Page 373 - Read Online
P. 373

Page 2 of 16                      Verkoeijen et al. J Cancer Metastasis Treat 2019;5:51  I  http://dx.doi.org/10.20517/2394-4722.2019.06

               while re-expression of EGFR in MTLn3-paxillinS178A cells fully restored EGF-driven cell motility and focal adhesion
               dynamics. Furthermore, re-expression of EGFR in MTLn3-paxillinS178A rescued spontaneous metastasis from
               breast to lung.


               Conclusion: Overall our data show an important role for JNK-mediated paxillin Ser178 phosphorylation in the
               regulation of EGFR expression and thereby, in EGF-driven cell migration and metastasis formation.

               Keywords: Paxillin, c-Jun NH2-terminal kinase, focal adhesion, epidermal growth factor receptor, cell migration,
               metastasis, breast cancer





               INTRODUCTION
               Breast cancer represents the most common type of cancer among women. The formation of metastases,
               which is a determinant of the prognosis of cancer patients, involves distinct cellular processes including
               cell migration,  invasion,  intra-  and  extravasation  and  proliferation.  These  processes  are  regulated  by
               growth factors, cytokines and cellular matrix molecules . An important regulator of cell proliferation
                                                                [1-6]
               and migration is the receptor tyrosine kinase epidermal growth factor receptor (EGFR) whose stimulation
               by epidermal growth factor (EGF) results in the activation of downstream signaling pathways including
               different mitogen-activated protein kinases cascades: extracellular signal-regulated kinase (ERK), p38 and
               c-Jun NH2-terminal kinase (JNK) [7-11] .


               The JNK group of kinases has essential roles in cancer development including regulation of the survival/
               proliferation balance as well as cell migration [7,9,12,13] . Dual Thr and Tyr phosphorylation of JNK by upstream
               MAP kinases results in JNK activation and nuclear translocation. In the nucleus, JNKs phosphorylate and
               activate transcription factors including members of the AP-1 family such as c-Jun. The JNK-AP-1 pathway
               regulates the expression of genes involved in the cell cycle, survival and apoptosis and extracellular matrix
               homeostasis [14-22] . Besides phosphorylation of transcription factors, several cytoskeleton-associated adaptor
               and signaling proteins have recently been identified as direct JNK substrates including β-catenin  and
                                                                                                    [23]
               paxillin [24,25] . Since both proteins are well known to be involved in cell-matrix and cell-cell contacts, the
               direct JNK-mediated phosphorylation of these targets may also influence cell migration.

               Paxillin is a 68 kD multidomain adaptor protein associated with focal adhesions [26-31] . It is long known
               to function as a scaffold to integrate multiple signaling pathways involved in matrix adhesion dynamics
               and cell migration [27,31-36] , and recently, it was shown to play a role in the nucleus in relation to mRNA
               transcription and subsequent translation [37,38] . Few studies have identified paxillin as a JNK substrate [8,24,25] .
               EGF-driven JNK activation results in JNK-mediated phosphorylation of paxillin at Ser178 and is required
               for cell migration [9,13,19,25,39] . In addition, expression of a phospho-defective Ser178 to Ala mutant of paxillin
               inhibited cell migration [9,19,24] . How JNK-mediated phosphorylation of paxillin regulates cell migration is still
               under investigation. It is still unclear how this phosphorylation event is relevant for breast cancer metastasis
               formation [40,41] .

               Here we explored the role and mechanism of paxillin Ser178 phosphorylation in breast tumor progression
               using the highly metastastic breast tumor cell line MTLn3 as a model. We show that ectopic expression of
               paxillinS178A significantly decreased EGF-dependent signaling and cell migration. Using an orthotopic
               mammary gland tumor/metastasis model, we demonstrate that the JNK-mediated phosphorylation of
               paxillin at Ser178 is essential for efficient metastasis of MTLn3 cells to the lung. Expression of mutant
               paxillinS178A was found to reduce EGFR expression, whereas re-expression of EGFR rescued the defected
               tumor cell migration and metastasis formation. Our data indicate that the phosphorylation of Ser178 of
   368   369   370   371   372   373   374   375   376   377   378