Characterization of Spontaneous and TGF-β-Induced Cell Motility of Primary Human Normal and Neoplastic Mammary Cells In Vitro Using Novel Real-Time Technology

The clinical complications derived from metastatic disease are responsible for the majority of all breast cancer related deaths. Since cell migration and invasion are a prerequisite for metastasis their assessment in patient cancer cells in vitro may have prognostic value for the tumor's metastatic capacity. We employed real-time cell analysis (RTCA) on the xCELLigence DP system to determine in vitro motility of patient-derived primary human breast cancer epithelial cells (HBCEC). Initially, the RTCA assay was validated using established human breast cancer cell lines with either an invasive (MDA-MB-231, MDA-MB-435s) or a non-invasive phenotype (MCF-7, MDA-MB-468), and primary NSCLC cells (Tu459). Previous standard assays of cell migration/invasion revealed that only MDA-MB-231, −435s, and Tu459 cells exhibited spontaneous and TGF-β1-stimulated migration and invasion through a Matrigel barrier. In the present study, the TGF-β1-stimulated activities could be blocked by SB431542, a potent kinase inhibitor of the TGF-β type I receptor ALK5. Application of the RTCA assay to patient-derived tumor cells showed that 4/4 primary HBCEC and primary NSCLC cells, but not normal human mammary epithelial cells (HMEC), displayed high spontaneous migratory and invasive activity which correlated with higher MMP-2 expression and uPA protein levels in HBCEC compared to HMEC. Upon treatment with TGF-β1, HBCEC exhibited morphologic and gene regulatory alterations indicative of epithelial-to-mesenchymal transition. However, exclusively the invasive but not the migratory activity of HBCEC was further enhanced by TGF-β1. This indicates the requirement for molecular, e.g. integrin interactions with Matrigel components in HBCEC in order to become responsive to pro-invasive TGF-β effects. Together, these results show for the first time that tumorigenic HBCEC but not normal HMEC possess a strong basal migratory as well as a basal and TGF-β1-inducible invasive potential. These findings qualify the RTCA assay as an in vitro migration/invasion testing system for patient-specific primary breast cancer cells.

[1]  Olivier De Wever,et al.  Comparative Analysis of Dynamic Cell Viability, Migration and Invasion Assessments by Novel Real-Time Technology and Classic Endpoint Assays , 2012, PloS one.

[2]  R. Hass,et al.  Mesenchymal stem cells as all-round supporters in a normal and neoplastic microenvironment , 2012, Cell Communication and Signaling.

[3]  H. Lehnert,et al.  The Src family kinase inhibitors PP2 and PP1 effectively block TGF-beta1-induced cell migration and invasion in both established and primary carcinoma cells , 2012, Cancer Chemotherapy and Pharmacology.

[4]  Stephanie Alexander,et al.  Cancer Invasion and the Microenvironment: Plasticity and Reciprocity , 2011, Cell.

[5]  Hendrik Lehnert,et al.  Interaction of tumor cells with the microenvironment , 2011, Cell Communication and Signaling.

[6]  P. Dijke,et al.  The TGF-β/Smad pathway induces breast cancer cell invasion through the up-regulation of matrix metalloproteinase 2 and 9 in a spheroid invasion model system , 2011, Breast Cancer Research and Treatment.

[7]  H. Lehnert,et al.  Differential roles of Smad2 and Smad3 in the regulation of TGF-β1-mediated growth inhibition and cell migration in pancreatic ductal adenocarcinoma cells: control by Rac1 , 2011, Molecular Cancer.

[8]  S. Böhm,et al.  Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC , 2011, Cell Communication and Signaling.

[9]  R. Hass,et al.  Extracellular signals in young and aging breast epithelial cells and possible connections to age-associated breast cancer development , 2011, Mechanisms of Ageing and Development.

[10]  P. ten Dijke,et al.  TGF-β Signaling in Breast Cancer Cell Invasion and Bone Metastasis , 2011, Journal of Mammary Gland Biology and Neoplasia.

[11]  M. Bollet,et al.  Management of Phyllodes Breast Tumors , 2011, The breast journal.

[12]  S. Sebens,et al.  Differential roles of Src in transforming growth factor-ß regulation of growth arrest, epithelial-to-mesenchymal transition and cell migration in pancreatic ductal adenocarcinoma cells. , 2011, International journal of oncology.

[13]  Ben Fabry,et al.  The role of the tissue microenvironment in the regulation of cancer cell motility and invasion , 2010, Cell Communication and Signaling.

[14]  F. Bianchini,et al.  Environmental control of invasiveness and metastatic dissemination of tumor cells: the role of tumor cell-host cell interactions , 2010, Cell Communication and Signaling.

[15]  A. Lavrentieva,et al.  Effects of hypoxic culture conditions on umbilical cord-derived human mesenchymal stem cells , 2010, Cell Communication and Signaling.

[16]  Jenny G. Parvani,et al.  The Pathophysiology of Epithelial-Mesenchymal Transition Induced by Transforming Growth Factor-β in Normal and Malignant Mammary Epithelial Cells , 2010, Journal of Mammary Gland Biology and Neoplasia.

[17]  D. Radisky,et al.  Matrix Metalloproteinase-Induced Epithelial-Mesenchymal Transition in Breast Cancer , 2010, Journal of Mammary Gland Biology and Neoplasia.

[18]  R. Hass,et al.  Cellular senescence of human mammary epithelial cells (HMEC) is associated with an altered MMP-7/HB-EGF signaling and increased formation of elastin-like structures , 2009, Mechanisms of Ageing and Development.

[19]  R. Hass,et al.  Characterization of human breast cancer epithelial cells (HBCEC) derived from long term cultured biopsies , 2009, Journal of experimental & clinical cancer research : CR.

[20]  Cornelia Kasper,et al.  Identification of subpopulations in mesenchymal stem cell-like cultures from human umbilical cord , 2009, Cell Communication and Signaling.

[21]  P. Porter Global trends in breast cancer incidence and mortality. , 2009, Salud publica de Mexico.

[22]  T. Motyl,et al.  Matrix metalloproteinase-2 involvement in breast cancer progression: a mini-review. , 2009, Medical science monitor : international medical journal of experimental and clinical research.

[23]  M. Sogayar,et al.  Correlation between MMPs and their inhibitors in breast cancer tumor tissue specimens and in cell lines with different metastatic potential , 2009, BMC Cancer.

[24]  C. Klein,et al.  The Metastasis Cascade , 2008, Science.

[25]  E. McSherry,et al.  Molecular basis of invasion in breast cancer. , 2007, Cellular and molecular life sciences : CMLS.

[26]  E. McSherry,et al.  Common Molecular Mechanisms of Mammary Gland Development and Breast Cancer , 2007, Cellular and Molecular Life Sciences.

[27]  M. Tiemann,et al.  [Coagulation and formation of malignant effusions]. , 2007, Hamostaseologie.

[28]  B. Sipos,et al.  Dissecting the role of TGF-beta type I receptor/ALK5 in pancreatic ductal adenocarcinoma: Smad activation is crucial for both the tumor suppressive and prometastatic function , 2007, Oncogene.

[29]  Wei Wei,et al.  Metastatic patterns in adenocarcinoma , 2006, Cancer.

[30]  Francis Lin,et al.  Differential effects of EGF gradient profiles on MDA-MB-231 breast cancer cell chemotaxis. , 2004, Experimental cell research.

[31]  Edgar D Staren,et al.  Matrix metalloproteinase expression in breast cancer. , 2003, The Journal of surgical research.

[32]  A. Reith,et al.  SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. , 2002, Molecular pharmacology.

[33]  N. Harbeck,et al.  Clinical relevance of invasion factors urokinase-type plasminogen activator and plasminogen activator inhibitor type 1 for individualized therapy decisions in primary breast cancer is greatest when used in combination. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[34]  N. Brünner,et al.  External quality assessment of trans-European multicentre antigen determinations (enzyme-linked immunosorbent assay) of urokinase-type plasminogen activator (uPA) and its type 1 inhibitor (PAI-1) in human breast cancer tissue extracts. , 1998, British Journal of Cancer.

[35]  H. Ungefroren,et al.  Transcriptional Regulation of the Human Biglycan Gene* , 1996, The Journal of Biological Chemistry.

[36]  Hui-Li Qu,et al.  Transforming growth factor-β 1 enhances the invasiveness of breast cancer cells by inducing a Smad2-dependent epithelial-to-mesenchymal transition. , 2013, Oncology reports.

[37]  P. Friedl,et al.  Cancer invasion and resistance: interconnected processes of disease progression and therapy failure. , 2012, Trends in molecular medicine.

[38]  N. Harbeck,et al.  Procedures for the quantitative protein determination of urokinase and its inhibitor, PAI-1, in human breast cancer tissue extracts by ELISA. , 2006, Methods in molecular medicine.

[39]  Joachim Wegener,et al.  Real-time impedance assay to follow the invasive activities of metastatic cells in culture. , 2002, BioTechniques.