Skip to main content

Advertisement

Log in

Noncanonical TGF-β Signaling During Mammary Tumorigenesis

  • Published:
Journal of Mammary Gland Biology and Neoplasia Aims and scope Submit manuscript

Abstract

Breast cancer is a heterogeneous disease comprised of at least five major tumor subtypes that coalesce as the second leading cause of cancer death in women in the United States. Although metastasis clearly represents the most lethal characteristic of breast cancer, our understanding of the molecular mechanisms that govern this event remains inadequate. Clinically, ~30% of breast cancer patients diagnosed with early-stage disease undergo metastatic progression, an event that (a) severely limits treatment options, (b) typically results in chemoresistance and low response rates, and (c) greatly contributes to aggressive relapses and dismal survival rates. Transforming growth factor-β (TGF-β) is a pleiotropic cytokine that regulates all phases of postnatal mammary gland development, including branching morphogenesis, lactation, and involution. TGF-β also plays a prominent role in suppressing mammary tumorigenesis by preventing mammary epithelial cell (MEC) proliferation, or by inducing MEC apoptosis. Genetic and epigenetic events that transpire during mammary tumorigenesis conspire to circumvent the tumor suppressing activities of TGF-β, thereby permitting late-stage breast cancer cells to acquire invasive and metastatic phenotypes in response to TGF-β. Metastatic progression stimulated by TGF-β also relies on its ability to induce epithelial-mesenchymal transition (EMT) and the expansion of chemoresistant breast cancer stem cells. Precisely how this metamorphosis in TGF-β function comes about remains incompletely understood; however, recent findings indicate that the initiation of oncogenic TGF-β activity is contingent upon imbalances between its canonical and noncanonical signaling systems. Here we review the molecular and cellular contributions of noncanonical TGF-β effectors to mammary tumorigenesis and metastatic progression.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3

Similar content being viewed by others

Abbreviations

AKT/PKB:

protein kinase B

BMDC:

bone marrow-derived cell

CSC:

cancer stem cell

ECM:

extracellular matrix

EGF:

epidermal growth factor

EMT:

epithelial-mesenchymal transition

ERK:

extracellular signal-regulated kinase

FAK:

focal adhesion kinase

HGF:

hepatocyte growth factor

hnRNP E1:

heterogeneous nuclear ribronucleoprotein E1

ILEI:

interleukin-like EMT inducer

JNK:

c-Jun N-terminal kinase

MAP kinase:

mitogen-activated protein kinase

MEC:

mammary epithelial cell

MET:

mesenchymal-epithelial transition

miR:

microRNA

MMP:

matrix metalloproteinase

MSP:

macrophage-stimulating protein

MTA3:

metastasis associated protein 3

mTOR:

mammalian target of rapamycin

NF-κB:

nuclear factor-κB

PAI:

plasminogen activator inhibitor

PDGF:

platelet-derived growth factor

PI3K:

phosphoinositide-3-kinase

TβR-I:

TGF-β type I receptor

TβR-II:

TGF-β type II receptor

TβR-III:

TGF-β type III receptor

TGF-β:

transforming growth factor-β

TRAF6:

TNF receptor-associated factor 6

uPA:

urokinase plasminogen activator

uPAR:

uPA receptor

ZO-1:

zonula occluden-1

References

  1. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60(5):277–300.

    Article  PubMed  Google Scholar 

  2. Nguyen DX, Bos PD, Massague J. Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer. 2009;9(4):274–84.

    Article  PubMed  CAS  Google Scholar 

  3. Nguyen DX, Massague J. Genetic determinants of cancer metastasis. Nat Rev Genet. 2007;8(5):341–52.

    Article  PubMed  CAS  Google Scholar 

  4. Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14(6):818–29.

    Article  PubMed  CAS  Google Scholar 

  5. Taylor MA, Parvani JG, Schiemann WP. The pathophysiology of epithelial-mesenchymal transition induced by transforming growth factor-β in normal and malignant mammary epithelial cells. J Mammary Gland Biol Neoplasia. 2010;15(2):169–90.

    Article  PubMed  Google Scholar 

  6. Altekruse SF, Kosary CL, Krapcho M, Neyman N, Aminous R, Waldron W, et al. SEER Cancer Statistics Review 1975–1007. National Cancer Institute. 2010.

  7. Kennecke H, Yerushalmi R, Woods R, Cheang MC, Voduc D, Speers CH, et al. Metastatic behavior of breast cancer subtypes. J Clin Oncol. 2010;28(20):3271–7.

    Article  PubMed  Google Scholar 

  8. Chiang AC, Massague J. Molecular basis of metastasis. N Engl J Med. 2008;359(26):2814–23.

    Article  PubMed  CAS  Google Scholar 

  9. Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2(8):563–72.

    Article  PubMed  CAS  Google Scholar 

  10. Weinstat-Saslow D, Steeg PS. Angiogenesis and colonization in the tumor metastatic process: basic and applied advances. FASEB J. 1994;8(6):401–7.

    PubMed  CAS  Google Scholar 

  11. Scheel C, Onder T, Karnoub A, Weinberg RA. Adaptation versus selection: the origins of metastatic behavior. Cancer Res. 2007;67(24):11476–9.

    Article  PubMed  CAS  Google Scholar 

  12. Talmadge JE. Clonal selection of metastasis within the life history of a tumor. Cancer Res. 2007;67(24):11471–5.

    Article  PubMed  CAS  Google Scholar 

  13. Talmadge JE, Fidler IJ. AACR centennial series: the biology of cancer metastasis: historical perspective. Cancer Res. 2010;70(14):5649–69.

    Article  PubMed  CAS  Google Scholar 

  14. Spratt JS, Spratt JA. Growth rates. In: Donegan WL editor. Cancer of the Breast. Spratt, J.S.; 2002. p. 443–476.

  15. Gupta GP, Massague J. Cancer metastasis: building a framework. Cell. 2006;127(4):679–95.

    Article  PubMed  CAS  Google Scholar 

  16. Li F, Tiede B, Massague J, Kang Y. Beyond tumorigenesis: cancer stem cells in metastasis. Cell Res. 2007;17(1):3–14.

    Article  PubMed  CAS  Google Scholar 

  17. Serra R, Crowley MR. TGF-β in mammary gland development and breast cancer. Breast Dis. 2003;18:61–73.

    PubMed  CAS  Google Scholar 

  18. Serra R, Crowley MR. Mouse models of transforming growth factor β impact in breast development and cancer. Endocr Relat Cancer. 2005;12(4):749–60.

    Article  PubMed  CAS  Google Scholar 

  19. Tian M, Schiemann WP. The TGF-β paradox in human cancer: an update. Future Oncol. 2009;5(2):259–71.

    Article  PubMed  CAS  Google Scholar 

  20. Barcellos-Hoff MH, Akhurst RJ. Transforming growth factor-β in breast cancer: too much, too late. Breast Cancer Res. 2009;11(1):202.

    Article  PubMed  CAS  Google Scholar 

  21. Tian M, Neil JR, Schiemann WP. Transforming growth factor-β and the hallmarks of cancer. Cell Signal. 2010; PMID: 20940046.

    Google Scholar 

  22. Bierie B, Moses HL. Tumour microenvironment: TGFβ: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer. 2006;6(7):506–20.

    Article  PubMed  CAS  Google Scholar 

  23. Rahimi RA, Leof EB. TGF-β signaling: a tale of two responses. J Cell Biochem. 2007;102(3):593–608.

    Article  PubMed  CAS  Google Scholar 

  24. Schiemann WP. Targeted TGF-β chemotherapies: friend or foe in treating human malignancies? Expert Rev Anticancer Ther. 2007;7(5):609–11.

    Article  PubMed  CAS  Google Scholar 

  25. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran S, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141(1):69–80.

    Article  PubMed  CAS  Google Scholar 

  26. Taube JH, Herschkowitz JI, Komurov K, Zhou AY, Gupta S, Yang J, et al. Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc Natl Acad Sci USA. 2010;107(35):15449–54.

    Article  PubMed  CAS  Google Scholar 

  27. Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA. 2009;106(33):13820–5.

    Article  PubMed  CAS  Google Scholar 

  28. Farmer P, Bonnefoi H, Anderle P, Cameron D, Wirapati P, Becette V, et al. A stroma-related gene signature predicts resistance to neoadjuvant chemotherapy in breast cancer. Nat Med. 2009;15(1):68–74.

    Article  PubMed  CAS  Google Scholar 

  29. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–15.

    Article  PubMed  CAS  Google Scholar 

  30. Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N, Yao J, et al. Molecular definition of breast tumor heterogeneity. Cancer Cell. 2007;11(3):259–73.

    Article  PubMed  CAS  Google Scholar 

  31. Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer. 2007;7(2):139–47.

    PubMed  CAS  Google Scholar 

  32. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor β in human disease. N Engl J Med. 2000;342(18):1350–8.

    Article  PubMed  CAS  Google Scholar 

  33. Massague J. TGF-β signal transduction. Annu Rev Biochem. 1998;67:753–91.

    Article  PubMed  CAS  Google Scholar 

  34. Chang H, Brown CW, Matzuk MM. Genetic analysis of the mammalian transforming growth factor-β superfamily. Endocr Rev. 2002;23(6):787–823.

    Article  PubMed  CAS  Google Scholar 

  35. Nishimura SL. Integrin-mediated transforming growth factor-β activation, a potential therapeutic target in fibrogenic disorders. Am J Pathol. 2009;175(4):1362–70.

    Article  PubMed  CAS  Google Scholar 

  36. Rifkin DB. Latent transforming growth factor-β (TGF-β) binding proteins: orchestrators of TGF-β availability. J Biol Chem. 2005;280(9):7409–12.

    PubMed  CAS  Google Scholar 

  37. Massague J, Gomis RR. The logic of TGFβ signaling. FEBS Lett. 2006;580(12):2811–20.

    Article  PubMed  CAS  Google Scholar 

  38. Massague J, Seoane J, Wotton D. Smad transcription factors. Genes Dev. 2005;19(23):2783–810.

    Article  PubMed  CAS  Google Scholar 

  39. Tsukazaki T, Chiang TA, Davison AF, Attisano L, Wrana JL. SARA, a FYVE domain protein that recruits Smad2 to the TGFβreceptor. Cell. 1998;95(6):779–91.

    Article  PubMed  CAS  Google Scholar 

  40. Miura S, Takeshita T, Asao H, Kimura Y, Murata K, Sasaki Y, et al. Hgs (Hrs), a FYVE domain protein, is involved in Smad signaling through cooperation with SARA. Mol Cell Biol. 2000;20(24):9346–55.

    Article  PubMed  CAS  Google Scholar 

  41. Faresse N, Colland F, Ferrand N, Prunier C, Bourgeade MF, Atfi A. Identification of PCTA, a TGIF antagonist that promotes PML function in TGF-β signalling. EMBO J. 2008;27(13):1804–15.

    Article  PubMed  CAS  Google Scholar 

  42. Hocevar BA, Smine A, Xu XX, Howe PH. The adaptor molecule Disabled-2 links the transforming growth factor β receptors to the Smad pathway. EMBO J. 2001;20(11):2789–801.

    Article  PubMed  CAS  Google Scholar 

  43. Penheiter SG, Singh RD, Repellin CE, Wilkes MC, Edens M, Howe PH, et al. Type II transforming growth factor-β receptor recycling is dependent upon the clathrin adaptor protein Dab2. Mol Biol Cell. 2010;21(22):4009–19.

    Article  PubMed  CAS  Google Scholar 

  44. Runyan CE, Hayashida T, Hubchak S, Curley JF, Schnaper HW. Role of SARA (SMAD anchor for receptor activation) in maintenance of epithelial cell phenotype. J Biol Chem. 2009;284(37):25181–9.

    Article  PubMed  CAS  Google Scholar 

  45. Chaudhury A, Hussey GS, Ray PS, Jin G, Fox PL, Howe PH. TGF-β-mediated phosphorylation of hnRNP E1 induces EMT via transcript-selective translational induction of Dab2 and ILEI. Nat Cell Biol. 2010;12(3):286–93.

    PubMed  CAS  Google Scholar 

  46. Hocevar BA, Prunier C, Howe PH. Disabled-2 (Dab2) mediates transforming growth factor β (TGFβ)-stimulated fibronectin synthesis through TGFβ-activated kinase 1 and activation of the JNK pathway. J Biol Chem. 2005;280(27):25920–7.

    Article  PubMed  CAS  Google Scholar 

  47. Hussey GS, Chaudhury A, Dawson AE, Lindner DJ, Knudsen CR, Wilce MC, et al. Identification of an mRNP complex regulating tumorigenesis at the translational elongation step. Mol Cell. 2011;41(4):419–31.

    Article  PubMed  CAS  Google Scholar 

  48. Kim W, Seok Kang Y, Soo Kim J, Shin NY, Hanks SK, Song WK. The integrin-coupled signaling adaptor p130Cas suppresses Smad3 function in transforming growth factor-β signaling. Mol Biol Cell. 2008;19(5):2135–46.

    Article  PubMed  CAS  Google Scholar 

  49. Wendt MK, Smith JA, Schiemann WP. p130Cas is required for mammary tumor growth and transforming growth factor-β-mediated metastasis through regulation of Smad2/3 activity. J Biol Chem. 2009;284(49):34145–56.

    Article  PubMed  CAS  Google Scholar 

  50. Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, et al. The MAD-related protein Smad7 associates with the TGFβ receptor and functions as an antagonist of TGFβ signaling. Cell. 1997;89:1165–73.

    Article  PubMed  CAS  Google Scholar 

  51. Nakao A, Afrakht M, Moren A, Nakayama T, Christian JL, Heuchel R, et al. Identification of Smad7, a TGFβ-inducible antagonist of TGF-β signalling. Nature. 1997;389:631–5.

    Article  PubMed  CAS  Google Scholar 

  52. Souchelnytskyi S, Nakayama T, Nakao A, Moren A, Heldin CH, Christian JL, et al. Physical and functional interaction of murine and xenopus Smad7 with bone morphogenetic protein receptors and transforming growth factor-β receptors. J Biol Chem. 1998;273:25364–70.

    Article  PubMed  CAS  Google Scholar 

  53. Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K, Imamura T, et al. Smurf1 interacts with transforming growth factor-β type I receptor through Smad7 and induces receptor degradation. J Biol Chem. 2001;276(16):12477–80.

    Article  PubMed  CAS  Google Scholar 

  54. Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H, Thomsen GH, et al. Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGFβ receptor for degradation. Mol Cell. 2000;6(6):1365–75.

    Article  PubMed  CAS  Google Scholar 

  55. Datta PK, Moses HL. STRAP and Smad7 synergize in the inhibition of transforming growth factor β signaling. Mol Cell Biol. 2000;20(9):3157–67.

    Article  PubMed  CAS  Google Scholar 

  56. Ibarrola N, Kratchmarova I, Nakajima D, Schiemann WP, Moustakas A, Pandey A, et al. Cloning of a novel signaling molecule, AMSH-2, that potentiates transforming growth factor β signaling. BMC Cell Biol. 2004;5:2.

    Article  PubMed  Google Scholar 

  57. Koinuma D, Shinozaki M, Komuro A, Goto K, Saitoh M, Hanyu A, et al. Arkadia amplifies TGF-β superfamily signalling through degradation of Smad7. EMBO J. 2003;22(24):6458–70.

    Article  PubMed  CAS  Google Scholar 

  58. Lin X, Duan X, Liang YY, Su Y, Wrighton KH, Long J, et al. PPM1A functions as a Smad phosphatase to terminate TGFβ signaling. Cell. 2006;125(5):915–28.

    Article  PubMed  CAS  Google Scholar 

  59. Lin X, Liang M, Feng XH. Smurf2 is a ubiquitin E3 ligase mediating proteasome-dependent degradation of Smad2 in transforming growth factor-β signaling. J Biol Chem. 2000;275(47):36818–22.

    Article  PubMed  CAS  Google Scholar 

  60. Lo RS, Massague J. Ubiquitin-dependent degradation of TGF-β-activated Smad2. Nat Cell Biol. 1999;1(8):472–8.

    Article  PubMed  CAS  Google Scholar 

  61. Fukuchi M, Imamura T, Chiba T, Ebisawa T, Kawabata M, Tanaka K, et al. Ligand-dependent degradation of Smad3 by a ubiquitin ligase complex of ROC1 and associated proteins. Mol Biol Cell. 2001;12(5):1431–43.

    PubMed  CAS  Google Scholar 

  62. Izzi L, Attisano L. Regulation of the TGFβ signalling pathway by ubiquitin-mediated degradation. Oncogene. 2004;23(11):2071–8.

    Article  PubMed  CAS  Google Scholar 

  63. Dupont S, Mamidi A, Cordenonsi M, Montagner M, Zacchigna L, Adorno M, et al. FAM/USP9x, a deubiquitinating enzyme essential for TGFβ signaling, controls Smad4 monoubiquitination. Cell. 2009;136(1):123–35.

    Article  PubMed  CAS  Google Scholar 

  64. Derynck R, Akhurst RJ, Balmain A. TGF-β signaling in tumor suppression and cancer progression. Nat Genet. 2001;29(2):117–29.

    Article  PubMed  CAS  Google Scholar 

  65. Massague J. TGFβ in cancer. Cell. 2008;134(2):215–30.

    Article  PubMed  CAS  Google Scholar 

  66. Derynck R, Akhurst RJ. Differentiation plasticity regulated by TGF-β family proteins in development and disease. Nat Cell Biol. 2007;9(9):1000–4.

    Article  PubMed  CAS  Google Scholar 

  67. Piek E, Moustakas A, Kurisaki A, Heldin CH, ten Dijke P. TGF-β type I receptor/ALK-5 and Smad proteins mediate epithelial to mesenchymal transdifferentiation in NMuMG breast epithelial cells. J Cell Sci. 1999;112:4557–68.

    PubMed  CAS  Google Scholar 

  68. Valcourt U, Kowanetz M, Niimi H, Heldin CH, Moustakas A. TGF-β and the Smad signaling pathway support transcriptomic reprogramming during epithelial-mesenchymal cell transition. Mol Biol Cell. 2005;16(4):1987–2002.

    Article  PubMed  CAS  Google Scholar 

  69. Wendt MK, Allington TM, Schiemann WP. Mechanisms of the epithelial-mesenchymal transition by TGF-β. Future Oncol. 2009;5(8):1145–68.

    Article  PubMed  CAS  Google Scholar 

  70. Oft M, Akhurst RJ, Balmain A. Metastasis is driven by sequential elevation of H-Ras and Smad2 levels. Nat Cell Biol. 2002;4(7):487–94.

    Article  PubMed  CAS  Google Scholar 

  71. Lehmann K, Janda E, Pierreux CE, Rytomaa M, Schulze A, McMahon M, et al. 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(20):2610–22.

    Article  PubMed  CAS  Google Scholar 

  72. Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox-2 inactivates Smad signaling and enhances EMT stimulated by TGF-β through a PGE2-dependent mechanisms. Carcinogenesis. 2008;29(11):2227–35.

    Article  PubMed  CAS  Google Scholar 

  73. Neil JR, Schiemann WP. Altered TAB1:IκB kinase interaction promotes transforming growth factor β-mediated nuclear factor-κB activation during breast cancer progression. Cancer Res. 2008;68(5):1462–70.

    Article  PubMed  CAS  Google Scholar 

  74. Neil JR, Tian M, Schiemann WP. x-linked inhibitor of apoptosis protein and its E3 ligase activity promote transforming growth factor-β-mediated nuclear factor-κB activation during breast cancer progression. J Biol Chem. 2009;284(32):21209–17.

    Article  PubMed  CAS  Google Scholar 

  75. Tian M, Schiemann WP. PGE2 receptor EP2 mediates the antagonistic effect of COX-2 on TGF-β signaling during mammary tumorigenesis. FASEB J. 2010;24(4):1105–16.

    Article  PubMed  CAS  Google Scholar 

  76. Kretzschmar M, Doody J, Timokhina I, Massague J. A mechanism of repression of TGFβ/ Smad signaling by oncogenic Ras. Genes Dev. 1999;13(7):804–16.

    Article  PubMed  CAS  Google Scholar 

  77. Funaba M, Zimmerman CM, Mathews LS. Modulation of Smad2-mediated signaling by extracellular signal-regulated kinase. J Biol Chem. 2002;277(44):41361–8.

    Article  PubMed  CAS  Google Scholar 

  78. Brown JD, DiChiara MR, Anderson KR, Gimbrone Jr MA, Topper JN. MEKK-1, a component of the stress (stress-activated protein kinase/c-Jun N-terminal kinase) pathway, can selectively activate Smad2-mediated transcriptional activation in endothelial cells. J Biol Chem. 1999;274(13):8797–805.

    Article  PubMed  CAS  Google Scholar 

  79. Engel ME, McDonnell MA, Law BK, Moses HL. Interdependent SMAD and JNK signaling in transforming growth factor-β-mediated transcription. J Biol Chem. 1999;274(52):37413–20.

    Article  PubMed  CAS  Google Scholar 

  80. Waddell DS, Liberati NT, Guo X, Frederick JP, Wang XF. Casein kinase Iepsilon plays a functional role in the transforming growth factor-β signaling pathway. J Biol Chem. 2004;279(28):29236–46.

    Article  PubMed  CAS  Google Scholar 

  81. Yakymovych I, Ten Dijke P, Heldin CH, Souchelnytskyi S. Regulation of Smad signaling by protein kinase C. FASEB J. 2001;15(3):553–5.

    PubMed  CAS  Google Scholar 

  82. Wicks SJ, Lui S, Abdel-Wahab N, Mason RM, Chantry A. Inactivation of smad-transforming growth factor β signaling by Ca(2+)-calmodulin-dependent protein kinase II. Mol Cell Biol. 2000;20(21):8103–11.

    Article  PubMed  CAS  Google Scholar 

  83. Guo X, Ramirez A, Waddell DS, Li Z, Liu X, Wang XF. Axin and GSK3βcontrol Smad3 protein stability and modulate TGF-βsignaling. Genes Dev. 2008;22(1):106–20.

    Article  PubMed  CAS  Google Scholar 

  84. Allington TM, Galliher-Beckley AJ, Schiemann WP. Activated Abl kinase inhibits oncogenic transforming growth factor-β signaling and tumorigenesis in mammary tumors. FASEB J. 2009;23(12):4231–43.

    Article  PubMed  CAS  Google Scholar 

  85. Galliher AJ, Schiemann WP. β3 integrin and Src facilitate transforming growth factor-β mediated induction of epithelial-mesenchymal transition in mammary epithelial cells. Breast Cancer Res. 2006;8(4):R42.

    Article  PubMed  CAS  Google Scholar 

  86. Galliher AJ, Schiemann WP. Src phosphorylates Tyr284 in TGF-β type II receptor and regulates TGF-β stimulation of p38 MAPK during breast cancer cell proliferation and invasion. Cancer Res. 2007;67(8):3752–8.

    Article  PubMed  CAS  Google Scholar 

  87. Galliher-Beckley AJ, Schiemann WP. Grb2 binding to Tyr284 in TβR-II is essential for mammary tumor growth and metastasis stimulated by TGF-β. Carcinogenesis. 2008;29(2):244–51.

    PubMed  CAS  Google Scholar 

  88. Wendt MK, Schiemann WP. Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis. Breast Cancer Res. 2009;11(5):R68.

    Article  PubMed  CAS  Google Scholar 

  89. Wendt MK, Smith JA, Schiemann WP. Transforming growth factor-β-induced epithelial-mesenchymal transition facilitates epidermal growth factor-dependent breast cancer progression. Oncogene. 2010;29(49):6485–98.

    Article  PubMed  CAS  Google Scholar 

  90. Taylor MA, Amin J, Kirschmann DA, Schiemann WP. Lysyl oxidase and hydrogen peroxide promote oncogenic signaling by transforming growth factor-βin mammary epithelial cells. Neoplasia. 2011;13:in press.

  91. Lee YH, Albig AR, Regner M, Schiemann BJ, Schiemann WP. Fibulin-5 initiates epithelial-mesenchymal transition (EMT) and enhances EMT induced by TGF-βin mammary epithelial cells via a MMP-dependent mechanism. Carcinogenesis. 2008;29(12):2243–51.

    Article  PubMed  CAS  Google Scholar 

  92. Moustakas A, Heldin CH. Non-Smad TGF-βsignals. J Cell Sci. 2005;118(Pt 16):3573–84.

    Article  PubMed  CAS  Google Scholar 

  93. Kang JS, Liu C, Derynck R. New regulatory mechanisms of TGF-β receptor function. Trends Cell Biol. 2009;19(8):385–94.

    Article  PubMed  CAS  Google Scholar 

  94. Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10(1):9–22.

    Article  PubMed  CAS  Google Scholar 

  95. Egeblad M, Nakasone ES, Werb Z. Tumors as organs: complex tissues that interface with the entire organism. Dev Cell. 2010;18(6):884–901.

    Article  PubMed  CAS  Google Scholar 

  96. Zutter MM. Integrin-mediated adhesion: tipping the balance between chemosensitivity and chemoresistance. Adv Exp Med Biol. 2007;608:87–100.

    Article  PubMed  CAS  Google Scholar 

  97. Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu J, et al. The integrin αvβ6 binds and activates latent TGFβ1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell. 1999;96(3):319–28.

    Article  PubMed  CAS  Google Scholar 

  98. Mu D, Cambier S, Fjellbirkeland L, Baron JL, Munger JS, Kawakatsu H, et al. The integrin αvβ8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-β1. J Cell Biol. 2002;157(3):493–507.

    Article  PubMed  CAS  Google Scholar 

  99. Sloan EK, Pouliot N, Stanley KL, Chia J, Moseley JM, Hards DK, et al. Tumor-specific expression of αvβ3 integrin promotes spontaneous metastasis of breast cancer to bone. Breast Cancer Res. 2006;8(2):R20.

    Article  PubMed  CAS  Google Scholar 

  100. Bandyopadhyay A, Agyin JK, Wang L, Tang Y, Lei X, Story BM, et al. Inhibition of pulmonary and skeletal metastasis by a transforming growth factor-β type I receptor kinase inhibitor. Cancer Res. 2006;66(13):6714–21.

    Article  PubMed  CAS  Google Scholar 

  101. Garamszegi N, Garamszegi SP, Samavarchi-Tehrani P, Walford E, Schneiderbauer MM, Wrana JL, et al. Extracellular matrix-induced transforming growth factor-β receptor signaling dynamics. Oncogene. 2010;29(16):2368–80.

    Article  PubMed  CAS  Google Scholar 

  102. van der Flier S, Chan CM, Brinkman A, Smid M, Johnston SR, Dorssers LC, et al. BCAR1/p130Cas expression in untreated and acquired tamoxifen-resistant human breast carcinomas. Int J Cancer. 2000;89(5):465–8.

    Article  PubMed  Google Scholar 

  103. Ta HQ, Thomas KS, Schrecengost RS, Bouton AH. A novel association between p130Cas and resistance to the chemotherapeutic drug adriamycin in human breast cancer cells. Cancer Res. 2008;68(21):8796–804.

    Article  PubMed  CAS  Google Scholar 

  104. Cabodi S, Tinnirello A, Bisaro B, Tornillo G, del Pilar Camacho-Leal M, Forni G, et al. p130Cas is an essential transducer element in ErbB2 transformation. FASEB J. 2010;24(10):3796–808.

    Article  PubMed  CAS  Google Scholar 

  105. Cabodi S, Tinnirello A, Di Stefano P, Bisaro B, Ambrosino E, Castellano I, et al. p130Cas as a new regulator of mammary epithelial cell proliferation, survival, and HER2-neu oncogene-dependent breast tumorigenesis. Cancer Res. 2006;66(9):4672–80.

    Article  PubMed  CAS  Google Scholar 

  106. Bhowmick NA, Zent R, Ghiassi M, McDonnell M, Moses HL. Integrin β1 signaling is necessary for transforming growth factor-β activation of p38MAPK and epithelial plasticity. J Biol Chem. 2001;276(50):46707–13.

    Article  PubMed  CAS  Google Scholar 

  107. Mizejewski GJ. Role of integrins in cancer: survey of expression patterns. Proc Soc Exp Biol Med. 1999;222(2):124–38.

    Article  PubMed  CAS  Google Scholar 

  108. Weaver VM, Lelievre S, Lakins JN, Chrenek MA, Jones JC, Giancotti F, et al. β4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium. Cancer Cell. 2002;2(3):205–16.

    Article  PubMed  CAS  Google Scholar 

  109. Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009;9(8):550–62.

    Article  PubMed  CAS  Google Scholar 

  110. Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga CL. Phosphatidylinositol 3-kinase function is required for transforming growth factor β-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem. 2000;275(47):36803–10.

    Article  PubMed  CAS  Google Scholar 

  111. Murillo MM, del Castillo G, Sanchez A, Fernandez M, Fabregat I. Involvement of EGF receptor and c-Src in the survival signals induced by TGF-β1 in hepatocytes. Oncogene. 2005;24(28):4580–7.

    Article  PubMed  CAS  Google Scholar 

  112. Jechlinger M, Sommer A, Moriggl R, Seither P, Kraut N, Capodiecci P, et al. Autocrine PDGFR signaling promotes mammary cancer metastasis. J Clin Invest. 2006;116(6):1561–70.

    Article  PubMed  CAS  Google Scholar 

  113. Uttamsingh S, Bao X, Nguyen KT, Bhanot M, Gong J, Chan JL, et al. Synergistic effect between EGF and TGF-β1 in inducing oncogenic properties of intestinal epithelial cells. Oncogene. 2008;27(18):2626–34.

    Article  PubMed  CAS  Google Scholar 

  114. Lamouille S, Derynck R. Cell size and invasion in TGF-β-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 2007;178(3):437–51.

    Article  PubMed  CAS  Google Scholar 

  115. Conery AR, Cao Y, Thompson EA, Townsend Jr CM, Ko TC, Luo K. Akt interacts directly with Smad3 to regulate the sensitivity to TGF-β induced apoptosis. Nat Cell Biol. 2004;6(4):366–72.

    Article  PubMed  CAS  Google Scholar 

  116. Remy I, Montmarquette A, Michnick SW. PKB/Akt modulates TGF-β signalling through a direct interaction with Smad3. Nat Cell Biol. 2004;6(4):358–65.

    Article  PubMed  CAS  Google Scholar 

  117. Xu J, Lamouille S, Derynck R. TGF-β-induced epithelial to mesenchymal transition. Cell Res. 2009;19(2):156–72.

    Article  PubMed  CAS  Google Scholar 

  118. Heldin CH, Landstrom M, Moustakas A. Mechanism of TGF-β signaling to growth arrest, apoptosis, and epithelial-mesenchymal transition. Curr Opin Cell Biol. 2009;21(2):166–76.

    Article  PubMed  CAS  Google Scholar 

  119. Hall A. Rho GTPases and the control of cell behaviour. Biochem Soc Trans. 2005;33(Pt 5):891–5.

    PubMed  CAS  Google Scholar 

  120. Hall A. The cytoskeleton and cancer. Cancer Metastasis Rev. 2009;28(1–2):5–14.

    Article  PubMed  Google Scholar 

  121. Bhowmick NA, Ghiassi M, Bakin A, Aakre M, Lundquist CA, Engel ME, et al. Transforming growth factor-β1 mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell. 2001;12(1):27–36.

    PubMed  CAS  Google Scholar 

  122. Viloria-Petit AM, David L, Jia JY, Erdemir T, Bane AL, Pinnaduwage D, et al. A role for the TGFβ-Par6 polarity pathway in breast cancer progression. Proc Natl Acad Sci USA. 2009;106(33):14028–33.

    Article  PubMed  CAS  Google Scholar 

  123. Ozdamar B, Bose R, Barrios-Rodiles M, Wang HR, Zhang Y, Wrana JL. Regulation of the polarity protein Par6 by TGFβ receptors controls epithelial cell plasticity. Science. 2005;307(5715):1603–9.

    Article  PubMed  CAS  Google Scholar 

  124. Hutchison N, Hendry BM, Sharpe CC. Rho isoforms have distinct and specific functions in the process of epithelial to mesenchymal transition in renal proximal tubular cells. Cell Signal. 2009;21(10):1522–31.

    Article  PubMed  CAS  Google Scholar 

  125. He Y, Northey JJ, Primeau M, Machado RD, Trembath R, Siegel PM, et al. CdGAP is required for transforming growth factor β- and Neu/ErbB-2-induced breast cancer cell motility and invasion. Oncogene. 2010;30(9):1032–45.

    PubMed  Google Scholar 

  126. Mythreye K, Blobe GC. The type III TGF-β receptor regulates epithelial and cancer cell migration through β-arrestin2-mediated activation of Cdc42. Proc Natl Acad Sci USA. 2009;106(20):8221–6.

    Article  PubMed  CAS  Google Scholar 

  127. Kong W, Yang H, He L, Zhao JJ, Coppola D, Dalton WS, et al. MicroRNA-155 is regulated by the transforming growth factor β/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Mol Cell Biol. 2008;28(22):6773–84.

    Article  PubMed  CAS  Google Scholar 

  128. Valastyan S, Benaich N, Chang A, Reinhardt F, Weinberg RA. Concomitant suppression of three target genes can explain the impact of a microRNA on metastasis. Genes Dev. 2009;23(22):2592–7.

    Article  PubMed  CAS  Google Scholar 

  129. Valastyan S, Reinhardt F, Benaich N, Calogrias D, Szasz AM, Wang ZC, et al. A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell. 2009;137(6):1032–46.

    Article  PubMed  CAS  Google Scholar 

  130. Atfi A, Djelloul S, Chastre E, Davis R, Gespach C. Evidence for a role of Rho-like GTPases and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in transforming growth factor β-mediated signaling. J Biol Chem. 1997;272(3):1429–32.

    PubMed  CAS  Google Scholar 

  131. Bakin AV, Rinehart C, Tomlinson AK, Arteaga CL. p38 mitogen-activated protein kinase is required for TGFβ-mediated fibroblastic transdifferentiation and cell migration. J Cell Sci. 2002;115(Pt 15):3193–206.

    PubMed  CAS  Google Scholar 

  132. Xie L, Law BK, Chytil AM, Brown KA, Aakre ME, Moses HL. Activation of the Erk pathway is required for TGF-β1-induced EMT in vitro. Neoplasia. 2004;6(5):603–10.

    Article  PubMed  CAS  Google Scholar 

  133. Shintani Y, Hollingsworth MA, Wheelock MJ, Johnson KR. Collagen I promotes metastasis in pancreatic cancer by activating c-Jun NH(2)-terminal kinase 1 and up-regulating N-cadherin expression. Cancer Res. 2006;66(24):11745–53.

    Article  PubMed  CAS  Google Scholar 

  134. Shintani Y, Wheelock MJ, Johnson KR. Phosphoinositide-3 kinase-Rac1-c-Jun NH2-terminal kinase signaling mediates collagen I-induced cell scattering and up-regulation of N-cadherin expression in mouse mammary epithelial cells. Mol Biol Cell. 2006;17(7):2963–75.

    Article  PubMed  CAS  Google Scholar 

  135. Sorrentino A, Thakur N, Grimsby S, Marcusson A, von Bulow V, Schuster N, et al. The type I TGF-β receptor engages TRAF6 to activate TAK1 in a receptor kinase-independent manner. Nat Cell Biol. 2008;10(10):1199–207.

    Article  PubMed  CAS  Google Scholar 

  136. Yamashita M, Fatyol K, Jin C, Wang X, Liu Z, Zhang YE. TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-β. Mol Cell. 2008;31(6):918–24.

    Article  PubMed  CAS  Google Scholar 

  137. Karin M. Nuclear factor-κB in cancer development and progression. Nature. 2006;441(7092):431–6.

    Article  PubMed  CAS  Google Scholar 

  138. Sovak MA, Arsura M, Zanieski G, Kavanagh KT, Sonenshein GE. The inhibitory effects of transforming growth factor β1 on breast cancer cell proliferation are mediated through regulation of aberrant nuclear factor-κB/Rel expression. Cell Growth Differ. 1999;10(8):537–44.

    PubMed  CAS  Google Scholar 

  139. Arsura M, Wu M, Sonenshein GE. TGF β1 inhibits NF-κB/Rel activity inducing apoptosis of B cells: transcriptional activation of IκBα. Immunity. 1996;5(1):31–40.

    Article  PubMed  CAS  Google Scholar 

  140. You HJ, How T, Blobe GC. The type III transforming growth factor-β receptor negatively regulates nuclear factor κB signaling through its interaction with β-arrestin2. Carcinogenesis. 2009;30(8):1281–7.

    Article  PubMed  CAS  Google Scholar 

  141. Chua HL, Bhat-Nakshatri P, Clare SE, Morimiya A, Badve S, Nakshatri H. NF-κB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: potential involvement of ZEB-1 and ZEB-2. Oncogene. 2007;26(5):711–24.

    Article  PubMed  CAS  Google Scholar 

  142. Huber MA, Azoitei N, Baumann B, Grunert S, Sommer A, Pehamberger H, et al. NF-κB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J Clin Invest. 2004;114(4):569–81.

    PubMed  CAS  Google Scholar 

  143. Kagan HM, Li W. Lysyl oxidase: properties, specificity, and biological roles inside and outside of the cell. J Cell Biochem. 2003;88(4):660–72.

    Article  PubMed  CAS  Google Scholar 

  144. Payne SL, Hendrix MJ, Kirschmann DA. Paradoxical roles for lysyl oxidases in cancer–a prospect. J Cell Biochem. 2007;101(6):1338–54.

    Article  PubMed  CAS  Google Scholar 

  145. Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, et al. Tensional homeostasis and the malignant phenotype. Cancer Cell. 2005;8(3):241–54.

    Article  PubMed  CAS  Google Scholar 

  146. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891–906.

    Article  PubMed  CAS  Google Scholar 

  147. Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009;15(1):35–44.

    Article  PubMed  CAS  Google Scholar 

  148. Erler JT, Bennewith KL, Nicolau M, Dornhofer N, Kong C, Le QT, et al. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 2006;440(7088):1222–6.

    Article  PubMed  CAS  Google Scholar 

  149. Payne SL, Hendrix MJ, Kirschmann DA. Lysyl oxidase regulates actin filament formation through the p130(Cas)/Crk/DOCK180 signaling complex. J Cell Biochem. 2006;98(4):827–37.

    Article  PubMed  CAS  Google Scholar 

  150. Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+ CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13(1):23–35.

    Article  PubMed  CAS  Google Scholar 

  151. Moustakas A, Heldin CH. Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007;98(10):1512–20.

    Article  PubMed  CAS  Google Scholar 

  152. Tomaskovic-Crook E, Thompson EW, Thiery JP. Epithelial to mesenchymal transition and breast cancer. Breast Cancer Res. 2009;11(6):213.

    Article  PubMed  CAS  Google Scholar 

  153. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420–8.

    Article  PubMed  CAS  Google Scholar 

  154. Padua D, Zhang XH, Wang Q, Nadal C, Gerald WL, Gomis RR, et al. TGFβ primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 2008;133(1):66–77.

    Article  PubMed  CAS  Google Scholar 

  155. Furuse M. Molecular basis of the core structure of tight junctions. Cold Spring Harb Perspect Biol. 2010;2(1):a002907.

    Article  PubMed  Google Scholar 

  156. Martin TA, Jiang WG. Loss of tight junction barrier function and its role in cancer metastasis. Biochim Biophys Acta. 2009;1788(4):872–91.

    PubMed  CAS  Google Scholar 

  157. Ebnet K, Suzuki A, Ohno S, Vestweber D. Junctional adhesion molecules (JAMs): more molecules with dual functions? J Cell Sci. 2004;117(Pt 1):19–29.

    Article  PubMed  CAS  Google Scholar 

  158. Schneeberger EE, Lynch RD. The tight junction: a multifunctional complex. Am J Physiol Cell Physiol. 2004;286(6):C1213–28.

    Article  PubMed  CAS  Google Scholar 

  159. Itoh M, Bissell MJ. The organization of tight junctions in epithelia: implications for mammary gland biology and breast tumorigenesis. J Mammary Gland Biol Neoplasia. 2003;8(4):449–62.

    Article  PubMed  Google Scholar 

  160. Lacher MD, Tiirikainen MI, Saunier EF, Christian C, Anders M, Oft M, et al. Transforming growth factor-β receptor inhibition enhances adenoviral infectability of carcinoma cells via up-regulation of Coxsackie and Adenovirus Receptor in conjunction with reversal of epithelial-mesenchymal transition. Cancer Res. 2006;66(3):1648–57.

    Article  PubMed  CAS  Google Scholar 

  161. Vincent T, Neve EP, Johnson JR, Kukalev A, Rojo F, Albanell J, et al. A SNAIL1-SMAD3/4 transcriptional repressor complex promotes TGF-β mediated epithelial-mesenchymal transition. Nat Cell Biol. 2009;11(8):943–50.

    Article  PubMed  CAS  Google Scholar 

  162. Grande M, Franzen A, Karlsson JO, Ericson LE, Heldin NE, Nilsson M. Transforming growth factor-β and epidermal growth factor synergistically stimulate epithelial to mesenchymal transition (EMT) through a MEK-dependent mechanism in primary cultured pig thyrocytes. J Cell Sci. 2002;115(Pt 22):4227–36.

    Article  PubMed  CAS  Google Scholar 

  163. Shiou SR, Singh AB, Moorthy K, Datta PK, Washington MK, Beauchamp RD, et al. Smad4 regulates claudin-1 expression in a transforming growth factor-β-independent manner in colon cancer cells. Cancer Res. 2007;67(4):1571–9.

    Article  PubMed  CAS  Google Scholar 

  164. Barrios-Rodiles M, Brown KR, Ozdamar B, Bose R, Liu Z, Donovan RS, et al. High-throughput mapping of a dynamic signaling network in mammalian cells. Science. 2005;307(5715):1621–5.

    Article  PubMed  CAS  Google Scholar 

  165. Murphy SJ, Dore JJ, Edens M, Coffey RJ, Barnard JA, Mitchell H, et al. Differential trafficking of transforming growth factor-β receptors and ligand in polarized epithelial cells. Mol Biol Cell. 2004;15(6):2853–62.

    Article  PubMed  CAS  Google Scholar 

  166. Yakovich AJ, Huang Q, Du J, Jiang B, Barnard JA. Vectorial TGFβ signaling in polarized intestinal epithelial cells. J Cell Physiol. 2010;224(2):398–404.

    Article  PubMed  CAS  Google Scholar 

  167. Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer. 2004;4(2):118–32.

    Article  PubMed  CAS  Google Scholar 

  168. Agiostratidou G, Hulit J, Phillips GR, Hazan RB. Differential cadherin expression: potential markers for epithelial to mesenchymal transformation during tumor progression. J Mammary Gland Biol Neoplasia. 2007;12(2–3):127–33.

    Article  PubMed  Google Scholar 

  169. Gravdal K, Halvorsen OJ, Haukaas SA, Akslen LA. A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer. Clin Cancer Res. 2007;13(23):7003–11.

    Article  PubMed  CAS  Google Scholar 

  170. Tomita K, van Bokhoven A, van Leenders GJ, Ruijter ET, Jansen CF, Bussemakers MJ, et al. Cadherin switching in human prostate cancer progression. Cancer Res. 2000;60(13):3650–4.

    PubMed  CAS  Google Scholar 

  171. Hazan RB, Phillips GR, Qiao RF, Norton L, Aaronson SA. Exogenous expression of N-cadherin in breast cancer cells induces cell migration, invasion, and metastasis. J Cell Biol. 2000;148(4):779–90.

    Article  PubMed  CAS  Google Scholar 

  172. Micalizzi DS, Farabaugh SM, Ford HL. Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia. 2010;15(2):117–34.

    Article  PubMed  Google Scholar 

  173. Christofori G. Changing neighbours, changing behaviour: cell adhesion molecule-mediated signalling during tumour progression. EMBO J. 2003;22(10):2318–23.

    Article  PubMed  CAS  Google Scholar 

  174. Kim KK, Wei Y, Szekeres C, Kugler MC, Wolters PJ, Hill ML, et al. Epithelial cell α3β1 integrin links β-catenin and Smad signaling to promote myofibroblast formation and pulmonary fibrosis. J Clin Invest. 2009;119(1):213–24.

    PubMed  CAS  Google Scholar 

  175. Kim Y, Kugler MC, Wei Y, Kim KK, Li X, Brumwell AN, et al. Integrin α3β1-dependent β-catenin phosphorylation links epithelial Smad signaling to cell contacts. J Cell Biol. 2009;184(2):309–22.

    Article  PubMed  CAS  Google Scholar 

  176. Tian YC, Phillips AO. Interaction between the transforming growth factor-βtype II receptor/Smad pathway and β-catenin during transforming growth factor-β1-mediated adherens junction disassembly. Am J Pathol. 2002;160(5):1619–28.

    Article  PubMed  CAS  Google Scholar 

  177. Vogelmann R, Nguyen-Tat MD, Giehl K, Adler G, Wedlich D, Menke A. TGFβ-induced downregulation of E-cadherin-based cell-cell adhesion depends on PI3-kinase and PTEN. J Cell Sci. 2005;118(Pt 20):4901–12.

    Article  PubMed  CAS  Google Scholar 

  178. Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, et al. TGF-β signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303(5659):848–51.

    Article  PubMed  CAS  Google Scholar 

  179. Kim BG, Li C, Qiao W, Mamura M, Kasprzak B, Anver M, et al. Smad4 signalling in T cells is required for suppression of gastrointestinal cancer. Nature. 2006;441(7096):1015–9.

    Article  PubMed  CAS  Google Scholar 

  180. Cheng N, Bhowmick NA, Chytil A, Gorksa AE, Brown KA, Muraoka R, et al. Loss of TGF-β type II receptor in fibroblasts promotes mammary carcinoma growth and invasion through upregulation of TGF-α-, MSP- and HGF-mediated signaling networks. Oncogene. 2005;24(32):5053–68.

    Article  PubMed  CAS  Google Scholar 

  181. Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature. 2004;432(7015):332–7.

    Article  PubMed  CAS  Google Scholar 

  182. Ignotz RA, Massague J. Transforming growth factor-β stimulates the expression of fibronectin and collagen and their incorporation into the extracellular matrix. J Biol Chem. 1986;261(9):4337–45.

    PubMed  CAS  Google Scholar 

  183. Barkan D, Kleinman H, Simmons JL, Asmussen H, Kamaraju AK, Hoenorhoff MJ, et al. Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res. 2008;68(15):6241–50.

    Article  PubMed  CAS  Google Scholar 

  184. Maschler S, Wirl G, Spring H, Bredow DV, Sordat I, Beug H, et al. Tumor cell invasiveness correlates with changes in integrin expression and localization. Oncogene. 2005;24(12):2032–41.

    Article  PubMed  CAS  Google Scholar 

  185. Wels J, Kaplan RN, Rafii S, Lyden D. Migratory neighbors and distant invaders: tumor-associated niche cells. Genes Dev. 2008;22(5):559–74.

    Article  PubMed  CAS  Google Scholar 

  186. Chandler EM, Saunders MP, Yoon CJ, Gourdon D, Fischbach C. Adipose progenitor cells increase fibronectin matrix strain and unfolding in breast tumors. Phys Biol. 2011;8(1):015008.

    Article  PubMed  CAS  Google Scholar 

  187. Barkan D, El Touny LH, Michalowski AM, Smith JA, Chu I, Davis AS, et al. Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res. 2010;70(14):5706–16.

    Article  PubMed  CAS  Google Scholar 

  188. Bianchini G, Qi Y, Alvarez RH, Iwamoto T, Coutant C, Ibrahim NK, et al. Molecular anatomy of breast cancer stroma and its prognostic value in estrogen receptor-positive and -negative cancers. J Clin Oncol. 2010;28(28):4316–23.

    Article  PubMed  Google Scholar 

  189. Wienke D, Davies GC, Johnson DA, Sturge J, Lambros MB, Savage K, et al. The collagen receptor Endo180 (CD280) Is expressed on basal-like breast tumor cells and promotes tumor growth in vivo. Cancer Res. 2007;67(21):10230–40.

    Article  PubMed  CAS  Google Scholar 

  190. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33.

    Article  PubMed  CAS  Google Scholar 

  191. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 2004;101(9):2999–3004.

    Article  PubMed  CAS  Google Scholar 

  192. Smeets A, Daemen A, Vanden Bempt I, Gevaert O, Claes B, Wildiers H, et al. Prediction of lymph node involvement in breast cancer from primary tumor tissue using gene expression profiling and miRNAs. Breast Cancer Res Treat. 2010: PMID: 21116709.

  193. Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65(16):7065–70.

    Article  PubMed  CAS  Google Scholar 

  194. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 2006;103(7):2257–61.

    Article  PubMed  CAS  Google Scholar 

  195. Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, et al. The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol. 2008;10(2):202–10.

    Article  PubMed  CAS  Google Scholar 

  196. Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007;449(7163):682–8.

    Article  PubMed  CAS  Google Scholar 

  197. Zhu S, Wu H, Wu F, Nie D, Sheng S, Mo YY. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 2008;18(3):350–9.

    Article  PubMed  CAS  Google Scholar 

  198. Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, et al. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008;451(7175):147–52.

    Article  PubMed  CAS  Google Scholar 

  199. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10(5):593–601.

    Article  PubMed  CAS  Google Scholar 

  200. Zavadil J, Narasimhan M, Blumenberg M, Schneider RJ. Transforming growth factor-β and microRNA:mRNA regulatory networks in epithelial plasticity. Cells Tissues Organs. 2007;185(1–3):157–61.

    Article  PubMed  CAS  Google Scholar 

  201. Zhu S, Si ML, Wu H, Mo YY. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem. 2007;282(19):14328–36.

    Article  PubMed  CAS  Google Scholar 

  202. Qian B, Katsaros D, Lu L, Preti M, Durando A, Arisio R, et al. High miR-21 expression in breast cancer associated with poor disease-free survival in early stage disease and high TGFβ1. Breast Cancer Res Treat. 2009;117(1):131–40.

    Article  PubMed  CAS  Google Scholar 

  203. Davis BN, Hilyard AC, Lagna G, Hata A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature. 2008;454(7200):56–61.

    Article  PubMed  CAS  Google Scholar 

  204. Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29(34):4741–51.

    Article  PubMed  CAS  Google Scholar 

  205. Pardal R, Clarke MF, Morrison SJ. Applying the principles of stem-cell biology to cancer. Nat Rev Cancer. 2003;3(12):895–902.

    Article  PubMed  CAS  Google Scholar 

  206. Gupta PB, Chaffer CL, Weinberg RA. Cancer stem cells: mirage or reality? Nat Med. 2009;15(9):1010–2.

    Article  PubMed  CAS  Google Scholar 

  207. Zhou BB, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov. 2009;8(10):806–23.

    Article  PubMed  CAS  Google Scholar 

  208. Polyak K, Weinberg RA. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer. 2009;9(4):265–73.

    Article  PubMed  CAS  Google Scholar 

  209. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100(7):3983–8.

    Article  PubMed  CAS  Google Scholar 

  210. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3(7):730–7.

    Article  PubMed  CAS  Google Scholar 

  211. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.

    Article  PubMed  CAS  Google Scholar 

  212. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318(5858):1917–20.

    Article  PubMed  CAS  Google Scholar 

  213. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.

    Article  PubMed  CAS  Google Scholar 

  214. Pera MF, Tam PP. Extrinsic regulation of pluripotent stem cells. Nature. 2010;465(7299):713–20.

    Article  PubMed  CAS  Google Scholar 

  215. Stadtfeld M, Hochedlinger K. Induced pluripotency: history, mechanisms, and applications. Genes Dev. 2010;24(20):2239–63.

    Article  PubMed  CAS  Google Scholar 

  216. Greber B, Lehrach H, Adjaye J. Fibroblast growth factor 2 modulates transforming growth factor βsignaling in mouse embryonic fibroblasts and human ESCs (hESCs) to support hESC self-renewal. Stem Cells. 2007;25(2):455–64.

    Article  PubMed  CAS  Google Scholar 

  217. James D, Levine AJ, Besser D, Hemmati-Brivanlou A. TGFβ/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells. Development. 2005;132(6):1273–82.

    Article  PubMed  CAS  Google Scholar 

  218. Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu F, Sonntag A, et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat Cell Biol. 2009;11(12):1487–95.

    Article  PubMed  CAS  Google Scholar 

  219. Li HX, Han M, Bernier M, Zheng B, Sun SG, Su M, et al. Kruppel-like factor 4 promotes differentiation by transforming growth factor-βreceptor-mediated Smad and p38 MAPK signaling in vascular smooth muscle cells. J Biol Chem. 2010;285(23):17846–56.

    Article  PubMed  CAS  Google Scholar 

  220. Wielenga VJ, Smits R, Korinek V, Smit L, Kielman M, Fodde R, et al. Expression of CD44 in Apc and Tcf mutant mice implies regulation by the WNT pathway. Am J Pathol. 1999;154(2):515–23.

    Article  PubMed  CAS  Google Scholar 

  221. Bennett CN, Green JE. Unlocking the power of cross-species genomic analyses: identification of evolutionarily conserved breast cancer networks and validation of preclinical models. Breast Cancer Res. 2008;10(5):213.

    Article  PubMed  CAS  Google Scholar 

  222. Aslakson CJ, Miller FR. Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 1992;52(6):1399–405.

    PubMed  CAS  Google Scholar 

  223. Santner SJ, Dawson PJ, Tait L, Soule HD, Eliason J, Mohamed AN, et al. Malignant MCF10CA1 cell lines derived from premalignant human breast epithelial MCF10AT cells. Breast Cancer Res Treat. 2001;65(2):101–10.

    Article  PubMed  CAS  Google Scholar 

  224. Rak JW, McEachern D, Miller FR. Sequential alteration of peanut agglutinin binding-glycoprotein expression during progression of murine mammary neoplasia. Br J Cancer. 1992;65(5):641–8.

    Article  PubMed  CAS  Google Scholar 

  225. Tsuji T, Ibaragi S, Shima K, Hu MG, Katsurano M, Sasaki A, et al. Epithelial-mesenchymal transition induced by growth suppressor p12CDK2-AP1 promotes tumor cell local invasion but suppresses distant colony growth. Cancer Res. 2008;68(24):10377–86.

    Article  PubMed  CAS  Google Scholar 

  226. McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince TA, et al. Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell. 2008;133(6):994–1005.

    Article  PubMed  CAS  Google Scholar 

  227. Kim MY, Oskarsson T, Acharyya S, Nguyen DX, Zhang XH, Norton L, et al. Tumor self-seeding by circulating cancer cells. Cell. 2009;139(7):1315–26.

    Article  PubMed  Google Scholar 

  228. Nomura M, Li E. Smad2 role in mesoderm formation, left-right patterning and craniofacial development. Nature. 1998;393(6687):786–90.

    PubMed  CAS  Google Scholar 

  229. Waldrip WR, Bikoff EK, Hoodless PA, Wrana JL, Robertson EJ. Smad2 signaling in extraembryonic tissues determines anterior-posterior polarity of the early mouse embryo. Cell. 1998;92(6):797–808.

    Article  PubMed  CAS  Google Scholar 

  230. Zhu Y, Richardson JA, Parada LF, Graff JM. Smad3 mutant mice develop metastatic colorectal cancer. Cell. 1998;94(6):703–14.

    Article  PubMed  CAS  Google Scholar 

  231. Datto MB, Frederick JP, Pan L, Borton AJ, Zhuang Y, Wang XF. Targeted disruption of Smad3 reveals an essential role in transforming growth factor β-mediated signal transduction. Mol Cell Biol. 1999;19(4):2495–504.

    PubMed  CAS  Google Scholar 

  232. Yang X, Letterio JJ, Lechleider RJ, Chen L, Hayman R, Gu H, et al. Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF-β. EMBO J. 1999;18(5):1280–91.

    Article  PubMed  CAS  Google Scholar 

  233. Kretschmer A, Moepert K, Dames S, Sternberger M, Kaufmann J, Klippel A. Differential regulation of TGF-βsignaling through Smad2, Smad3 and Smad4. Oncogene. 2003;22(43):6748–63.

    Article  PubMed  CAS  Google Scholar 

  234. Dzwonek J, Preobrazhenska O, Cazzola S, Conidi A, Schellens A, van Dinther M, et al. Smad3 is a key nonredundant mediator of transforming growth factor βsignaling in Nme mouse mammary epithelial cells. Mol Cancer Res. 2009;7(8):1342–53.

    Article  PubMed  CAS  Google Scholar 

  235. Piek E, Ju WJ, Heyer J, Escalante-Alcalde D, Stewart CL, Weinstein M, et al. Functional characterization of transforming growth factor β signaling in Smad2- and Smad3-deficient fibroblasts. J Biol Chem. 2001;276(23):19945–53.

    Article  PubMed  CAS  Google Scholar 

  236. 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(1):9–33.

    Article  PubMed  CAS  Google Scholar 

  237. Petersen M, Pardali E, van der Horst G, Cheung H, van den Hoogen C, van der Pluijm G, et al. Smad2 and Smad3 have opposing roles in breast cancer bone metastasis by differentially affecting tumor angiogenesis. Oncogene. 2010;29(9):1351–61.

    Article  PubMed  CAS  Google Scholar 

  238. Tian F, DaCosta Byfield S, Parks WT, Yoo S, Felici A, Tang B, et al. Reduction in Smad2/3 signaling enhances tumorigenesis but suppresses metastasis of breast cancer cell lines. Cancer Res. 2003;63(23):8284–92.

    PubMed  CAS  Google Scholar 

  239. Tang B, Vu M, Booker T, Santner SJ, Miller FR, Anver MR, et al. TGF-βswitches from tumor suppressor to prometastatic factor in a model of breast cancer progression. J Clin Invest. 2003;112(7):1116–24.

    PubMed  CAS  Google Scholar 

  240. Tian F, Byfield SD, Parks WT, Stuelten CH, Nemani D, Zhang YE, et al. Smad-binding defective mutant of transforming growth factor β type I receptor enhances tumorigenesis but suppresses metastasis of breast cancer cell lines. Cancer Res. 2004;64(13):4523–30.

    Article  PubMed  CAS  Google Scholar 

  241. Korpal M, Yan J, Lu X, Xu S, Lerit DA, Kang Y. Imaging transforming growth factor-β signaling dynamics and therapeutic response in breast cancer bone metastasis. Nat Med. 2009;15(8):960–6.

    Article  PubMed  CAS  Google Scholar 

  242. Giampieri S, Manning C, Hooper S, Jones L, Hill CS, Sahai E. Localized and reversible TGFβ signalling switches breast cancer cells from cohesive to single cell motility. Nat Cell Biol. 2009;11(11):1287–96.

    Article  PubMed  CAS  Google Scholar 

  243. Giampieri S, Pinner S, Sahai E. Intravital imaging illuminates transforming growth factor β signaling switches during metastasis. Cancer Res. 2010;70(9):3435–9.

    Article  PubMed  CAS  Google Scholar 

  244. Butcher DT, Alliston T, Weaver VM. A tense situation: forcing tumour progression. Nat Rev Cancer. 2009;9(2):108–22.

    Article  PubMed  CAS  Google Scholar 

  245. Paszek MJ, Weaver VM. The tension mounts: mechanics meets morphogenesis and malignancy. J Mammary Gland Biol Neoplasia. 2004;9(4):325–42.

    Article  PubMed  Google Scholar 

  246. Kumar S, Weaver VM. Mechanics, malignancy, and metastasis: the force journey of a tumor cell. Cancer Metastasis Rev. 2009;28(1–2):113–27.

    Article  PubMed  Google Scholar 

  247. Sugahara KN, Teesalu T, Karmali PP, Kotamraju VR, Agemy L, Girard OM, et al. Tissue-penetrating delivery of compounds and nanoparticles into tumors. Cancer Cell. 2009;16(6):510–20.

    Article  PubMed  CAS  Google Scholar 

  248. Kota J, Chivukula RR, O’Donnell KA, Wentzel EA, Montgomery CL, Hwang HW, et al. Therapeutic microRNA delivery suppresses tumorigenesis in a murine liver cancer model. Cell. 2009;137(6):1005–17.

    Article  PubMed  CAS  Google Scholar 

  249. Tsuda N, Ishiyama S, Li Y, Ioannides CG, Abbruzzese JL, Chang DZ. Synthetic microRNA designed to target glioma-associated antigen 1 transcription factor inhibits division and induces late apoptosis in pancreatic tumor cells. Clin Cancer Res. 2006;12(21):6557–64.

    Article  PubMed  CAS  Google Scholar 

  250. Bhardwaj A, Singh S, Singh AP. MicroRNA-based cancer therapeutics: big hope from small RNAs. Mol Cell Pharmacol. 2010;2(5):213–9.

    PubMed  CAS  Google Scholar 

  251. Weiler J, Hunziker J, Hall J. Anti-miRNA oligonucleotides (AMOs): ammunition to target miRNAs implicated in human disease? Gene Ther. 2006;13(6):496–502.

    Article  PubMed  CAS  Google Scholar 

  252. Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, et al. Silencing of microRNAs in vivo with ‘antagomirs’. Nature. 2005;438(7068):685–9.

    Article  PubMed  CAS  Google Scholar 

  253. Gumireddy K, Young DD, Xiong X, Hogenesch JB, Huang Q, Deiters A. Small-molecule inhibitors of microrna miR-21 function. Angew Chem Int Ed Engl. 2008;47(39):7482–4.

    Article  PubMed  CAS  Google Scholar 

  254. Ebert MS, Neilson JR, Sharp PA. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat Methods. 2007;4(9):721–6.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Schiemann Laboratory for critical comments and reading of the manuscript. W.P.S. was supported by grants from the National Institutes of Health (CA129359), the Komen Foundation (BCTR0706967), and the Department of Defense (BC084651), while M.A.T. was supported by the Department of Defense (BC093128).

Financial Support

W.P.S. was supported by grants from the National Institutes of Health (CA129359), the Komen Foundation (BCTR0706967), and the Department of Defense (BC084651), while M.A.T. was supported by the Department of Defense (BC093128).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to William P. Schiemann.

Additional information

Jenny G. Parvani and Molly A. Taylor were equal contributors to this work.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Parvani, J.G., Taylor, M.A. & Schiemann, W.P. Noncanonical TGF-β Signaling During Mammary Tumorigenesis. J Mammary Gland Biol Neoplasia 16, 127–146 (2011). https://doi.org/10.1007/s10911-011-9207-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10911-011-9207-3

Keywords

Navigation