Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-8kt4b Total loading time: 0 Render date: 2024-06-24T12:16:06.399Z Has data issue: false hasContentIssue false

11 - The Continuum of Epithelial Mesenchymal Transition – Implication of Hybrid States for Migration and Survival in Development and Cancer

from VARIOUS PROPERTIES OF CANCER CELLS

Published online by Cambridge University Press:  05 June 2012

Lilian Soon
Affiliation:
The University of Sydney, Australia
Anthony Tachtsidis
Affiliation:
University of Melbourne, Australia
Sandra Fok
Affiliation:
The University of Sydney, Australia
Elizabeth D. Williams
Affiliation:
Monash University, Australia
Donald F. Newgreen
Affiliation:
Royal Children's Hospital, Australia
Erik W. Thompson
Affiliation:
University of Melbourne, Australia
David Lyden
Affiliation:
Weill Cornell Medical College, New York
Danny R. Welch
Affiliation:
Weill Cornell Medical College, New York
Bethan Psaila
Affiliation:
Imperial College of Medicine, London
Get access

Summary

EMT AND CELL MIGRATION – EMBRYONIC NECESSITIES CO-OPTED BY INVASIVE CANCER

The concept of the epithelial–mesenchymal transition (EMT) originated from studies of events in development, particularly those preceding the onset of cell migration [1]. These were initially brought together and popularized by the efforts of the late Elizabeth Hay and colleagues [2, 3]. One of the most intensively studied examples of EMT and cell migration is the generation of migratory neural crest mesenchyme from the neurectodermal epithelium [4] (Figure 11.1). These cells go on to form the autonomic and sensory nervous systems. Even prior to this, the emergence of the primary mesenchyme from the epithelial epiblast during gastrulation (the first EMT) results in the formation of highly motile cells that are critical to the development of the body plan [1] (Figure 11.2A). Further EMTs occur in other epithelia after the neural crest EMT to generate the cells that form muscle, bone, and connective tissues (Figure 11.2B). Such cellular plasticity is fundamental to embryological development and is regulated largely at the transcriptional level. Various transcriptional repressors of E-cadherin (and other cadherins), such as Snail (Snail 1), Slug (Snail 2), Twist, Zeb1 (δEF1), Zeb2 (SIP), and E47/E12, regulate EMT in developmental system (reviewed in [5]).

Commitment to lineage differentiation in normal cells is more pliant than first thought, and cellular transition is emerging as a major mechanism of adult tissue homeostasis [6].

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 117 - 130
Publisher: Cambridge University Press
Print publication year: 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Shook, D, Keller, R (2003) Mechanisms, mechanics and function of epithelial-mesenchymal transitions in early development. Mech Dev. 120: 1351–1383.CrossRefGoogle ScholarPubMed
Trelstad, R (2004) The extracellular matrix in development and regeneration. An interview with Elizabeth D. Hay. Int J Dev Biol. 48: 687–694.CrossRefGoogle Scholar
Hay, ED (2005) The mesenchymal cell, its role in the embryo, and the remarkable signaling mechanisms that create it. Dev Dyn. 233: 706–720.CrossRefGoogle Scholar
Duband, JL, Monier, F, Delannet, M, Newgreen, D (1995) Epithelium-mesenchyme transition during neural crest development. Acta Anat (Basel). 154: 63–78.CrossRefGoogle ScholarPubMed
Peinado, H, Olmeda, D, Cano, A (2007) Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype?Nat Rev Cancer. 7: 415–428.CrossRefGoogle ScholarPubMed
Prindull, G, Zipori, D (2004) Environmental guidance of normal and tumor cell plasticity: epithelial mesenchymal transitions as a paradigm. Blood. 103: 2892–2899.CrossRefGoogle Scholar
Huber, MA, Kraut, N, Beug, H (2005) Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol. 17: 548–558.CrossRefGoogle ScholarPubMed
Birchmeier, W, Behrens, J (1994) Cadherin expression in carcinomas: role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta. 1198: 11–26.Google ScholarPubMed
Thiery, JP (2003) Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol. 15: 740–746.CrossRefGoogle ScholarPubMed
Thompson, EW, Newgreen, DF, Tarin, D (2005) Carcinoma invasion and metastasis: a role for epithelial-mesenchymal transition?Cancer Res. 65: 5991–5995; discussion 5995.CrossRefGoogle ScholarPubMed
Tarin, D, Thompson, EW, Newgreen, DF (2005) The fallacy of epithelial mesenchymal transition in neoplasia. Cancer Res. 65: 5996–6000; discussion 6000–6001.CrossRefGoogle ScholarPubMed
Xue, C, Plieth, D, Venkov, C, Xu, C, Neilson, EG (2003) The gatekeeper effect of epithelial-mesenchymal transition regulates the frequency of breast cancer metastasis. Cancer Res. 63: 3386–3394.Google ScholarPubMed
Trimboli, AJ, Fukino, K, Bruin, A et al. (2008) Direct evidence for epithelial-mesenchymal transitions in breast cancer. Cancer Res. 68: 937–945.CrossRefGoogle ScholarPubMed
Charafe-Jauffret, E, Ginestier, C, Monville, F et al. (2006) Gene expression profiling of breast cell lines identifies potential new basal markers. Oncogene. 25: 2273–2284.CrossRefGoogle ScholarPubMed
Neve, RM, Chin, K, Fridlyand, J et al. (2006) A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 10: 515–527.CrossRefGoogle Scholar
Blick, T, Widodo, E, Hugo, H et al. (2008) Epithelial mesenchymal transition traits in human breast cancer cell lines. Clin Exp Metastasis. 25: 629–642.CrossRefGoogle ScholarPubMed
Brabletz, T, Jung, A, Spaderna, S, Hlubek, F, Kirchner, T (2005) Opinion: migrating cancer stem cells – an integrated concept of malignant tumour progression. Nat Rev Cancer. 5: 744–749.CrossRefGoogle ScholarPubMed
Shipitsin, M, Campbell, LL, Argani, P et al. (2007) Molecular definition of breast tumor heterogeneity. Cancer Cell. 11: 259–273.CrossRefGoogle ScholarPubMed
Mani, SA, Guo, W, Liao, MJ et al. (2008) The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 133: 704–715.CrossRefGoogle ScholarPubMed
Hollier, BG, Evans, K, Mani, SA (2009) The epithelial-to-mesenchymal transition and cancer stem cells: a coalition against cancer therapies. J Mammary Gland Biol Neoplasia. 14: 29–43.CrossRefGoogle ScholarPubMed
Kulesa, PM, Fraser, SE (2000) In ovo time-lapse analysis of chick hindbrain neural crest cell migration shows cell interactions during migration to the branchial arches. Development. 127: 1161–1172.Google ScholarPubMed
Young, HM, Bergner, AJ, Anderson, RB et al. (2004) Dynamics of neural crest-derived cell migration in the embryonic mouse gut. Dev Biol. 270: 455–473.CrossRefGoogle ScholarPubMed
Friedl, P, Hegerfeldt, Y, Tusch, M (2004) Collective cell migration in morphogenesis and cancer. Int J Dev Biol. 48: 441–449.CrossRefGoogle ScholarPubMed
Thompson, EW, Williams, ED (2008) EMT and MET in carcinoma – clinical observations, regulatory pathways and new models. Clin Exp Metastasis. 25: 591–592.CrossRefGoogle ScholarPubMed
Zavadil, J, Haley, J, Kalluri, R, Muthuswamy, SK, Thompson, E (2008) Epithelial-mesenchymal transition. Cancer Res. 68: 9574–9577.CrossRefGoogle ScholarPubMed
Hugo, H, Ackland, ML, Blick, T et al. (2007) Epithelial-mesenchymal and mesenchymal-epithelial transitions in carcinoma progression. J Cell Physiol. 213: 374–383.CrossRefGoogle ScholarPubMed
Yang, J, Weinberg, RA (2008) Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 14: 818–829.CrossRefGoogle ScholarPubMed
Thiery, JP, Sleeman, JP (2006) Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 7: 131–142.CrossRefGoogle ScholarPubMed
Korsching, E, Packeisen, J, Liedtke, C et al. (2005) The origin of vimentin expression in invasive breast cancer: epithelial-mesenchymal transition, myoepithelial histogenesis or histogenesis from progenitor cells with bilinear differentiation potential?J Pathol. 206: 451–457.CrossRefGoogle ScholarPubMed
Sarrio, D, Rodriguez-Pinilla, SM, Hardisson, D, Cano, A, Moreno-Bueno, G, Palacios, J (2008) Epithelial-mesenchymal transition in breast cancer relates to the basal-like phenotype. Cancer Res. 68: 989–997.CrossRefGoogle ScholarPubMed
Come, C, Arnoux, V, Bibeau, F, Savagner, P (2004) Roles of the transcription factors snail and slug during mammary morphogenesis and breast carcinoma progression. J Mammary Gland Biol Neoplasia. 9: 183–193.CrossRefGoogle ScholarPubMed
Come, C, Magnino, F, Bibeau, F et al. (2006) Snail and slug play distinct roles during breast carcinoma progression. Clin Cancer Res. 12: 5395–5402.CrossRefGoogle ScholarPubMed
Aigner, K, Dampier, B, Descovich, L et al. (2007) The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene. 26: 6979–6988.CrossRefGoogle ScholarPubMed
Eger, A, Aigner, K, Sonderegger, S et al. (2005) DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene. 24: 2375–2385.CrossRefGoogle ScholarPubMed
Blanco, MJ, Moreno-Bueno, G, Sarrio, D et al. (2002) Correlation of Snail expression with histological grade and lymph node status in breast carcinomas. Oncogene. 21: 3241–3246.CrossRefGoogle ScholarPubMed
Elloul, S, Elstrand, MB, Nesland, JM et al. (2005) Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease aggressiveness in metastatic ovarian and breast carcinoma. Cancer. 103: 1631–1643.CrossRefGoogle ScholarPubMed
Martin, TA, Goyal, A, Watkins, G, Jiang, WG (2005) Expression of the transcription factors Snail, Slug, and Twist and their clinical significance in human breast cancer. Ann Surg Oncol. 12: 488–496.CrossRefGoogle ScholarPubMed
Mironchik, Y, Winnard, PT Jr, Vesuna, F et al. (2005) Twist overexpression induces in vivo angiogenesis and correlates with chromosomal instability in breast cancer. Cancer Res. 65: 10801–10809.CrossRefGoogle ScholarPubMed
Turashvili, G, Bouchal, J, Baumforth, K et al. (2007) Novel markers for differentiation of lobular and ductal invasive breast carcinomas by laser microdissection and microarray analysis. BMC Cancer. 7: 55.CrossRefGoogle ScholarPubMed
Wu, X, Chen, H, Parker, B et al. (2006) HOXB7, a homeodomain protein, is overexpressed in breast cancer and confers epithelial-mesenchymal transition. Cancer Res. 66: 9527–9534.CrossRefGoogle ScholarPubMed
Baumgart, E, Cohen, MS, Neto, BS et al. (2007) Identification and prognostic significance of an epithelial-mesenchymal transition expression profile in human bladder tumors. Clin Cancer Res. 13: 1685–1694.CrossRefGoogle ScholarPubMed
Koksal, IT, Ates, M, Danisman, A et al. (2006) Reduced E-cadherin and alpha-catenin expressions have no prognostic role in bladder carcinoma. Pathol Oncol Res. 12: 13–19.CrossRefGoogle ScholarPubMed
Sanchez-Carbayo, M, Socci, ND, Charytonowicz, E et al. (2002) Molecular profiling of bladder cancer using cDNA microarrays: defining histogenesis and biological phenotypes. Cancer Res. 62: 6973–6980.Google ScholarPubMed
Shariat, SF, Matsumoto, K, Casella, R, Jian, W, Lerner, SP (2005) Urinary levels of soluble E-cadherin in the detection of transitional cell carcinoma of the urinary bladder. Eur Urol. 48: 69–76.CrossRefGoogle ScholarPubMed
Serdar, A, Turhan, C, Soner, G et al. (2005) The prognostic importance of E-cadherin and p53 gene expression in transitional bladder carcinoma patients. Int Urol Nephrol. 37: 485–492.CrossRefGoogle ScholarPubMed
Szekely, E, Torok, V, Szekely, T, Riesz, P, Romics, I (2006) E-cadherin expression in transitional cell carcinomas. Pathol Oncol Res. 12: 73–77.CrossRefGoogle ScholarPubMed
Otto, T, Bex, A, Schmidt, U, Raz, A, Rubben, H (1997) Improved prognosis assessment for patients with bladder carcinoma. Am J Pathol. 150: 1919–1923.Google ScholarPubMed
Fidler, IJ (2002) Critical determinants of metastasis. Semin Cancer Biol. 12: 89–96.CrossRefGoogle Scholar
Barrallo-Gimeno, A, Nieto, MA (2005) The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development. 132: 3151–3161.CrossRefGoogle ScholarPubMed
Moody, SE, Perez, D, Pan, TC et al. (2005) The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell. 8: 197–209.CrossRefGoogle ScholarPubMed
Willipinski-Stapelfeldt, B, Riethdorf, S, Assmann, V et al. (2005) Changes in cytoskeletal protein composition indicative of an epithelial-mesenchymal transition in human micrometastatic and primary breast carcinoma cells. Clin Cancer Res. 11: 8006–8014.CrossRefGoogle ScholarPubMed
Frisch, SM, Screaton, RA (2001) Anoikis mechanisms. Curr Opin Cell Biol. 13: 555–562.CrossRefGoogle ScholarPubMed
Przybylo, JA, Radisky, DC (2007) Matrix metalloprote-inase-induced epithelial-mesenchymal transition: Tumor progression at Snail's pace. Int J Biochem Cell Biol. 39: 1082–1088.CrossRefGoogle ScholarPubMed
Thomson, S, Buck, E, Petti, F et al. (2005) Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. Cancer Res. 65: 9455–9462.CrossRefGoogle ScholarPubMed
Thiery, JP (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2: 442–454.CrossRefGoogle ScholarPubMed
Tothill, RW, Kowalczyk, A, Rischin, D et al. (2005) An expression-based site of origin diagnostic method designed for clinical application to cancer of unknown origin. Cancer Res. 65: 4031–4040.CrossRefGoogle ScholarPubMed
Weigelt, B, Glas, AM, Wessels, LF et al. (2003) Gene expression profiles of primary breast tumors maintained in distant metastases. Proc Natl Acad Sci USA. 100: 15901–15905.CrossRefGoogle ScholarPubMed
Weigelt, B, Hu, Z, He, X et al. (2005) Molecular portraits and 70-gene prognosis signature are preserved throughout the metastatic process of breast cancer. Cancer Res. 65: 9155–9158.CrossRefGoogle ScholarPubMed
Chaffer, CL, Brennan, JP, Slavin, JL et al. (2006) Mesenchymal-to-epithelial transition facilitates bladder cancer metastasis: role of fibroblast growth factor receptor-2. Cancer Res. 66: 11271–11278.CrossRefGoogle ScholarPubMed
Summers, JL, Falor, WH, Ward, RM, Fiorito, J, Brezler, MR, Abreu, AJ (1983) Identical genetic profiles in primary and metastatic bladder tumors. J Urol. 129: 827–828.CrossRefGoogle ScholarPubMed
Chaffer, CL, Dopheide, B, McCulloch, DR et al. (2005) Upregulated MT1-MMP/TIMP-2 axis in the TSU-Pr1-B1/B2 model of metastatic progression in transitional cell carcinoma of the bladder. Clin Exp Metastasis. 22: 115–125.CrossRefGoogle ScholarPubMed
Yates, CC, Shepard, CR, Stolz, DB, Wells, A (2007) Co-culturing human prostate carcinoma cells with hepatocytes leads to increased expression of E-cadherin. Br J Cancer. 96: 1246–1252.CrossRefGoogle ScholarPubMed
Ackland, ML, Newgreen, DF, Fridman, M et al. (2003) Epidermal growth factor-induced epithelio-mesenchymal transition in human breast carcinoma cells. Lab Invest. 83: 435–448.CrossRefGoogle ScholarPubMed
Lee, JM, Dedhar, S, Kalluri, R, Thompson, EW (2006) The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol. 172: 973–981.CrossRefGoogle ScholarPubMed
Klymkowsky, MW, Savagner, P (2009) Epithelial-mesenchymal transition: a cancer researcher's conceptual friend and foe. Am J Pathol. 174: 1588–1593.CrossRefGoogle ScholarPubMed
Christiansen, JJ, Rajasekaran, AK (2006) Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer Res. 66: 8319–8326.CrossRefGoogle ScholarPubMed
Moustakas, A, Heldin, CH (2007) Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 98: 1512–1520.CrossRefGoogle ScholarPubMed
Batlle, E, Sancho, E, Franci, C et al. (2000) The transcription factor Snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol. 2: 84–89.CrossRefGoogle ScholarPubMed
Cano, A, Perez-Moreno, MA, Rodrigo, I et al. (2000) The transcription factor Snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol. 2: 76–83.CrossRefGoogle ScholarPubMed
Bolos, V, Peinado, H, Perez-Moreno, MA, Fraga, MF, Esteller, M, Cano, A (2003) The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci. 116: 499–511.CrossRefGoogle ScholarPubMed
Jeanes, A, Gottardi, CJ, Yap, AS (2008) Cadherins and cancer: how does cadherin dysfunction promote tumor progression?Oncogene. 27: 6920–6929.CrossRefGoogle ScholarPubMed
Vestweber, D, Kemler, R, Ekblom, P (1985) Cell-adhesion molecule uvomorulin during kidney development. Dev Biol. 112: 213–221.CrossRefGoogle ScholarPubMed
Dahl, U, Sjodin, A, Larue, L et al. (2002) Genetic dissection of cadherin function during nephrogenesis. Mol Cell Biol. 22: 1474–1487.CrossRefGoogle ScholarPubMed
Suyama, K, Shapiro, I, Guttman, M, Hazan, RB (2002) A signaling pathway leading to metastasis is controlled by N-cadherin and the FGF receptor. Cancer Cell. 2: 301–314.CrossRefGoogle ScholarPubMed
Halbleib, JM, Nelson, WJ (2006) Cadherins in development: cell adhesion, sorting, and tissue morphogenesis. Genes Dev. 20: 3199–3214.CrossRefGoogle ScholarPubMed
Reynolds, AB, Roczniak-Ferguson, A (2004) Emerging roles for p120-catenin in cell adhesion and cancer. Oncogene. 23: 7947–7956.CrossRefGoogle ScholarPubMed
Maeda, M, Johnson, E, Mandal, SH et al. (2006) Expression of inappropriate cadherins by epithelial tumor cells promotes endocytosis and degradation of E-cadherin via competition for p120(ctn). Oncogene. 25: 4595–4604.CrossRefGoogle Scholar
Derycke, L, Wever, O, Stove, V et al. (2006) Soluble N-cadherin in human biological fluids. Int J Cancer. 119: 2895–2900.CrossRefGoogle ScholarPubMed
Johnson, E, Theisen, CS, Johnson, KR, Wheelock, MJ (2004) R-cadherin influences cell motility via Rho family GTPases. J Biol Chem. 279: 31041–31049.CrossRefGoogle ScholarPubMed
Friedl, P, Wolf, K (2003) Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 3: 362–374.CrossRefGoogle ScholarPubMed
Carragher, NO (2009) Profiling distinct mechanisms of tumour invasion for drug discovery: imaging adhesion, signalling and matrix turnover. Clin Exp Metastasis. 26: 381–397.CrossRefGoogle ScholarPubMed
Tchou-Wong, KM, Fok, SYY, Rubin, JS et al. (2006) Rapid chemokinetic movement and the invasive potential of lung cancer cells; a functional molecular study. BMC Cancer. 6: 151.Google Scholar
Sanz-Moreno, V, Gadea, G, Ahn, J et al. (2008) Rac activation and inactivation control plasticity of tumor cell movement. Cell. 135: 510–523.CrossRefGoogle ScholarPubMed
Sahai, E, Marshall, CJ (2003) Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol. 5: 711–719.CrossRefGoogle ScholarPubMed
Gadea, G, Sanz-Moreno, V, Self, A, Godi, A, Marshall, CJ (2008) DOCK10-mediated Cdc42 activation is necessary for amoeboid invasion of melanoma cells. Curr Biol. 18: 1456–1465.CrossRefGoogle ScholarPubMed
Fackler, OT, Grosse, R (2008) Cell motility through plasma membrane blebbing. J Cell Biol. 181: 879–884.CrossRefGoogle ScholarPubMed
Trinkaus, JP (1973) Surface activity and locomotion of Fundulus deep cells during blastula and gastrula stages. Dev Biol. 30: 69–103.CrossRefGoogle ScholarPubMed
Lammermann, T, Bader, BL, Monkley, SJ et al. (2008) Rapid leukocyte migration by integrin-independent flowing and squeezing. Nature. 453: 51–55.CrossRefGoogle ScholarPubMed
Liotta, , Mandler, R, Murano, G et al. (1996) Tumor cell autocrine motility factor. Proc Natl Acad Sci USA. 83: 3302–3306.CrossRefGoogle Scholar
Muller, A, Homey, B, Soto, H et al. (2001) Involvement of chemokine receptors in breast cancer metastasis. Nature. 410: 50–56.CrossRefGoogle ScholarPubMed
Kohn, EC, Francis, EA, Liotta, , Schiffmann, E (1990) Heterogeneity of the motility responses in malignant tumor cells: a biological basis for the diversity and homing of metastatic cells. Int J Cancer. 46: 287–292.CrossRefGoogle ScholarPubMed
Zigmond, SH (1977) Ability of polymorphonuclear leukocytes to orient in gradients of chemotactic factors. J Cell Biol. 75: 606–616.CrossRefGoogle ScholarPubMed
Zicha, D, Dunn, G, Jones, G (1997) Analyzing chemotaxis using the Dunn direct-viewing chamber. Methods Mol Biol. 75: 449–457.Google ScholarPubMed
Fok, S, Domachuk, P, Rosengarten, G et al. (2008) Planar microfluidic chamber for generation of stable and steep chemoattractant gradients. Biophys J. 95: 1523–1530.CrossRefGoogle ScholarPubMed
Price, JT, Thompson, EW (1999) Models for studying cellular invasion of basement membranes. Methods Mol Biol. 129: 231–249.Google ScholarPubMed
Newgreen, DF, Murphy, M (2000) Neural crest cell outgrowth cultures and the analysis of cell migration. Methods Mol Biol. 137: 201–211.Google ScholarPubMed
Ahlstrom, JD, Erickson, CA (2009) The neural crest epithelial-mesenchymal transition in 4D: a ‘tail’ of multiple non-obligatory cellular mechanisms. Development. 136: 1801–1812.CrossRefGoogle ScholarPubMed
Sauka-Spengler, T, Bronner-Fraser, M (2008) A gene regulatory network orchestrates neural crest formation. Nat Rev Mol Cell Biol. 9: 557–568.CrossRefGoogle ScholarPubMed
Carmona-Fontaine, C, Matthews, HK, Kuriyama, S et al. (2008) Contact inhibition of locomotion in vivo controls neural crest directional migration. Nature. 456: 957–961.CrossRefGoogle ScholarPubMed
Neri, A, Welch, D, Kawaguchi, T, Nicolson, GL (1982) Development and biologic properties of malignant cell sublines and clones of a spontaneously metastasizing rat mammary adenocarcinoma. J Natl Cancer Inst. 68: 507–517.Google ScholarPubMed
Edmonds, BT, Wyckoff, J, Yeung, YG et al. (1996) Elongation factor-1 alpha is an overexpressed actin binding protein in metastatic rat mammary adenocarcinoma. J Cell Sci. 109(Pt 11): 2705–2714.Google ScholarPubMed
Kaufmann, AM, Khazaie, K, Wiedemuth, M et al. (1994) Expression of epidermal growth factor receptor correlates with metastatic potential of 13762NF rat mammary adenocarcinoma cells. Int J Oncol. 4: 1149–1155.Google ScholarPubMed
Wyckoff, JB, Insel, L, Khazaie, K, Lichtner, RB, Condeelis, JS, Segall, JE (1998) Suppression of ruffling by the EGF receptor in chemotactic cells. Exp Cell Res. 242: 100–109.CrossRefGoogle ScholarPubMed
Lebret, SC, Newgreen, DF, Thompson, EW, Ackland, ML (2007) Induction of epithelial to mesenchymal transition in PMC42-LA human breast carcinoma cells by carcinoma-associated fibroblast secreted factors. Breast Cancer Res. 9: R19.CrossRefGoogle ScholarPubMed
Lebret, SC, Newgreen, DF, Waltham, MC, Price, JT, Thompson, EW, Ackland, ML (2006) Myoepithelial molecular markers in human breast carcinoma PMC42-LA cells are induced by extracellular matrix and stromal cells. In Vitro Cell Dev Biol Anim. 42: 298–307.Google ScholarPubMed
Whitehead, RH, Bertoncello, I, Webber, LM, Pedersen, JS (1983) A new human breast carcinoma cell line (PMC42) with stem cell characteristics. I. Morphologic characterization. J Natl Cancer Inst. 70: 649–661.Google ScholarPubMed
Whitehead, RH, Monaghan, P, Webber, LM, Bertoncello, I, Vitali, AA (1983) A new human breast carcinoma cell line (PMC42) with stem cell characteristics. II. Characterization of cells growing as organoids. J Natl Cancer Inst. 71: 1193–1203.Google ScholarPubMed
Whitehead, RH, Quirk, SJ, Vitali, AA, Funder, JW, Sutherland, RL, Murphy, LC (1984) A new human breast carcinoma cell line (PMC42) with stem cell characteristics. III. Hormone receptor status and responsiveness. J Natl Cancer Inst. 73: 643–648.Google ScholarPubMed
Monaghan, P, Whitehead, RH, Perusinghe, N, O'Hare, MJ (1985) An immunocytochemical and ultrastructural study of heterogeneity in the human breast carcinoma cell line PMC42. Cancer Res. 45: 5088–5097.Google ScholarPubMed
Ackland, ML, Michalczyk, A, Whitehead, RH (2001) PMC42, A novel model for the differentiated human breast. Exp Cell Res. 263: 14–22.CrossRefGoogle ScholarPubMed
Lo, HW, Hsu, SC, Xia, W et al. (2007) Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 67: 9066–9076.CrossRefGoogle ScholarPubMed
Vantyghem, SA, Allan, AL, Postenka, CO et al. (2005) A new model for lymphatic metastasis: development of a variant of the MDA-MB-468 human breast cancer cell line that aggressively metastasizes to lymph nodes. Clin Exp Metastasis. 22: 351–361.CrossRefGoogle ScholarPubMed
Rodenhiser, DI, Andrews, J, Kennette, W et al. (2008) Epigenetic mapping and functional analysis in a breast cancer metastasis model using whole-genome promoter tiling microarrays. Breast Cancer Res. 10: R62.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×