus (high signal condition); (iii) M3 tumors in vivo following transduction with shSmad3 to block Smad3-mediated responses (low signal condition) or shGFP control lentivirus (high signal condition). Results are mean +/−SEM for three to six independent samples/group, normalized to low signaling condition. *statistically significant (P <0.05) for high vs. low signaling condition, unpaired t test. dnTβRII, dominant-negative type II TGF-β receptor; TGF-β, transforming growth factor beta.
Figure 6
Figure 6
Meta-analyses correlating TGF-β/Smad3 target genes with outcome in human breast cancer datasets. Kaplan-Meier analyses were performed using the online GOBO tool to assess the association of the TGF-β-regulated gene sets with distant metastasis-free survival (DMFS) in meta-analyses across multiple breast cancer cohorts (1,379 tumors from eight datasets). Patient datasets were dichotomized to higher than median expression (black) or lower than median expression (grey) of the gene set. P values were determined by the log-rank test. (A,B) Kaplan-Meier plots for survival of all patients in the GOBO datasets, using the set of TGF-β/Smad3 target genes that were uniquely regulated in M3 (the TSTSS). This gene set was designed to be enriched in genes involved in tumor suppression. Weighting (positive or negative) of individual target genes is based on the directionality of TGF-β-regulated gene expression observed in M3 tumors in vivo(A) or in M3 cells in vitro(B) as indicated. (C,D) Correlation of the TSTSS (using the in vivo directional weighting) with DMFS in ER+ (n = 856) (C), and ER- (n = 320) (D) patient subsets of the GOBO cohorts. ns, not significant. ER, estrogen receptor; GOBO, gene expression-based outcome for breast cancer online; TGF-β, transforming growth factor beta; TSS, transcriptional start site; TSTSS, TGF-β/Smad3 tumor suppressor signature.
Figure 7
Figure 7
Loss of TGF-β signaling leads to reduced tumor differentiation. (A) The TSTSS is weakly anticorrelated with proliferation, as assessed using a metaPCNA index, in ER+ but not ER- breast cancers (TCGA cohort). (B) Expression of the TSTSS inversely correlates with tumor grade in ER+ breast cancer (n = 904 patients), as assessed using GOBO tool. (C) Correlation between the TSTSS and luminal differentiation, as assessed using a meta-differentiation index, in ER+ and ER- tumors in the TCGA cohort. (D) Immunohistochemical staining of cytokeratin 8 (CK8) and ER-α in primary tumors from M3 cells expressing shGFP, shSmad2 or shSmad3. Scale bars represent 100 μm. (E) Quantitation of % area occupied by structures with a differentiated glandular histology in M3 tumors expressing shGFP (control), shSmad2 or ShSmad3. Results are mean +/−SEM for five tumors/group. (F) Quantitation of CK8 and ER staining was performed using Image-Pro Plus software. Each datapoint represents the mean of five fields/tumor, and results are shown as mean +/−SEM for five tumors/group. *P <0.05 for one-way ANOVA with Dunnett’s multiple comparison test; ns, not significant; hpf, high power field. ER, estrogen receptor; GOBO, gene expression-based outcome for breast cancer online; TGF-β, transforming growth factor beta; TSTSS, TGF-β/Smad3 tumor suppressor signature.
Figure 8
Figure 8
Ephrin signaling contributes to the tumor suppressive effects of TGF-β in ER+ breast cancer. (A) Pathway enrichment in the TSTSS assessed by Ingenuity Pathway Analysis. Fisher’s exact test with Benjamini-Hochberg (B-H) correction. The dotted line represents the P <0.05 significance threshold. (B) Correlation of TSTSS with meta-EFNA index in ER+ tumors of the TCGA cohort. (C) Association of meta-EFNA index with distant metastasis-free survival (DMFS) in ER+ breast cancer (n = 856 patients) using the GOBO tool. (D) Smad3 ChIP-QPCR at the EFNA1 locus in M3 and M4 cells. (E) ChIP-QPCR for H3AcK9/14 at the EFNA1 locus to identify active chromatin. (F) Time course of EFNA1 mRNA induction by TGF-β (2 ng/ml) in M3 and M4 cells. Results are mean +/−SEM of three determinations. *P <0.05. (G) Western blot of effect of TGF-β treatment of M3 cells on Ephrin A1 (EFNA1) expression and oncogenic signaling through phosphorylation of the EphA2 receptor on S897. EFNA-Fc was used as a positive control for activation of the EphA2 signaling path. (H) Western blot showing knockdown of EFNA1 in M3 cells. (I) Knockdown of EFNA enhances tumorigenesis in M3 cells. n = 8 to 10 mice/group. *P >0.05 one-way ANOVA, Tukey’s multiple comparison test. ChIP, chromatin immunoprecipitation; ER, estrogen receptor; GOBO, Gene expression-based Outcome for Breast cancer Online; H3AcK9/14, histone H3 acetylated on lysine 9 or 14; QPCR, quantitative polymerase chain reaction; TGF-β, transforming growth factor beta; TSTSS, TGF-β/Smad3 tumor suppressor signature.

References

    1. Reis-Filho JS, Weigelt B, Fumagalli D, Sotiriou C. Molecular profiling: moving away from tumor philately. Sci Transl Med. 2010;2:ps43.
    1. Gatza ML, Lucas JE, Barry WT, Kim JW, Wang Q, Crawford MD, Datto MB, Kelley M, Mathey-Prevot B, Potti A, Nevins JR. A pathway-based classification of human breast cancer. Proc Natl Acad Sci U S A. 2010;107:6994–6999.
    1. Massague J. TGFβ in cancer. Cell. 2008;134:215–230.
    1. Ikushima H, Miyazono K. TGFβ signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–424.
    1. Pardali K, Moustakas A. Actions of TGF-β as tumor suppressor and pro-metastatic factor in human cancer. Biochim Biophys Acta. 2007;1775:21–62.
    1. Akhurst RJ, Hata A. Targeting the TGFβ signalling pathway in disease. Nat Rev Drug Discov. 2012;11:790–811.
    1. Glick AB, Weinberg WC, Wu IH, Quan W, Yuspa SH. Transforming growth factor β1 suppresses genomic instability independent of a G1 arrest, p53, and Rb. Cancer Res. 1996;56:3645–3650.
    1. Tremain R, Marko M, Kinnimulki V, Ueno H, Bottinger E, Glick A. Defects in TGF-β signaling overcome senescence of mouse keratinocytes expressing v-Ha-ras. Oncogene. 2000;19:1698–1709.
    1. Guasch G, Schober M, Pasolli HA, Conn EB, Polak L, Fuchs E. Loss of TGFβ signaling destabilizes homeostasis and promotes squamous cell carcinomas in stratified epithelia. Cancer Cell. 2007;12:313–327.
    1. Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komuro A, Kano MR, Sugimoto K, Miyazono K. Autophagy is activated by TGF-β and potentiates TGF-β-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 2009;69:8844–8852.
    1. Bierie B, Chung CH, Parker JS, Stover DG, Cheng N, Chytil A, Aakre M, Shyr Y, Moses HL. Abrogation of TGF-β signaling enhances chemokine production and correlates with prognosis in human breast cancer. J Clin Invest. 2009;119:1571–1582.
    1. Novitskiy SV, Pickup MW, Gorska AE, Owens P, Chytil A, Aakre M, Wu H, Shyr Y, Moses HL. TGF-β receptor II loss promotes mammary carcinoma progression by Th17 dependent mechanisms. Cancer Discov. 2011;1:430–441.
    1. Cheng N, Bhowmick NA, Chytil A, Gorksa AE, Brown KA, Muraoka R, Arteaga CL, Neilson EG, Hayward SW, Moses HL. Loss of TGF-β type II receptor in fibroblasts promotes mammary carcinoma growth and invasion through upregulation of TGF-alpha-, MSP- and HGF-mediated signaling networks. Oncogene. 2005;24:5053–5068.
    1. Lehmann K, Janda E, Pierreux CE, Rytomaa M, Schulze A, McMahon M, Hill CS, Beug H, Downward J. Raf induces TGFβ production while blocking its apoptotic but not invasive responses: a mechanism leading to increased malignancy in epithelial cells. Genes Dev. 2000;14:2610–2622.
    1. Oft M, Peli J, Rudaz C, Schwarz H, Beug H, Reichmann E. TGF-β1 and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells. Genes Dev. 1996;10:2462–2477.
    1. Seton-Rogers SE, Lu Y, Hines LM, Koundinya M, LaBaer J, Muthuswamy SK, Brugge JS. Cooperation of the ErbB2 receptor and transforming growth factor β in induction of migration and invasion in mammary epithelial cells. Proc Natl Acad Sci U S A. 2004;101:1257–1262.
    1. Scheel C, Eaton EN, Li SH, Chaffer CL, Reinhardt F, Kah KJ, Bell G, Guo W, Rubin J, Richardson AL, Weinberg RA. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell. 2011;145:926–940.
    1. Bruna A, Greenwood W, Le QJ, Teschendorff A, Miranda-Saavedra D, Rueda OM, Sandoval JL, Vidakovic AT, Saadi A, Pharoah P, Singl J, Caldas C. TGFβ induces the formation of tumour-initiating cells in claudin-low breast cancer. Nat Commun. 2012;3:1055.
    1. Gorelik L, Flavell RA. Immune-mediated eradication of tumors through the blockade of transforming growth factor-β signaling in T cells. Nat Med. 2001;7:1118–1122.
    1. Pardali E, ten Dijke P. Transforming growth factor-β signaling and tumor angiogenesis. Front Biosci. 2009;14:4848–4861.
    1. Lewis MP, Lygoe KA, Nystrom ML, Anderson WP, Speight PM, Marshall JF, Thomas GJ. Tumour-derived TGF-β1 modulates myofibroblast differentiation and promotes HGF/SF-dependent invasion of squamous carcinoma cells. Br J Cancer. 2004;90:822–832.
    1. Moses H, Barcellos-Hoff MH. TGF-β biology in mammary development and breast cancer. Cold Spring Harb Perspect Biol. 2011;3:a003277.
    1. Bierie B, Moses HL. Gain or loss of TGFβ signaling in mammary carcinoma cells can promote metastasis. Cell Cycle. 2009;8:3319–3327.
    1. Siegel PM, Shu W, Cardiff RD, Muller WJ, Massague J. Transforming growth factor β signaling impairs Neu-induced mammary tumorigenesis while promoting pulmonary metastasis. Proc Natl Acad Sci U S A. 2003;100:8430–8435.
    1. Tang B, Vu M, Booker T, Santner SJ, Miller FR, Anver MR, Wakefield LM. TGF-β switches from tumor suppressor to prometastatic factor in a model of breast cancer progression. J Clin Invest. 2003;112:1116–1124.
    1. Bierie B, Stover DG, Abel TW, Chytil A, Gorska AE, Aakre M, Forrester E, Yang L, Wagner KU, Moses HL. Transforming growth factor-β regulates mammary carcinoma cell survival and interaction with the adjacent microenvironment. Cancer Res. 2008;68:1809–1819.
    1. Forrester E, Chytil A, Bierie B, Aakre M, Gorska AE, Sharif-Afshar AR, Muller WJ, Moses HL. Effect of conditional knockout of the type II TGF-β receptor gene in mammary epithelia on mammary gland development and polyomavirus middle T antigen induced tumor formation and metastasis. Cancer Res. 2005;65:2296–2302.
    1. Kohn EA, Yang YA, Du Z, Nagano Y, Van Schyndle CM, Herrmann MA, Heldman M, Chen JQ, Stuelten CH, Flanders KC, Wakefield LM. Biological responses to TGF-β in the mammary epithelium show a complex dependency on Smad3 gene dosage with important implications for tumor progression. Mol Cancer Res. 2012;10:1389–1399.
    1. Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J, Silliman N, Szabo S, Dezso Z, Ustyanksky V, Nikoskaya T, Nikolsky Y, Karchin R, Wilson PA, Kaminker JS, Zhang Z, Croshaw R, Willis J, Dawson D, Shipitsin M, Willson JK, Sukumar S, Polyak K, Park BH, Pethiyagoda CL, Pant PV. et al.The genomic landscapes of human breast and colorectal cancers. Science. 2007;318:1108–1113.
    1. Brown KA, Aakre ME, Gorska AE, Price JO, Eltom SE, Pietenpol JA, Moses HL. Induction by transforming growth factor-β1 of epithelial to mesenchymal transition is a rare event in vitro. Breast Cancer Res. 2004;6:R215–R231.
    1. Gobbi H, Dupont WD, Simpson JF, Plummer WD Jr, Schuyler PA, Olson SJ, Arteaga CL, Page DL. Transforming growth factor-β and breast cancer risk in women with mammary epithelial hyperplasia. J Natl Cancer Inst. 1999;91:2096–2101.
    1. Wang SE, Xiang B, Guix M, Olivares MG, Parker J, Chung CH, Pandiella A, Arteaga CL. Transforming growth factor β engages TACE and ErbB3 to activate phosphatidylinositol-3 kinase/Akt in ErbB2-overexpressing breast cancer and desensitizes cells to trastuzumab. Mol Cell Biol. 2008;28:5605–5620.
    1. Coulouarn C, Factor VM, Thorgeirsson SS. Transforming growth factor-β gene expression signature in mouse hepatocytes predicts clinical outcome in human cancer. Hepatology. 2008;47:2059–2067.
    1. Padua D, Zhang XH, Wang Q, Nadal C, Gerald WL, Gomis RR, Massague J. TGFβ primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 2008;133:66–77.
    1. Xu XL, Kapoun AM. Heterogeneous activation of the TGFβ pathway in glioblastomas identified by gene expression-based classification using TGFβ-responsive genes. J Transl Med. 2009;7:12.
    1. Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N, Yao J, Nikolskaya T, Serebryiskaya T, Beroukhim R, Hu M, Halushka MK, Sukumar S, Parker LM, Anderson KS, Harris LN, Garber JE, Richardson AL, Schnitt SJ, Nikolsky Y, Gelman RS, Polyak K. Molecular definition of breast tumor heterogeneity. Cancer Cell. 2007;11:259–273.
    1. Santner SJ, Dawson PJ, Tait L, Soule HD, Eliason J, Mohamed AN, Wolman SR, Heppner GH, Miller FR. Malignant MCF10CA1 cell lines derived from premalignant human breast epithelial MCF10AT cells. Breast Cancer Res Treat. 2001;65:101–110.
    1. Strickland LB, Dawson PJ, Santner SJ, Miller FR. Progression of premalignant MCF10AT generates heterogeneous malignant variants with characteristic histologic types and immunohistochemical markers. Breast Cancer Res Treat. 2000;64:235–240.
    1. Soule HD, Maloney TM, Wolman SR, Peterson WD Jr, Brenz R, McGrath CM, Russo J, Pauley RJ, Jones RF, Brooks SC. Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res. 1990;50:6075–6086.
    1. Visscher DW, Nanjia-Makker P, Heppner G, Shekhar PV. Tamoxifen suppresses histologic progression to atypia and DCIS in MCFIOAT xenografts, a model of early human breast cancer. Breast Cancer Res Treat. 2001;65:41–47.
    1. Shekhar PV, Chen ML, Werdell J, Heppner GH, Miller FR, Christman JK. Transcriptional activation of functional endogenous estrogen receptor gene expression in MCF10AT cells: a model for early breast cancer. Int J Oncol. 1998;13:907–915.
    1. Shekhar MP, Werdell J, Santner SJ, Pauley RJ, Tait L. Breast stroma plays a dominant regulatory role in breast epithelial growth and differentiation: implications for tumor development and progression. Cancer Res. 2001;61:1320–1326.
    1. Shekhar MP, Nangia-Makker P, Wolman SR, Tait L, Heppner GH, Visscher DW. Direct action of estrogen on sequence of progression of human preneoplastic breast disease. Am J Pathol. 1998;152:1129–1132.
    1. Kadota M, Yang HH, Gomez B, Sato M, Clifford RJ, Meerzaman D, Dunn BK, Wakefield LM, Lee MP. Delineating genetic alterations for tumor progression in the MCF10A series of breast cancer cell lines. PLoS One. 2010;5:e9201.
    1. Michelucci A, Di CC, Lami A, Collecchi P, Caligo A, Decarli N, Leopizzi M, Aretini P, Bertacca G, Porta RP, Ricci S, Della Rocca C, Stanta G, Bevilacqua G, Cazzana A. PIK3CA in breast carcinoma: a mutational analysis of sporadic and hereditary cases. Diagn Mol Pathol. 2009;18:200–205.
    1. Bild AH, Parker JS, Gustafson AM, Acharya CR, Hoadley KA, Anders C, Marcom PK, Carey LA, Potti A, Nevins JR, Perou CM. An integration of complementary strategies for gene-expression analysis to reveal novel therapeutic opportunities for breast cancer. Breast Cancer Res. 2009;11:R55.
    1. Kohn EA, Du Z, Sato M, Van Schyndle CM, Welsh MA, Yang YA, Stuelten CH, Tang B, Ju W, Bottinger EP, Wakefield LM. A novel approach for the generation of genetically modified mammary epithelial cell cultures yields new insights into TGFβ signaling in the mammary gland. Breast Cancer Res. 2010;12:R83.
    1. Liang G, Lin JC, Wei V, Yoo C, Cheng JC, Nguyen CT, Weisenberger DJ, Egger G, Takai D, Gonzales FA, Jones PA. Distinct localization of histone H3 acetylation and H3-K4 methylation to the transcription start sites in the human genome. Proc Natl Acad Sci U S A. 2004;101:7357–7362.
    1. Karmodiya K, Krebs AR, Oulad-Abdelghani M, Kimura H, Tora L. H3K9 and H3K14 acetylation co-occur at many gene regulatory elements, while H3K14ac marks a subset of inactive inducible promoters in mouse embryonic stem cells. BMC Genomics. 2012;13:424.
    1. Kadota M, Yang HH, Hu N, Wang C, Hu Y, Taylor PR, Buetow KH, Lee MP. Allele-specific chromatin immunoprecipitation studies show genetic influence on chromatin state in human genome. PLoS Genet. 2007;3:e81.
    1. Nicol JW, Helt GA, Blanchard SG Jr, Raja A, Loraine AE. The Integrated Genome Browser: free software for distribution and exploration of genome-scale datasets. Bioinformatics. 2009;25:2730–2731.
    1. The Galaxy Project. [ ]
    1. Silverman BW. Density Estimation. London, UK: Chapman and Hall; 1986.
    1. The R Project for Statistical Computing. [ ]
    1. Genomatix Software Tools. [ ]
    1. Ringner M, Fredlund E, Hakkinen J, Borg A, Staaf J. GOBO: gene expression-based outcome for breast cancer online. PLoS One. 2011;6:e17911.
    1. GOBO: gene expression-based outcome for breast cancer online. [ ]
    1. van de Vijver MJ, He YD, van’t Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsam D, Witteveen A, Glas A, Delahaye L, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999–2009.
    1. Broad Institute Genome Data Analysis Center. [ ]
    1. Venet D, Dumont JE, Detours V. Most random gene expression signatures are significantly associated with breast cancer outcome. PLoS Comput Biol. 2011;7:e1002240.
    1. Ge X, Yamamoto S, Tsutsumi S, Midorikawa Y, Ihara S, Wang SM, Aburatani H. Interpreting expression profiles of cancers by genome-wide survey of breadth of expression in normal tissues. Genomics. 2005;86:127–141.
    1. Lim E, Wu D, Pa B, Bouras T, Asselin-Labat ML, Vaillant F, Yagita H, Lindeman GJ, Smyth GK, Visvader JE. Transcriptome analyses of mouse and human mammary cell subpopulations reveal multiple conserved genes and pathways. Breast Cancer Res. 2010;12:R21.
    1. Kim J, Villadsen R, Sorlie T, Fogh L, Gronlund SZ, Fridriksdottir AJ, Kuhn I, Rank F, Wielenga VT, Solvang H, Edwards PA, Borresen-Dale AL, Ronnov-Jessen L, Bissell MJ, Petersen OW. Tumor initiating but differentiated luminal-like breast cancer cells are highly invasive in the absence of basal-like activity. Proc Natl Acad Sci U S A. 2012;109:6124–6129.
    1. Gao H, Chakraborty G, Lee-Lim AP, Mo Q, Decker M, Vonica A, Shen R, Brogi E, Brivanlou AH, Giancotti FG. The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites. Cell. 2012;150:764–779.
    1. Tang B, Yoo N, Vu M, Mamura M, Nam JS, Ooshima A, Du Z, Desprez PY, Anver MR, Michalowska AM, Shih J, Parks WT, Wakefield LM. Transforming growth factor-β can suppress tumorigenesis through effects on the putative cancer stem or early progenitor cell and committed progeny in a breast cancer xenograft model. Cancer Res. 2007;67:8643–8652.
    1. Schmierer B, Hill CS. TGFβ-SMAD signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol. 2007;8:970–982.
    1. Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-β signaling. J Cell Biochem. 2007;101:9–33.
    1. Matsuzaki K, Okazaki K. Transforming growth factor-β during carcinogenesis: the shift from epithelial to mesenchymal signaling. J Gastroenterol. 2006;41:295–303.
    1. Zhu S, Wang W, Clarke DC, Liu X. Activation of Mps1 promotes transforming growth factor-β-independent Smad signaling. J Biol Chem. 2007;282:18327–18338.
    1. Lee BH, Chen W, Stippec S, Cobb MH. Biological cross-talk between WNK1 and the transforming growth factor β-Smad signaling pathway. J Biol Chem. 2007;282:17985–17996.
    1. Seong HA, Jung H, Ha H. Murine protein serine/threonine kinase 38 stimulates TGF-β signaling in a kinase-dependent manner via direct phosphorylation of Smad proteins. J Biol Chem. 2010;285:30959–30970.
    1. Chung AC, Zhang H, Kong YZ, Tan JJ, Huang XR, Kopp JB, Lan HY. Advanced glycation end-products induce tubular CTGF via TGF-β-independent Smad3 signaling. J Am Soc Nephrol. 2010;21:249–260.
    1. Koinuma D, Tsutsumi S, Kamimura N, Taniguchi H, Miyazawa K, Sunamura M, Imamura T, Miyazono K, Aburatani H. Chromatin immunoprecipitation on microarray analysis of Smad2/3 binding sites reveals roles of ETS1 and TFAP2A in transforming growth factor β signaling. Mol Cell Biol. 2009;29:172–186.
    1. Mizutani A, Koinuma D, Tsutsumi S, Kamimura N, Morikawa M, Suzuki HI, Imamura T, Miyazono K, Aburatani H. Cell-type specific target selection by combinatorial binding of Smad2/3 and hepatocyte nuclear factor 4{alpha} in HepG2 cells. J Biol Chem. 2011;286:29848–29860.
    1. Zhang Y, Handley D, Kaplan T, Yu H, Bais AS, Richards T, Pandit KV, Zeng Q, Benos PV, Friedman N, Eickelberg O, Kaminski N. High throughput determination of TGFβ1/SMAD3 targets in A549 lung epithelial cells. PLoS One. 2011;6:e20319.
    1. Zhang Y, Feng XH, Derynck R. Smad3 and Smad4 cooperate with c-Jun/c-Fos to mediate TGF-β-induced transcription. Nature. 1998;394:909–913.
    1. Sundqvist A, Zieba A, Vasilaki E, Herrera HC, Soderberg O, Koinuma D, Miyazono K, Heldin CH, Landegren U, ten Dijke P, van Dam H. Specific interactions between Smad proteins and AP-1 components determine TGFβ-induced breast cancer cell invasion. Oncogene. 2013;32:3606–3615.
    1. The Cancer Genome Atlas. [ ]
    1. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, Graf S, Ha G, Haffari G, Bashashati A, Russell R, McKinney S, Langerod A, Green A, Provenzano E, Wishart G, Pinder S, Watson P, Markowetz F, Murphy L, Ellis I, Purushotham A, Borresen-Dale AL, Brenton JD, Tavare S, Caldas C. et al.The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352.
    1. Mosley JD, Keri RA. Cell cycle correlated genes dictate the prognostic power of breast cancer gene lists. BMC Med Genomics. 2008;1:11.
    1. Rakha EA, Reis-Filho JS, Baehner F, Dabbs DJ, Decker T, Eusebi V, Fox SB, Ichihara S, Jacquemier J, Lakhani SR, Palacios J, Richardson AL, Schnitt SJ, Schmitt FC, Tan PH, Tse GM, Badve S, Ellis IO. Breast cancer prognostic classification in the molecular era: the role of histological grade. Breast Cancer Res. 2010;12:207.
    1. Pasquale EB. Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat Rev Cancer. 2010;10:165–180.
    1. Vaught D, Brantley-Sieders DM, Chen J. Eph receptors in breast cancer: roles in tumor promotion and tumor suppression. Breast Cancer Res. 2008;10:217.
    1. Miao H, Wang B. EphA receptor signaling-complexity and emerging themes. Semin Cell Dev Biol. 2012;23:16–25.
    1. Kennedy BA, Deatherage DE, Gu F, Tang B, Chan MW, Nephew KP, Huang TH, Jin VX. ChIP-seq defined genome-wide map of TGFβ/SMAD4 targets: implications with clinical outcome of ovarian cancer. PLoS One. 2011;6:e22606.
    1. Mullen AC, Orlando DA, Newman JJ, Loven J, Kumar RM, Bilodeau S, Reddy J, Guenther MG, Dekoter RP, Young RA. Master transcription factors determine cell-type-specific responses to TGF-β signaling. Cell. 2011;147:565–576.
    1. Kendrick H, Regan JL, Magnay FA, Grigoriadis A, Mitsopoulos C, Zvelebil M, Smalley MJ. Transcriptome analysis of mammary epithelial subpopulations identifies novel determinants of lineage commitment and cell fate. BMC Genomics. 2008;9:591.
    1. Ewan KB, Oketch-Rabah HA, Ravani SA, Shyamala G, Moses HL, Barcellos-Hoff MH. Proliferation of estrogen receptor-alpha-positive mammary epithelial cells is restrained by transforming growth factor-β1 in adult mice. Am J Pathol. 2005;167:409–417.
    1. Noblitt LW, Bangari DS, Shukla S, Knapp DW, Mohammed S, Kinch MS, Mittal SK. Decreased tumorigenic potential of EphA2-overexpressing breast cancer cells following treatment with adenoviral vectors that express EphrinA1. Cancer Gene Ther. 2004;11:757–766.
    1. Brantley-Sieders DM, Zhuang G, Hicks D, Fang WB, Hwang Y, Cates JM, Coffman K, Jackson D, Bruckheimer E, Muraoka-Cook RS, Chen J. The receptor tyrosine kinase EphA2 promotes mammary adenocarcinoma tumorigenesis and metastatic progression in mice by amplifying ErbB2 signaling. J Clin Invest. 2008;118:64–78.
    1. Zelinski DP, Zantek ND, Stewart JC, Irizarry AR, Kinch MS. EphA2 overexpression causes tumorigenesis of mammary epithelial cells. Cancer Res. 2001;61:2301–2306.
    1. Brantley-Sieders DM, Fang WB, Hwang Y, Hicks D, Chen J. Ephrin-A1 facilitates mammary tumor metastasis through an angiogenesis-dependent mechanism mediated by EphA receptor and vascular endothelial growth factor in mice. Cancer Res. 2006;66:10315–10324.

Source: PubMed

3
Abonner