Skip to main content
Erschienen in: Molecular Cancer 1/2015

Open Access 01.12.2015 | Short communication

Erk2 but not Erk1 regulates crosstalk between Met and EGFR in squamous cell carcinoma cell lines

verfasst von: Simone Gusenbauer, Emanuele Zanucco, Pjotr Knyazev, Axel Ullrich

Erschienen in: Molecular Cancer | Ausgabe 1/2015

Abstract

Background

Squamous cell carcinoma (SCC) is the most common type of tongue and larynx cancer and a common type of lung cancer. In this study, we attempted to specifically evaluate the signaling pathway underlying HGF/Met induced EGFR ligand release in SSCs. The Met proto-oncogene encodes for a tyrosine kinase receptor which is often hyperactivated in human cancers. Met activation correlates with poor patient outcome. Several studies revealed a role of Met in receptor-crosstalk inducing either activation of other receptors, or inducing their resistance to targeted cancer treatments. In an epithelial tumor cell line screen we recently showed that the Met ligand HGF blocks the EGFR tyrosine kinase and at the same time activates transcriptional upregulation and accumulation in the supernatant of the EGFR ligand amphiregulin (Oncogene 32:3846–56, 2013). In the present work we describe the pathway responsible for the amphiregulin induction.

Findings

Amphiregulin is transcriptionally upregulated and is released into the supernatant. We show that Erk2 but not Erk1 mediates amphiregulin upregulation upon treatment with monocyte derived HGF. A siRNA knockdown of Erk2 completely abolishes amphiregulin release in squamous cell carcinomas.

Conclusions

These results identify Erk2 as the key downstream signal transducer between Met activation and EGFR ligand upregulation in squamous cell carcinoma cell lines derived from tongue, larynx and lung.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12943-015-0319-z) contains supplementary material, which is available to authorized users.
Simone Gusenbauer and Emanuele Zanucco contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SG and EZ planned and performed the experiments. PK generated the MAD-NT cell line. SG and PK helped to draft the manuscript. AU designed the study and drafted the manuscript. All authors read and approved the final manuscript.
Abkürzungen
B
Blocking antibody
CHX
Cycloheximide
CM
Conditioned medium
EGF
Epidermal growth factor
EGFR
Epidermal growth factor receptor
Erk1 and Erk2
Extracellular signal-regulated kinases 1 and 2
G418
Geneticin
HER2
Human epidermal growth factor receptor 2
HGF
Hepatocyte growth factor
IP
Immunoprecipitation
MAPK
Mitogen-activated protein kinase
Met
Mesenchymal-epithelial transition factor
PI3K
Phosphoinositid-3-kinase
TAM
Tumor associated macrophage
TK
Tyrosine kinase

Findings

A common feature of many human epithelial cancers is a constitutive activation of the Ras/Raf/MEK/Erk signal transduction pathway [1-4]. This hyperactivation can be either induced via activating mutations in members of this pathway or via binding of extracellular signaling molecules to a receptor on the cell-surface. These extracellular signaling molecules include cytokines, extracellular matrix components, GPCR ligands, neurotransmitters and growth factors. Growth factors bind to specific cell-surface receptors harboring intrinsic protein kinase activity, the so called receptor tyrosine kinases (=TK). Following ligand binding, growth-factor receptor tyrosine kinases such as Met, the EGF receptor family, the fibroblast growth factor receptor family or the vascular endothelial growth factor receptor family become activated and recruit intracellular adaptor proteins to the cytoplasmic tails of the activated receptors. A signaling cascade is triggered involving Ras, Raf and MEK. Activated MEK activates the extracellular signal regulated kinases 1 and 2 (Erk1/2) by phosphorylation of Threonine and Tyrosine residues within their activation loop. Once Erk1 and Erk2 are phosphorylated they regulate the transcription of genes involved in a range of fundamental cellular processes including cell survival, proliferation, motility, and differentiation [5]. The two isoforms Erk1 and Erk2 share a high level of amino acid identity (88%), are ubiquitously expressed in mammalian cells, are coactivated in response to extracellular stimuli [6,7] and recognize the same substrates [8-11]. However various studies describe a nonredundant function of Erk1 and Erk2. Analyses of mouse models show that disruption of the Erk2 gene leads to early embryonic lethality, while disruption of Erk1 does not [12,13]. In vitro studies have also suggested that Erk1 and Erk2 may exert distinct functions in certain cellular contexts. For example, a knockdown of Erk2 expression restrains hepatocyte cell division, whereas Erk1 silencing specifically improves long-term hepatocyte survival [14,15]. In breast epithelial cells Erk2 but not Erk1 induces epithelial-to-mesenchymal transformation [16]. Other studies reported that osteosarcoma cells regulate the expression of gp130 via Erk2 [17]. Furthermore it has been reported that siRNA knockdown of Erk1 in fibroblasts enhances Erk2 signaling and results in enhanced cell proliferation [18].
In our study we reveal an Erk2 dependent crosstalk between tumor stroma associated HGF/Met signaling and tumor cell associated EGFR signaling. HGF is a frequently detected ligand in the tumor stroma, mainly released by tumor-associated macrophages (TAMs) and by stromal fibroblasts [19-21]. Met receptor activation in cancer cells upon HGF binding, was shown to trigger several pro-tumorigenic pathways [22-25]. However, the complex crosstalk between epithelial tumor cells and stromal cells is yet poorly understood. Several studies have shown diverse mechanisms of transactivation between Met and the EGF receptor family [26-34]: the hyperactivation of Met, for example, was shown to play a role in resistance formation to EGF-receptor-family-blocking agents [26,27,32]. Scheving et al. demonstrated that inhibition of EGFR TK blocks HGF-induced DNA synthesis in primary hepatocytes, indicating that the proliferative actions of HGF may be secondary via new synthesis or processing of EGFR ligands [29]. Similarly, Spix et al. blocked HGF-induced scattering of human corneal limbal epithelial cells with an EGFR TK inhibitor [30]. Finally Reznik and coworkers demonstrated that HGF stimulation of glioblastoma cells induces EGFR activation via new transcription of EGFR ligands [31].
Here, we attempted to specifically investigate the signaling pathway underlying HGF/Met induced EGFR ligand release in SCCs derived from different tissues. Amphiregulin protein release upon HGF stimulation could be observed in SCCs of the tongue, lung and larynx (Figure 1A). In order to investigate which signal transducer downstream of Met activation mediates the upregulation of amphiregulin, we used, due to the high amphiregulin production, SCC9 cells as a preliminary model system. The amphiregulin transcript induction peaked within the first two hours after HGF stimulation (Figure 1B). Amphiregulin protein accumulation started after 4–8 hours and peaked after 24 hours (Figure 1C). To test whether the amphiregulin release depends on new protein synthesis or on shedding of existing pro-forms, the effect of the translation inhibitors cycloheximide (=CHX) and geneticin (=G418) was investigated. Both inhibitors abrogated amphiregulin release into the supernatant, suggesting that amphiregulin release fully depends on new protein synthesis (Figure 1D). Furthermore, SCC9 cells were incubated with inhibitors for MEK and for PI3 kinase, prior to HGF stimulation. mRNA levels of amphiregulin were measured after 2 hours and protein levels were measured after 24 hours of stimulation. The inhibitor specificity and efficacy was analyzed 5 minutes after HGF stimulation and is shown in Additional file 1: Figure S1. Notably, full inhibition of amphiregulin mRNA (Figure 1E) and protein (Figure 1F) induction was achieved with the MEK inhibitor UO126, while only a minor effect was observed with the PI3K inhibitor at the protein level (Figure 1F). These experiments prove the regulation on transcript level and reveal a MAPK-pathway-dependent amphiregulin production.
Consistent with our previous findings, although the EGFR ligand amphiregulin is present in the cell culture medium, the EGF receptor does not become phosphorylated on tyrosine residues after HGF treatment [32] (Figure 2A). Therefore, in order to test if the released amphiregulin is capable of activating EGFR, the following two-step experiment was performed: first, SCC9 cells were stimulated with HGF. After 24 hours the conditioned medium (=CM) was collected. Second, fresh, untreated SCC9 cells were stimulated for three minutes with this CM, lysed and immunobloted for phospho-tyrosine and EGFR. If active EGFR ligands were shed, an activation of the EGFR should occur now. Indeed, strong EGFR and HER2 phosphorylation was observed and a blocking antibody experiment (verification of antibody specificity in Additional file 2: Figure S2) revealed amphiregulin to be the major contributor of activation (Figure 2B).
Monocytes, macrophages and fibroblasts are thought to be the major sources of HGF in the tumor stroma [35-37]. It was of interest, whether a complex CM derived from a monocytic cell line, is capable to induce EGFR ligand release. Therefore, CM of MAD-NT cells, a HGF producing subclone of the promyelocytic leukemia cell line HL60, was used (Additional file 3: Figure S3). Amphiregulin was released upon MAD-NT CM by SCC9 cells and a HGF-blocking antibody as well as the Met inhibitor PHA-665752 abrogated the amphiregulin production (Figure 2C). The Met inhibitor efficacy was analyzed 3 minutes after HGF stimulation and is shown in Additional file 4: Figure S4. In the next step, SCC9 cells were stimulated for 10 minutes with HGF and with MAD-NT CM and assayed in immunoblot analysis for the activation of the downstream signal transducers Erk1/2, Akt, p38 MAPK and SAPK/JNK. All tested signal transducers got activated by HGF and by MAD-NT CM. Both, the HGF-blocking antibody as well as the Met inhibitor PHA-665752 reduced activation of Erk1/2, Akt, p38 MAPK and SAPK/JNK (Figure 2D). These data indicate, that monocytes can be a major source of HGF and that HGF is a very potent activator of crucial signaling cascades.
To evaluate which MAPK is responsible for the EGFR ligand induction, siRNA knockdown experiments of Erk1 and Erk2 were performed. The CM of MAD-NT cells was used as HGF source. A Knockdown of Erk2 dramatically reduced the release of amphiregulin, whereas a knockdown of Erk1 showed no effect. To exclude the possibility of an off-target effect, two different siRNAs for Erk2 were used. Similar results were obtained with both Erk2 siRNAs. Interestingly a double knockdown of Erk1 and Erk2 further reduced the production of amphiregulin, indicating that Erk1 could partly compensate for the loss of Erk2 (Figure 3A). Moreover Erk2 depletion blocks HGF induced amphiregulin release in tongue-, lung- and larynx-derived SCC cell lines. Notably, although in NCI-H2882 cells an Erk2 knockdown dramatically reduced basal amphiregulin production, HGF stimulation was still able to augment amphiregulin release similar to baseline levels (Figure 3B). Altogether these data suggest that in different human SCCs the upregulation and release of amphiregulin upon HGF stimulation is mediated via Erk2.
In this study we have shown that HGF is a strong inducer of the EGFR ligand amphiregulin in different SCC cell lines and that amphiregulin acts as a potent activator of EGFR and HER2 in HGF untreated SCC9 cells. However in HGF treated cells EGFR and HER2 cannot be further activated by amphiregulin. Therefore it is likely, that the produced amphiregulin is provided for other cell types of the tumor stroma, which did not get in contact with HGF before or amphiregulin is provided for cell types, where the EGFR is not blocked by HGF. A second possibility is that amphiregulin might induce the recruitment and proliferation of stromal cells like endothelial cells and fibroblasts (Figure 4) which promote tumor progression. Interestingly Amin et al. compared tumor-associated endothelial cells and normal endothelial cells and found that tumor-derived endothelial cells express EGFR, HER2 and HER4, whereas their normal counterparts express HER2, HER3 and HER4 [38]. As a consequence of the gain of EGFR and the loss of HER3, tumor vasculature responds to EGFR ligands. In their study they suggest that this receptor exchange promotes tumor angiogenesis. Similarly Cascone et al. showed in a mouse xenograft model of human lung andenocarcinoma an upregulation and hyperactivation of stromal EGFR in blood vessel pericytes of anti-VEGF treatment resistant tumors. In this elegant study they were able to distinguish between stromal (mouse) and cancer (human) cell specific changes of total and of phosphorylated EGFR levels [39]. Several other studies show a proangiogenic and tumor supporting effect of EGFR signaling inside the tumor vasculature [40,41]. A third possibility is, that the tumor-produced EGFR ligands exert their function in distant tissues and influence the generation of the metastatic niche. In this study, we identified MAPK signaling as the underlying pathway for new amphiregulin mRNA and protein syntheses. We intended to specifically inhibit Erk1 and 2 by siRNA knockdown and could demonstrate that Erk2 but not Erk1 is responsible for amphiregulin upregulation. In addition we demonstrate the ability of a monocytic cell line to induce amphiregulin release in a HGF/Met/MAPK-dependent manner. Our study grants further investigations which cell type in the tumor microenvironment or in distant tissues benefits from the released EGFR ligand amphiregulin during cancer progression. The detailed understanding of stromal signaling may be critical for the development of successful treatments and for the improvement of combination regimens.

Materials and methods

Cell culture

Human squamous cell carcinoma cell lines derived from different tissues were as follows: tongue-derived SCC4, SCC9 and SCC15, larynx-derived UM-SCC-17B and lung-derived NCI-H2882 and HCC95. The acute promyelocytic leukemia cell line HL-60 clone MAD-NT (Macrophage differentiation, non-terminal) has undergone spontaneous differentiation into semiadherent cells and was generated in our laboratory. This cell line has been used due to its ability to spontaneously produce high levels of HGF (Additional file 3: Figure S3). All cell lines were obtained from the American Type Culture Collection, except HCC95 and NCI-H2882 cells which were a kind gift from Prof. Roman Thomas (University of Cologne) and UM-SCC-17B cells which were a kind gift from Prof. Thomas Wustrow (Ludwig-Maximilian-University of Munich). The ligands and blocking antibodies were purchased from R & D Systems. Geneticin was from Invitrogen, UO126 was from Cell Signaling Technologies, cycloheximide and Wortmannin were from Sigma, PHA-665752 was from Biomol. The siRNAs for Erk1 (cat# J-003592-10), Erk2#1 (cat# J-003555-12), Erk2#2 (cat# J-003555-14) and the non-targeting siRNA (cat# D-001810-03) were purchased from Dharmacon and transfection was performed with Lipofectamine RNAiMax (Invitrogen).

Quantitative RT-PCR

Total RNA was transcribed into complementary DNA using First Strand cDNA Synthesis Kit (Fermentas). SYBR Green Master Mix (Roche) was used for RT-qPCR measurement. The used primers were: amphiregulin → 5′-TGGTGCTGTCGCTCTTGATA-3′, ← 5′-GCCAGGTATTTGTGGTTCGT-3′; reference gene HPRT1 → 5′-GCTATAAATTCTTTGCTGACCTGCTG-3′, ← 5′-AATTACTTTTATGTCCCCTGTTGACTGG-3′.

ELISA

Amphiregulin and HGF protein was measured using sandwich ELISA (R&D Systems).

Immunoblotting and immunoprecipitation

For immunoprecipitation 0.5 mg of total protein, 1 μg of homemade monoclonal EGFR antibody (clone 108.1; has been characterized before [44]) and 5 μg of monoclonal HER2 antibody (clone 13D1B1; has been characterized before [44]) together with 10 μl of protein A-sepharose (GE Healthcare) were used. For the HGF immunoprecipitation 1 ml of HL60 and MAD-NT cell CM and 20 μg monoclonal HGF antibody (R&D Systems) together with 15 μl of protein A-sepharose were used. For immunoblotting the following antibodies were used: p-tyrosine (homemade clone 4G10), pAkt S473 (Cell Signaling Technologies), pErk1/2 T202/Y204 (Cell Signaling Technologies), pSAPK/JNK T183/Y185 (Cell Signaling Technologies), Erk1/2 (Santa Cruz Biotechnology), EGFR (Cell Signaling Technologies), HER2 (Millipore), HGF (R&D Systems), pMet Y1234/Y1235 (Cell Signaling Technologies), pp38 T180/Y182 (Cell Signaling Technologies) and tubulin (Sigma).

Acknowledgements

We thank Philip Vlaicu for sharing his knowledge that MAD-NT cells produce high amounts of HGF.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SG and EZ planned and performed the experiments. PK generated the MAD-NT cell line. SG and PK helped to draft the manuscript. AU designed the study and drafted the manuscript. All authors read and approved the final manuscript.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54.CrossRefPubMed Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54.CrossRefPubMed
2.
Zurück zum Zitat Collisson EA, Trejo CL, Silva JM, Gu S, Korkola JE, Heiser LM, et al. A central role for RAF–− > MEK–− > ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2012;2:685–93.CrossRefPubMedCentralPubMed Collisson EA, Trejo CL, Silva JM, Gu S, Korkola JE, Heiser LM, et al. A central role for RAF–− > MEK–− > ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2012;2:685–93.CrossRefPubMedCentralPubMed
3.
Zurück zum Zitat Choueiri TK, Cheville J, Palescandolo E, Fay AP, Kantoff PW, Atkins MB, et al. BRAF mutations in metanephric adenoma of the kidney. Eur Urol. 2012;62:917–22.CrossRefPubMed Choueiri TK, Cheville J, Palescandolo E, Fay AP, Kantoff PW, Atkins MB, et al. BRAF mutations in metanephric adenoma of the kidney. Eur Urol. 2012;62:917–22.CrossRefPubMed
4.
Zurück zum Zitat Singer G, Oldt 3rd R, Cohen Y, Wang BG, Sidransky D, Kurman RJ, et al. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst. 2003;95:484–6.CrossRefPubMed Singer G, Oldt 3rd R, Cohen Y, Wang BG, Sidransky D, Kurman RJ, et al. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst. 2003;95:484–6.CrossRefPubMed
5.
Zurück zum Zitat Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 2004;4:937–47.CrossRefPubMed Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 2004;4:937–47.CrossRefPubMed
6.
Zurück zum Zitat Lewis TS, Shapiro PS, Ahn NG. Signal transduction through MAP kinase cascades. Adv Cancer Res. 1998;74:49–139.CrossRefPubMed Lewis TS, Shapiro PS, Ahn NG. Signal transduction through MAP kinase cascades. Adv Cancer Res. 1998;74:49–139.CrossRefPubMed
7.
Zurück zum Zitat Meloche S. Cell cycle reentry of mammalian fibroblasts is accompanied by the sustained activation of p44mapk and p42mapk isoforms in the G1 phase and their inactivation at the G1/S transition. J Cell Physiol. 1995;163:577–88.CrossRefPubMed Meloche S. Cell cycle reentry of mammalian fibroblasts is accompanied by the sustained activation of p44mapk and p42mapk isoforms in the G1 phase and their inactivation at the G1/S transition. J Cell Physiol. 1995;163:577–88.CrossRefPubMed
8.
Zurück zum Zitat Voisin L, Saba-El-Leil MK, Julien C, Fremin C, Meloche S. Genetic demonstration of a redundant role of extracellular signal-regulated kinase 1 (ERK1) and ERK2 mitogen-activated protein kinases in promoting fibroblast proliferation. Mol Cell Biol. 2010;30:2918–32.CrossRefPubMedCentralPubMed Voisin L, Saba-El-Leil MK, Julien C, Fremin C, Meloche S. Genetic demonstration of a redundant role of extracellular signal-regulated kinase 1 (ERK1) and ERK2 mitogen-activated protein kinases in promoting fibroblast proliferation. Mol Cell Biol. 2010;30:2918–32.CrossRefPubMedCentralPubMed
9.
Zurück zum Zitat Lefloch R, Pouyssegur J, Lenormand P. Single and combined silencing of ERK1 and ERK2 reveals their positive contribution to growth signaling depending on their expression levels. Mol Cell Biol. 2008;28:511–27.CrossRefPubMedCentralPubMed Lefloch R, Pouyssegur J, Lenormand P. Single and combined silencing of ERK1 and ERK2 reveals their positive contribution to growth signaling depending on their expression levels. Mol Cell Biol. 2008;28:511–27.CrossRefPubMedCentralPubMed
10.
Zurück zum Zitat Robbins DJ, Zhen E, Owaki H, Vanderbilt CA, Ebert D, Geppert TD, et al. Regulation and properties of extracellular signal-regulated protein kinases 1 and 2 in vitro. J Biol Chem. 1993;268:5097–106.PubMed Robbins DJ, Zhen E, Owaki H, Vanderbilt CA, Ebert D, Geppert TD, et al. Regulation and properties of extracellular signal-regulated protein kinases 1 and 2 in vitro. J Biol Chem. 1993;268:5097–106.PubMed
11.
Zurück zum Zitat Gonzalez FA, Raden DL, Davis RJ. Identification of substrate recognition determinants for human ERK1 and ERK2 protein kinases. J Biol Chem. 1991;266:22159–63.PubMed Gonzalez FA, Raden DL, Davis RJ. Identification of substrate recognition determinants for human ERK1 and ERK2 protein kinases. J Biol Chem. 1991;266:22159–63.PubMed
12.
Zurück zum Zitat Hatano N, Mori Y, Oh-hora M, Kosugi A, Fujikawa T, Nakai N, et al. Essential role for ERK2 mitogen-activated protein kinase in placental development. Genes Cells. 2003;8:847–56.CrossRefPubMed Hatano N, Mori Y, Oh-hora M, Kosugi A, Fujikawa T, Nakai N, et al. Essential role for ERK2 mitogen-activated protein kinase in placental development. Genes Cells. 2003;8:847–56.CrossRefPubMed
13.
Zurück zum Zitat Yao Y, Li W, Wu J, Germann UA, Su MS, Kuida K, et al. Extracellular signal-regulated kinase 2 is necessary for mesoderm differentiation. Proc Natl Acad Sci U S A. 2003;100:12759–64.CrossRefPubMedCentralPubMed Yao Y, Li W, Wu J, Germann UA, Su MS, Kuida K, et al. Extracellular signal-regulated kinase 2 is necessary for mesoderm differentiation. Proc Natl Acad Sci U S A. 2003;100:12759–64.CrossRefPubMedCentralPubMed
14.
Zurück zum Zitat Fremin C, Bessard A, Ezan F, Gailhouste L, Regeard M, Le Seyec J, et al. Multiple division cycles and long-term survival of hepatocytes are distinctly regulated by extracellular signal-regulated kinases ERK1 and ERK2. Hepatology. 2009;49:930–9.CrossRefPubMed Fremin C, Bessard A, Ezan F, Gailhouste L, Regeard M, Le Seyec J, et al. Multiple division cycles and long-term survival of hepatocytes are distinctly regulated by extracellular signal-regulated kinases ERK1 and ERK2. Hepatology. 2009;49:930–9.CrossRefPubMed
15.
Zurück zum Zitat Fremin C, Ezan F, Boisselier P, Bessard A, Pages G, Pouyssegur J, et al. ERK2 but not ERK1 plays a key role in hepatocyte replication: an RNAi-mediated ERK2 knockdown approach in wild-type and ERK1 null hepatocytes. Hepatology. 2007;45:1035–45.CrossRefPubMed Fremin C, Ezan F, Boisselier P, Bessard A, Pages G, Pouyssegur J, et al. ERK2 but not ERK1 plays a key role in hepatocyte replication: an RNAi-mediated ERK2 knockdown approach in wild-type and ERK1 null hepatocytes. Hepatology. 2007;45:1035–45.CrossRefPubMed
16.
Zurück zum Zitat Shin S, Dimitri CA, Yoon SO, Dowdle W, Blenis J. ERK2 but not ERK1 induces epithelial-to-mesenchymal transformation via DEF motif-dependent signaling events. Mol Cell. 2010;38:114–27.CrossRefPubMedCentralPubMed Shin S, Dimitri CA, Yoon SO, Dowdle W, Blenis J. ERK2 but not ERK1 induces epithelial-to-mesenchymal transformation via DEF motif-dependent signaling events. Mol Cell. 2010;38:114–27.CrossRefPubMedCentralPubMed
17.
Zurück zum Zitat Bonito NA, Drechsler J, Stoecker S, Carmo CR, Seckl MJ, Hermanns HM, et al. Control of gp130 expression by the mitogen-activated protein kinase ERK2. Oncogene. 2014;33:2255–63.CrossRefPubMed Bonito NA, Drechsler J, Stoecker S, Carmo CR, Seckl MJ, Hermanns HM, et al. Control of gp130 expression by the mitogen-activated protein kinase ERK2. Oncogene. 2014;33:2255–63.CrossRefPubMed
18.
Zurück zum Zitat Vantaggiato C, Formentini I, Bondanza A, Bonini C, Naldini L, Brambilla R. ERK1 and ERK2 mitogen-activated protein kinases affect Ras-dependent cell signaling differentially. J Biol. 2006;5:14.CrossRefPubMedCentralPubMed Vantaggiato C, Formentini I, Bondanza A, Bonini C, Naldini L, Brambilla R. ERK1 and ERK2 mitogen-activated protein kinases affect Ras-dependent cell signaling differentially. J Biol. 2006;5:14.CrossRefPubMedCentralPubMed
19.
Zurück zum Zitat Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–31.CrossRefPubMed Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–31.CrossRefPubMed
20.
Zurück zum Zitat Liguori M, Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages as incessant builders and destroyers of the cancer stroma. Cancers (Basel). 2011;3:3740–61.CrossRef Liguori M, Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages as incessant builders and destroyers of the cancer stroma. Cancers (Basel). 2011;3:3740–61.CrossRef
21.
Zurück zum Zitat White JR, Harris RA, Lee SR, Craigon MH, Binley K, Price T, et al. Genetic amplification of the transcriptional response to hypoxia as a novel means of identifying regulators of angiogenesis. Genomics. 2004;83:1–8.CrossRefPubMed White JR, Harris RA, Lee SR, Craigon MH, Binley K, Price T, et al. Genetic amplification of the transcriptional response to hypoxia as a novel means of identifying regulators of angiogenesis. Genomics. 2004;83:1–8.CrossRefPubMed
22.
Zurück zum Zitat Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 1991;251:802–4.CrossRefPubMed Bottaro DP, Rubin JS, Faletto DL, Chan AM, Kmiecik TE, Vande Woude GF, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 1991;251:802–4.CrossRefPubMed
23.
Zurück zum Zitat Ponzetto C, Bardelli A, Zhen Z, Maina F, Dalla Zonca P, Giordano S, et al. A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell. 1994;77:261–71.CrossRefPubMed Ponzetto C, Bardelli A, Zhen Z, Maina F, Dalla Zonca P, Giordano S, et al. A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell. 1994;77:261–71.CrossRefPubMed
24.
Zurück zum Zitat Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–25.CrossRefPubMed Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–25.CrossRefPubMed
25.
Zurück zum Zitat Gao CF, Vande Woude GF. HGF/SF-Met signaling in tumor progression. Cell Res. 2005;15:49–51.CrossRefPubMed Gao CF, Vande Woude GF. HGF/SF-Met signaling in tumor progression. Cell Res. 2005;15:49–51.CrossRefPubMed
26.
Zurück zum Zitat Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.CrossRefPubMed Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.CrossRefPubMed
27.
Zurück zum Zitat Shattuck DL, Miller JK, Carraway 3rd KL, Sweeney C. Met receptor contributes to trastuzumab resistance of Her2-overexpressing breast cancer cells. Cancer Res. 2008;68:1471–7.CrossRefPubMed Shattuck DL, Miller JK, Carraway 3rd KL, Sweeney C. Met receptor contributes to trastuzumab resistance of Her2-overexpressing breast cancer cells. Cancer Res. 2008;68:1471–7.CrossRefPubMed
28.
Zurück zum Zitat Suda K, Murakami I, Katayama T, Tomizawa K, Osada H, Sekido Y, et al. Reciprocal and complementary role of MET amplification and EGFR T790M mutation in acquired resistance to kinase inhibitors in lung cancer. Clin Cancer Res. 2010;16:5489–98.CrossRefPubMed Suda K, Murakami I, Katayama T, Tomizawa K, Osada H, Sekido Y, et al. Reciprocal and complementary role of MET amplification and EGFR T790M mutation in acquired resistance to kinase inhibitors in lung cancer. Clin Cancer Res. 2010;16:5489–98.CrossRefPubMed
29.
Zurück zum Zitat Scheving LA, Stevenson MC, Taylormoore JM, Traxler P, Russell WE. Integral role of the EGF receptor in HGF-mediated hepatocyte proliferation. Biochem Biophys Res Commun. 2002;290:197–203.CrossRefPubMed Scheving LA, Stevenson MC, Taylormoore JM, Traxler P, Russell WE. Integral role of the EGF receptor in HGF-mediated hepatocyte proliferation. Biochem Biophys Res Commun. 2002;290:197–203.CrossRefPubMed
30.
Zurück zum Zitat Spix JK, Chay EY, Block ER, Klarlund JK. Hepatocyte growth factor induces epithelial cell motility through transactivation of the epidermal growth factor receptor. Exp Cell Res. 2007;313:3319–25.CrossRefPubMedCentralPubMed Spix JK, Chay EY, Block ER, Klarlund JK. Hepatocyte growth factor induces epithelial cell motility through transactivation of the epidermal growth factor receptor. Exp Cell Res. 2007;313:3319–25.CrossRefPubMedCentralPubMed
31.
Zurück zum Zitat Reznik TE, Sang Y, Ma Y, Abounader R, Rosen EM, Xia S, et al. Transcription-dependent epidermal growth factor receptor activation by hepatocyte growth factor. Mol Cancer Res. 2008;6:139–50.CrossRefPubMedCentralPubMed Reznik TE, Sang Y, Ma Y, Abounader R, Rosen EM, Xia S, et al. Transcription-dependent epidermal growth factor receptor activation by hepatocyte growth factor. Mol Cancer Res. 2008;6:139–50.CrossRefPubMedCentralPubMed
32.
Zurück zum Zitat Gusenbauer S, Vlaicu P, Ullrich A. HGF induces novel EGFR functions involved in resistance formation to tyrosine kinase inhibitors. Oncogene. 2013;32:3846–56.CrossRefPubMed Gusenbauer S, Vlaicu P, Ullrich A. HGF induces novel EGFR functions involved in resistance formation to tyrosine kinase inhibitors. Oncogene. 2013;32:3846–56.CrossRefPubMed
33.
Zurück zum Zitat Han C, Michalopoulos GK, Wu T. Prostaglandin E2 receptor EP1 transactivates EGFR/MET receptor tyrosine kinases and enhances invasiveness in human hepatocellular carcinoma cells. J Cell Physiol. 2006;207:261–70.CrossRefPubMed Han C, Michalopoulos GK, Wu T. Prostaglandin E2 receptor EP1 transactivates EGFR/MET receptor tyrosine kinases and enhances invasiveness in human hepatocellular carcinoma cells. J Cell Physiol. 2006;207:261–70.CrossRefPubMed
34.
Zurück zum Zitat Guo A, Villen J, Kornhauser J, Lee KA, Stokes MP, Rikova K, et al. Signaling networks assembled by oncogenic EGFR and c-Met. Proc Natl Acad Sci U S A. 2008;105:692–7.CrossRefPubMedCentralPubMed Guo A, Villen J, Kornhauser J, Lee KA, Stokes MP, Rikova K, et al. Signaling networks assembled by oncogenic EGFR and c-Met. Proc Natl Acad Sci U S A. 2008;105:692–7.CrossRefPubMedCentralPubMed
35.
Zurück zum Zitat Coffelt SB, Hughes R, Lewis CE. Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta. 2009;1796:11–8.PubMed Coffelt SB, Hughes R, Lewis CE. Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta. 2009;1796:11–8.PubMed
36.
Zurück zum Zitat Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–8.CrossRefPubMed Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–8.CrossRefPubMed
38.
Zurück zum Zitat Amin DN, Hida K, Bielenberg DR, Klagsbrun M. Tumor endothelial cells express epidermal growth factor receptor (EGFR) but not ErbB3 and are responsive to EGF and to EGFR kinase inhibitors. Cancer Res. 2006;66:2173–80.CrossRefPubMed Amin DN, Hida K, Bielenberg DR, Klagsbrun M. Tumor endothelial cells express epidermal growth factor receptor (EGFR) but not ErbB3 and are responsive to EGF and to EGFR kinase inhibitors. Cancer Res. 2006;66:2173–80.CrossRefPubMed
39.
Zurück zum Zitat Cascone T, Herynk MH, Xu L, Du Z, Kadara H, Nilsson MB, et al. Upregulated stromal EGFR and vascular remodeling in mouse xenograft models of angiogenesis inhibitor-resistant human lung adenocarcinoma. J Clin Invest. 2011;121:1313–28.CrossRefPubMedCentralPubMed Cascone T, Herynk MH, Xu L, Du Z, Kadara H, Nilsson MB, et al. Upregulated stromal EGFR and vascular remodeling in mouse xenograft models of angiogenesis inhibitor-resistant human lung adenocarcinoma. J Clin Invest. 2011;121:1313–28.CrossRefPubMedCentralPubMed
40.
Zurück zum Zitat Nolan-Stevaux O, Truitt MC, Pahler JC, Olson P, Guinto C, Lee DC, et al. Differential contribution to neuroendocrine tumorigenesis of parallel egfr signaling in cancer cells and pericytes. Genes Cancer. 2010;1:125–41.CrossRefPubMedCentralPubMed Nolan-Stevaux O, Truitt MC, Pahler JC, Olson P, Guinto C, Lee DC, et al. Differential contribution to neuroendocrine tumorigenesis of parallel egfr signaling in cancer cells and pericytes. Genes Cancer. 2010;1:125–41.CrossRefPubMedCentralPubMed
41.
Zurück zum Zitat Yokoi K, Sasaki T, Bucana CD, Fan D, Baker CH, Kitadai Y, et al. Simultaneous inhibition of EGFR, VEGFR, and platelet-derived growth factor receptor signaling combined with gemcitabine produces therapy of human pancreatic carcinoma and prolongs survival in an orthotopic nude mouse model. Cancer Res. 2005;65:10371–80.CrossRefPubMedCentralPubMed Yokoi K, Sasaki T, Bucana CD, Fan D, Baker CH, Kitadai Y, et al. Simultaneous inhibition of EGFR, VEGFR, and platelet-derived growth factor receptor signaling combined with gemcitabine produces therapy of human pancreatic carcinoma and prolongs survival in an orthotopic nude mouse model. Cancer Res. 2005;65:10371–80.CrossRefPubMedCentralPubMed
42.
Zurück zum Zitat Moon WS, Park HS, Yu KH, Park MY, Kim KR, Jang KY, et al. Expression of betacellulin and epidermal growth factor receptor in hepatocellular carcinoma: implications for angiogenesis. Hum Pathol. 2006;37:1324–32.CrossRefPubMed Moon WS, Park HS, Yu KH, Park MY, Kim KR, Jang KY, et al. Expression of betacellulin and epidermal growth factor receptor in hepatocellular carcinoma: implications for angiogenesis. Hum Pathol. 2006;37:1324–32.CrossRefPubMed
43.
Zurück zum Zitat Shiurba RA, Eng LF, Vogel H, Lee YL, Horoupian DS, Urich H. Epidermal growth factor receptor in meningiomas is expressed predominantly on endothelial cells. Cancer. 1988;62:2139–44.CrossRefPubMed Shiurba RA, Eng LF, Vogel H, Lee YL, Horoupian DS, Urich H. Epidermal growth factor receptor in meningiomas is expressed predominantly on endothelial cells. Cancer. 1988;62:2139–44.CrossRefPubMed
44.
Zurück zum Zitat Ruschel A, Ullrich A. Protein tyrosine kinase Syk modulates EGFR signalling in human mammary epithelial cells. Cell Signal. 2004;16:1249–61.CrossRefPubMed Ruschel A, Ullrich A. Protein tyrosine kinase Syk modulates EGFR signalling in human mammary epithelial cells. Cell Signal. 2004;16:1249–61.CrossRefPubMed
Metadaten
Titel
Erk2 but not Erk1 regulates crosstalk between Met and EGFR in squamous cell carcinoma cell lines
verfasst von
Simone Gusenbauer
Emanuele Zanucco
Pjotr Knyazev
Axel Ullrich
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2015
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-015-0319-z

Weitere Artikel der Ausgabe 1/2015

Molecular Cancer 1/2015 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.