Distinct regulatory effect of the p34SEI-1 oncoprotein on cancer metastasis in HER2/neu-positive and -negative cells

  • Authors:
    • Samil Jung
    • Jiyeon Ohk
    • Dongjun Jeong
    • Chengping Li
    • Soonduck Lee
    • Jingjing Duan
    • Changjin Kim
    • Jong-Seok Lim
    • Young Yang
    • Keun-Il Kim
    • Myeong-Sok Lee
  • View Affiliations

  • Published online on: April 28, 2014     https://doi.org/10.3892/ijo.2014.2403
  • Pages: 189-196
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The p34SEI-1 oncoprotein is involved in a transcriptional regulation, cell cycle regulation, apoptosis, development and many other important cellular functions. Our present study suggests that p34SEI-1 can promote metastasis by enhancing migration and invasion of cancer cells. Consistently, p34SEI-1 expression was found to be increased as the tumor invasiveness progressed in human breast tissues. p34SEI-1 may promote cancer metastasis by activating the PI3K/AKT signaling pathway. In this process, p34SEI-1 activates two different serine/threonine kinases, AKT or ILK, depending on the expression status of HER2/neu oncogene. In HER2/neu suppressed cancer cells, p34SEI-1 promoted metastasis mainly by activating AKT via phosphorylation of the 473 serine residue. In HER2/neu expressing cancer cells, p34SEI-1 overexpression downregulates HER2/neu expression, leading to the activation of another crucial serine/threonine kinase ILK due to phosphorylation of the 178 threonine residue instead of AKT. These results suggest that p34SEI-1 affects cancer metastasis by regulating two different signaling pathways depending on the HER2/neu expression level, in which AKT and ILK modulation can be stimulated by p34SEI-1 overexpression.

Introduction

Most of the cancer deaths result from metastasis. In many cases, survival rate of cancer patients with metastatic tumors is much lower than those of patients with localized tumors. Metastasis of breast cancer, the most common form of cancer in woman, is responsible for nearly 90% of deaths from breast cancer. Although breast cancer comprises about 25% of all cancers, the survival rates of patients with breast cancer are relatively high as long as cancer is detected and treated before it metastasizes. Therefore, better understanding of the mechanism that promotes metastasis of breast cancer would be helpful to design effective therapies.

The phosphoinositol-3-kinase (PI3K)/AKT pathway is involved in multiple cellular processes including cell differentiation, proliferation, survival, angiogenesis, invasion and metastasis (18). PI3K phosphorylates phosphatidylinositol 4,5-bisphosphate (PIP2), yielding phosphatidylinositol 3,4,5-triphosphate (PIP3), which recruits and activates phosphatidylinositol-dependent kinase 1 (PDK1). Activated PDK1 phosphorylates serine/threonine-specific protein kinase (AKT), also known as protein kinase B (PKB). AKT activation can be induced by phosphatase and tensin homolog (PTEN) abnormality (911). PTEN tumor suppressor is a negative regulator of the PI3K/AKT pathway, in which PTEN dephosphorylates PIP3 to PIP2 (12). Mutation of deletion of PTEN is frequently found to be mutated or deleted in a broad range of cancers, resulting in the hyper-activation of the PI3K/AKT signaling pathway. In addition, the HER2/neu proto-oncogene (also known as ErbB-2, CD340 or p185) is the most oncogenic signaling activator of PI3K/AKT, in which HER2/neu can phosphorylate PIP2 and in turn induce AKT activation (13,14). HER2/neu plays important roles in the development and progression of certain aggressive types of breast cancer and is found at a relatively high level in most aggressive tumors. Activated AKT acts as a key regulator for many events related with tumor malignancies including cell survival, proliferation, growth, angiogenesis and metastasis (7,12,1418). The critical steps to initiate the metastasis are the phosphorylation of AKT on serine 473 residue and regulation of its downstream target proteins. GSK3β is one of important downstream target proteins involved in metastasis. GSK3β inhibition promotes metastasis by affecting β-catenin, a dual function protein that regulates the coordination of cell-cell adhesion and gene transcription (1922). Deregulated β-catenin expression is associated with many cancers including malignant breast tumors (21,22). GSK3β can be inhibited in at least two ways in PI3K/AKT signaling pathway: activation of AKT to directly phosphorylate GSK3β on serine 9 residue (19) and directly/indirectly by integrin-liked kinase (ILK) (23). ILK is a multifunctional serine/threonine protein kinase that mediates a variety of cellular responses to integrin stimulation by extracellular matrix proteins. The mechanism of ILK activation is not fully understood but it is reported that p21 activated kinase (PAK) can phosphorylate ILK on threonine 173 for its activation (24). It was demonstrated that purified recombinant ILK phosphorylates recombinant GSK3β in vitro (25). It means that ILK can directly phosphorylate GSK3β. Furthermore, inhibition of ILK suppresses tumorigenesis and tumor growth (24,26). Exposure to ILK inhibitor suppresses the snail and β-catenin protein stability and transcriptional activity as well as GSK3β phosphorylation indicating that ILK activity affects the epithelial-mesenchymal transition (EMT) process. Despite these facts, its effect on cancer metastasis still remains unclear and controversial. It has been believed that ILK is a component of the PI3K-AKT pathway by phosphorylating AKT. However, it has been reported that ILK acts as an inhibitor of AKT phosphorylation even though cell proliferation was induced by stably overexpression of ILK (26,27).

With the goal of elucidating the roles of PI3K/AKT signaling pathway in cancer metastasis, we have previously reported the participation of the p34SEI-1 oncoprotein in this pathway. p34SEI-1 enhances cancer cell survival and promotes tumorigenesis by inducing NEDD4-1-mediated PTEN degradation (28). NEDD4-1 negatively regulates PTEN as a proto-oncogenic E3 ubiquitin ligase for PTEN and in turn activates the PI3K/AKT signaling pathway (29,30). Considering the vital roles of PI3K/AKT signaling pathway in metastasis and the indirect effect of p34SEI-1 on this pathway, it was suspected that p34SEI-1 may play an important role in the development of cancer metastasis. p34SEI-1 has multiple biological functions including transcription regulation, cell cycle regulation, inhibition of apoptosis and tumorigenesis (3133). Furthermore, p34SEI-1 increases chromosomal instability, which is closely related to cancer invasiveness (34,35). We previously showed that the expression level of p34SEI-1 is significantly increased in cancer tissue compared to normal tissues, in which p34SEI-1 stabilizes X-linked inhibitor of apoptosis protein (XIAP) leading to an anti-apoptotic effect (33). XIAP also promotes progression of metastasis by activating the oncogenic NF-κB transcriptional factor, fibronectin-related gene expression and cell motility kinase such as focal adhesion kinase (FAK) or Src (36,37).

All these data implicate that p34SEI-1 may be involved in the progression of metastatic cancers. We therefore investigated whether or not p34SEI-1 has metastatic potential and the nature of the mechanism.

Materials and methods

Cell lines, cell culture and materials

Five cancer cell lines were used in this study. MCF7, T47D, HEK293T and MDA-MB-231 cancer cells were cultured in DMEM medium and SKBR3 cells were grown in RPMI medium (Welgene Inc., Daegu, Korea). All media were supplemented with 10% FBS (Gibco-BRL, Carlsbad, CA, USA) and 1% antibiotic-anti-mycotic (Gibco-BRL). All cultures were grown at 37°C in a humidified atmosphere composed of 95% air and 5% CO2. LY294004 (cat no. 440202) and Cpd22 (cat no. 407331) were purchased from Calbiochem (La Jolla, CA, USA).

Reverse transcription (RT)-PCR

Total RNA was extracted from MDA-MB-231 cells with the RNeasy mini kit (cat no. 74106; Qiagen, Hilden, Germany) following the manufacturer’s instructions. For reverse transcription, 1 μg RNA of each sample was subjected to cDNA synthesis using an oligo (dT) primer and the ImProm-II™ Reverse Transcription System (A3800; Promega, Madison, WI, USA). PCR amplification was performed using 10 ng cDNA, different sets of primers, and AccuPower PCR PreMix system (Bioneer, Daejeon, Korea). The amplification reaction was carried out using a PCR Thermal Cycler Dice (Applied Biosystems, Foster City, CA, USA). Each gene product was amplified using corresponding pairs of primers, in which β-actin gene product was used as an internal control. The oligonucleotide sequences for RT-PCR analysis were: pRT-p34SEI-1-RT forward, 5′-AGGACCTCAGCCACAT TGAG-3′ and reverse, 5′-GGTGCCCAAAGTTCATTGTC-3′; pRT-HER2/neu-RT forward, 5′-CTGAACTGGTGTATGC AGAT-3′ and reverse, 5′-CCACACAGTCACACCATAA-3′; pRT-NEDD4-1 forward, 5′-TGGGACATCACTTTGT GATC-3′ and reverse, 5′-TGAGGCTTTTACTGGGGTC-3′; pRT-β-catenin forward, 5′-CATTTCCAATCTACTAATGC-3′ and 5′-CTGCATTCTGACTTTCAGTA-3′; pRT-c-MYC forward, 5′-ACCAGCAGCGACTCTGAGGA-3′ and reverse, 5′-TGACCCTCTTGGCAGCAGGATAGTCC-3′; pRT-ACTB forward, 5′-AGGTCGGAGTCAACGGATTTG-3′ and reverse, 5′-GTGATGGCATGGACTGTGGT-3′.

Western blot analysis

Cells were recovered from culture by centrifugation at 3,000 rpm for 1 min and washed twice in an ice-cold phosphate-buffered saline (PBS) buffer. The cells were then lysed in RIPA lysis buffer and the protein was quantified using a protein assay kit (Bio-Rad, Hercules, CA, USA). Approximately 25 μg of total protein per sample was subjected to 12% SDS-PAGE and the resolved proteins were transferred to Immobilon transfer membranes (cat no. IPVH00010, Millipore, Billerica, MA, USA). The filter was blocked in 5% non-fat dry milk/0.1% Tween-20/Tris-buffered saline (TBS) followed by incubation with each corresponding antibody and immune-detection was accomplished using the Power Opti-ECL Western blotting detection reagent (Bionote, Hwaseong, Korea). Antibodies used in this study were purchased as follows: p34SEI-1 (ALX-804-645; Enzo Life Sciences, Farmingdale, NY, USA), NEDD4-1 (sc-25508) and PTEN (sc-7974; from Santa Cruz Biotechnology, Santa Cruz, CA, USA), E-cadherin (cat no. 610181) and N-cadherin (cat no. 610921; from BD Biosciences, Franklin Lakes, NJ, USA), vimentin (sc-7557), fibronectin (sc-8422), HER2/neu (sc-33684; from Santa Cruz Biotechnology), pAKTser473 (cat no. 9271; Cell Signaling, Danvers, MA, USA), pILKthr178 (sc-130196; Santa Cruz Biotechnology), pGSK3βser9 (cat no. 2435-1; Epitomics, Burlingame, CA, USA) and γ-tubulin (sc-7396; Santa Cruz Biotechnology).

Overexpression or suppression of p34SEI-1 or HER2/neu gene

For overexpression of p34SEI-1, cells were plated at 1×106 cells in a 60-mm-diameter culture dish and transfected with 4 μg of either the control empty vector (pEF-BOS-EX) or the C-terminally Flag-tagged human p34SEI-1 protein (pEF-p34SEI-1-Flag) for 6 h in serum free medium using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA). For knockdown of p34SEI-1 expression, MDA-MB231 cells were plated as above and transfected with 4 μg of either empty control vector (pLKO.1) or p34SEI-1 directed shRNA expression vector [pLKO.1/p34SEI-1-short hairpin (sh) RNA] for 6 h in serum free medium using Lipofectamine 2000. After replacing the DNA-Lipofectamine complex-containing medium with complete, antibiotic-free growth medium, transfected cells were incubated for 48 h. For knockdown of endogenous HER2/neu, SKBR3 cells were plated 4×105 in a 60-mm-diameter culture dish and transiently transfected with 200 pmol of either scramble control RNA (scRNA) or a HER2/neu silencing siRNA (siHER2/neu) for 2 h in serum free medium using Lipofectamine 2000. After replacing the RNA-Lipofectamine complex-containing medium with complete growth medium, transfected cells were incubated for 48 h. pEF-BOS-EX and pEF-p34SEI-1-Flag plasmids were kindly provided by Dr Rikiro Fukunaga (Osaka University, Osaka, Japan) and pLKO.1/p34SEI-1-shRNA plasmid was obtained from OriGene (http://www.origene.com). HER2/neu oligonucleotides were chemically synthesized by ST Pharm Co. Ltd (Seoul, Korea).

Wound healing migration assay

Cell migration was estimated by a wound-healing migration assay and monitored by microscopy. MCF7 and MDA-MB231 cells were transfected with p34SEI-1 overexpressing pEF-p34SEI-1-Flag or p34SEI-1 suppressing pLKO.1/p34SEI-1-shRNA vector with each corresponding control vector, respectively. Each cell line was fully cultured in a 60-mm-diameter culture dish and a scratch was made on the monolayer using a sterile white tip. The distance of migration by cancer cells was measured after 48 h.

Matrigel invasion assay

In vitro invasion assay was performed using a Transwell membrane apparatus (Corning Life Sciences, Tewksbury, MA, USA) and Matrigel (BD Biosciences, Seoul, Korea). SKBR3, MCF7 and MDA-MB-231 breast cancer cells (2×105 cells) were placed in the Matrigel-coated upper chamber of the apparatus. Medium to each cell line was placed in the lower chamber as a source of chemo-attractants. Incubation was carried out for 24 h at 37°C. The capacity of these cells to invade through the semi-solid Matrigel was estimated by fluorescence.

Immunohistochemistry

IHC data using human tissue samples were kindly provided by Dr Chang-Jin Kim at Soonchunhyang University Hospital (Chonan, Korea) and it was obtained as previously described (28).

Results

Enhanced cancer cell migration and invasion by p34SEI-1 overexpression

To investigate whether p34SEI-1 oncoprotein is involved in the development of metastasis, the effect of p34SEI-1 on migration and invasion was tested by wound healing migration and Matrigel invasion assays. In the wound healing assay, cell mobility was enhanced in MCF7 cells transfected with p34SEI-1 overexpressing pEF-p34SEI-1-Flag vector, while it was reduced in MDA-MB-231 cells transfected with p34SEI-1 suppressing pLKO.1/p34SEI-1-shRNA vector compared to control cells (Fig. 1A). The Matrigel invasion assay showed that overexpression of p34SEI-1 in SKBR3 and MCF7 cells increased the invasiveness compared to control cells, while knockdown of p34SEI-1 in MDA-MB 231 cells decreased invasiveness (Fig. 1B). These data strongly suggest that p34SEI-1 exerts a positive effect on the cell migration and invasion in vitro implying the involvement of p34SEI-1 in metastasis. During the EMT process, epithelial markers like E-cadherin are diminished, while expression of mesenchymal markers including N-cadherin, vimentin and fibronectin increase due to activation of matrix metalloproteinases (MMPs) inducing metastasis (38). Accordingly, the expression levels of EMT-related proteins were checked using western blot analysis. The representative epithelial marker E-cadherin was decreased but its antagonist N-cadherin was increased in SKBR3 and MCF7 cells after transfection with pEF-p34SEI-1-Flag vector (Fig. 1C). Furthermore, vimentin another mesenchymal marker, was also increased in SKBR3 and MCF7 cells. The opposite result was obtained in MDA-MB-231 cells transfected with pLKO.1/p34SEI-1-shRNA vector compared to control (Fig. 1C). Taken together, the findings indicate that p34SEI-1 promotes metastasis by enhancing migration and invasion of cancer cells.

Change of p34SEI-1 expression level in tissue samples with different degrees of tumor invasiveness

To support the conclusion that p34SEI-1 has metastatic potential, immunohistochemistry was performed to test whether p34SEI-1 expression is clinically related with the metastasis of breast cancer. Indicated tissue samples were stained with p34SEI-1 antibody and the degree of positive signals was estimated. Normal lobular and ductal hyperplastic cells showed no strong positive signal (0 of 20 samples) while 8 of 20 (40%) ductal carcinoma in situ and 13 of 20 (70%) invasive ductal carcinoma samples displayed strong positive signals (Fig. 2). The invasive carcinoma showed more strong expression than non-invasive ductal carcinoma in situ. The representative immunohistochemistry data are shown in Fig. 2, in which p34SEI-1 was not or very weakly expressed in normal lobular epithelial or ductal hyperplasia breast cancer tissues, but was more strongly expressed in invasive ductal carcinoma tissue samples than in ductal carcinoma in situ. The data indicate that p34SEI-1 expression increases as the tumor invasiveness progresses in human breast tissues, strongly relating p34SEI-1 with breast cancer metastasis.

Positive effect of p34SEI-1 on metastasis via regulation of PI3K/AKT signaling pathway

To elucidate the mechanism of how p34SEI-1 promotes metastasis during cancer cell tumorigenesis, western blot analysis was employed to check the expression levels of the main components of the PI3K-AKT pathway, NEDD4-1, PTEN, phosphorylation of AKT on serine 473 residue (pAKTser473) and phosphorylation of GSK3β on serine 9 residue (pGSK3βser9) after overexpression or suppression of p34SEI-1. In MCF7, T47D and HEK293T cells transfected with pEF-p34SEI-1-Flag, p34SEI-1 induced increased pAKTser473 and pGSK3βser9 protein levels at least partly by inducing NEDD4-1-mediated PTEN degradation as we previously reported (20). In MDA-MB231 cells transfected with pLKO.1/p34SEI-1-shRNA vector, suppression of p34SEI-1 resulted in a decrease of NEDD4-1 and an increase of PTEN compared to the control. Consequently, the protein levels of pAKTser473 and pGSK3βser9 decreased (Fig. 3A). However, SKBR3 cells transfected with pEF-p34SEI-1-Flag revealed a different expression pattern, in which pAKTser473 was unexpectedly diminished. More interestingly, expression levels of GSK3β phosphorylation and β-catenin, downstream target of GSK3β, were not affected by the AKT inactivation (Fig. 3A). This fact implies the presence of another upstream kinase regulating GSK3β phosphorylation regardless of AKT inactivation. This speculation was supported by RT-PCR results showing that both classes had very similar expression pattern to the NEDD4-1, β-catenin, and its downstream target, c-MYC, at the transcriptional level. Overexpression of p34SEI-1 produced an increase of NEDD4-1, β-catenin and c-MYC in SKBR3, MCF7 and HEK293T cells, whereas, in MDA-MB231 cells p34SEI-1 was suppressive (Fig. 3B). The data implicated an unknown kinase in this pathway downstream of NEDD4-1 and upstream of GSK3β. Collectively, the data indicate that p34SEI-1 may promote cancer metastasis using distinct signaling pathways in two different types of cancer cell lines with different genetic background.

Effect of p34SEI-1 overexpression on the activation of AKT and ILK in HER2/neu-positive and -negative cancer cell lines

p34SEI-1 seems to promote metastasis by activating the PI3K/AKT signaling pathway. During this process, overexpression of p34SEI-1 decreased the phosphorylation of AKT on 473 serine residue in SKBR3 cells but increased phosphorylation in MCF7, T47D and HEK293T cells. Considering that SKBR3 is HER2/neu-positive cell line, but MCF7, T47D and HEK293T are HER2/neu-negative cell lines, it was assumed that HER2/neu might be responsible for the decrease of pAKTser473 protein level in p34SEI-1 overexpressing SKBR3 cells, since HER2/neu is a positive regulator of the PI3K/AKT signaling pathway and its expression was significantly decreased by p34SEI-1 overexpression (Fig. 4A). This result was consistent with the view that p34SEI-1 suppresses HER2/neu expression and downregulated HER2/neu inhibits the phosphorylation of AKT at the 473 serine residue. Unexpectedly, the phosphorylation level of GSK3β on 9 serine residue was increased despite AKT inactivation (Fig. 3A). This finding indicated that GSK3β might be phosphorylated by another factor rather than AKT in SKBR3 cells. To elucidate the mechanism, ILK was at first suspected to be responsible for GSK3β phosphorylation because ILK is known to affect PI3K/AKT signaling pathway by directly phosphorylating the 9 serine residue of GSK3β (23). The phosphorylation levels of ILK on the 178 threonine residue were checked in both HER2/neu-positive and -negative cell lines. pILKthr178 protein level was significantly increased in HER2/neu-positive SKBR3 cells while no change was found in HER2/neu-negative MCF7, HEK293 and T47D cells (Fig. 4A). The protein level of pILKthr178 was significantly induced when phosphorylation of pAKTser473 was inhibited by decreased HER2/neu. However, it was not changed in HER2/neu-negative cells, in which AKT phosphorylation was induced (Fig. 4A). This observation indicates that p34SEI-1 overexpression promotes cancer metastasis by inducing ILK instead of AKT when HER2/neu is diminished or depleted. Further RT-PCR analysis showed that the negative effect of p34SEI-1 on HER2/neu expression occurred both at the transcriptional and the translational levels (Fig. 4B).

Taken together, the data demonstrates that p34SEI-1 activates the PI3K/AKT signaling pathway using at least two different types of signaling pathways depending on HER2/neu expression status.

HER2/neu-dependent switching relationship between AKT and ILK signaling pathways

Decreased HER2/neu activity by p34SEI-1 overexpression might be responsible for the activation of ILK signaling in SKBR3 cells. This idea prompted the assessment of the dependence of the activation of ILK on HER2/neu. The phosphorylation of AKT, ILK and GSK3β was checked at the protein level after SKBR3 and MCF7 cells were transfected with HER2/neu specific siRNA or overexpressing pHER2/neu vector. In HER2/neu silenced SKBR3 cells, pAKTser473 protein level was decreased, probably due to HER2/neu mediated inhibition of PI3K, a direct downstream target of HER2/neu. Surprisingly, both pILKthr178 and pGSK3βser9 protein levels were increased after treatment of HER2/neu silencing siRNA, in which GSK3β was thought to be phosphorylated by the ILK rather than AKT (Fig. 5A). On the other hand, the expression levels of same proteins were also checked after MCF7 was transfected with HER2/neu overexpressing vector. Overexpression of HER2/neu highly promoted the phosphorylation of AKT on 473 serine residue but reduced that of ILK on the 178 threonine residue. PTEN was not affected by neither HER2/neu inhibition or overexpression (Fig. 5A). In an extended experiment, the switching relationship between AKT and ILK was investigated using Cpd22 of ILK inhibitor and LY29002 of PI3K/AKT inhibitor. In both groups, PTEN was decreased whenever p34SEI-1 was overexpressed probably due to p34SEI-1 mediated NEDD4-1 activation as we showed before (28). Very importantly, both groups showed an inverse relationship between pAKTser473 and pILKthr178 expression levels. When HER2/neu-positive SKBR3 cells were treated with Cpd22, pAKTser473 protein level increased even under p34SEI-1 overexpression (Fig. 5B). When HER2/neu-negative MCF7 cells were treated with LY29002, the ratio of ILK phosphorylation was significantly elevated (Fig. 5B). LY29002 treatment after p34SEI-1 overexpression produced an even higher level of pILKthr178 expression (Fig. 5B). It may be explained by the strong switching relationship between AKT and ILK. In both cases, p34SEI-1 overexpression and treatment of Cpd22 and LY29002 tremendously increased the phosphorylation of GSK3β on the 9 serine residue in both HER2/neu-positive and -negative cell lines. This may be the basis for progression of metastasis because either AKT or ILK phosphorylation could promote the metastasis under the circumstance that one of them is inhibited (Fig. 5B).

Taken together, the data demonstrate that p34SEI-1 induces the activation of either AKT or ILK signaling in a HER2/neu-dependent manner (Fig. 6).

Discussion

The present study shows that p34SEI-1 exerts a positive effect on cancer metastasis by inducing migration and invasion of cancer cells. p34SEI-1 appears to promote metastasis by activating the PI3K/AKT signaling pathway, in which two different serine/threonine kinases, AKT and ILK, are alternatively activated depending on HER2/neu expression. In HER2/neu suppressed cells, p34SEI-1 overexpression increased pAKTser473 protein level and in turn activated PI3K/AKT signaling pathway at least partly via NEDD4-1 mediated PTEN ubiquitination. However, pAKTser473 protein was unexpectedly diminished despite exuberant p34SEI-1 overexpression in HER2/neu expressing SKBR3 cancer cells. In HER2/neu strongly positive SKBR3 cells, p34SEI-1 reduced HER2/neu, leading to the inhibition of AKT phosphorylation. Instead, p34SEI-1 activated another multifunctional serine/threonine protein kinase ILK to promote metastasis. This result suggests that p34SEI-1 affects cancer metastasis in a HER2/neu-dependent manner. Interestingly, our data also showed the inverse relationship in the expression levels of AKT and ILK proteins, implying that AKT and ILK have a switching relationship with each other in HER2/neu-dependent manner. Considering all these results, we suggest that drug resistance or recurrence of metastatic cancer may be caused by the switching relationship between AKT and ILK after the treatment of HER2/neu overexpressing breast cancers with a monoclonal antibody targeting HER2/neu oncogene (39). However, the exact mechanism of HER2/neu inhibition by p34SEI-1 overexpression is still not clear and needs further study.

We showed that p34SEI-1 can downregulate HER2/neu at the transcriptional and protein levels. We also found that p21 enhancer activator 3 (PEA3) is at least partly responsible for the p34SEI-1 mediated HER2/neu downregulation at the transcription level (data not shown). Interestingly, p34SEI-1 overexpression increased the expression levels of PEA3 gene at the transcription level (data not shown). PEA3 can bind to the promoter region of HER2/neu directly (40). PEA3 facilitates cancer invasion via regulation of PI3K/AKT-related proteins and MMP13. Inhibition of PEA3 diminishes the non-adherent tumor growth, migration and invasion via downregulation of EMT markers in a variety of tumors including breast cancer (41). Therefore, we assumed that p34SEI-1 might negatively affect HER2/neu expression by regulating PEA3 gene expression or by interacting with it. However, more precise experiments such as ChIP assay need to be performed to make the mechanism clear.

In summary, our data demonstrate that p34SEI-1 activates the PI3K/AKT signaling pathway by positively regulating at least two different types of AKT or ILK-mediated signaling pathways depending on HER2/neu expression status. Taken together, p34SEI-1 would be considered as blocking metastatic breast cancer, and it might be used for the prevention and treatment of metastatic breast cancer.

Abbreviations:

p34SEI-1

34-kD protein encoding SEI-1 (selected with Ink4a-1 as bait) gene;

HER2/neu

human epidermal growth factor receptor 2;

ILK

integrin-linked kinase;

PI3K

phosphoinositide-3 kinase;

AKT

serine/threonine-specific protein kinase

Acknowledgements

This study was supported by Sookmyung Women’s University (2011).

References

1. 

Morgensztern D and McLeod HL: PI3K/Akt/mTOR pathway as a target for cancer therapy. Anticancer Drugs. 16:797–803. 2005. View Article : Google Scholar : PubMed/NCBI

2. 

Yap TA, Garrett MD, Walton MI, Raynaud F, de Bono JS and Workman P: Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises. Curr Opin Pharmacol. 8:393–412. 2008. View Article : Google Scholar

3. 

LoPiccolo J, Blumenthal GM, Bernstein WB and Dennis PA: Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat. 11:32–50. 2008. View Article : Google Scholar : PubMed/NCBI

4. 

Song G, Ouyang G and Bao S: The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med. 9:59–71. 2005. View Article : Google Scholar : PubMed/NCBI

5. 

DeFeo-Jones D, Barnett SF, Fu S, et al: Tumor cell sensitization to apoptotic stimuli by selective inhibition of specific Akt/PKB family members. Mol Cancer Ther. 4:271–279. 2005.PubMed/NCBI

6. 

Qiao M, Sheng S and Pardee AB: Metastasis and AKT activation. Cell Cycle. 7:2991–2996. 2008. View Article : Google Scholar

7. 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

8. 

Brader S and Eccles SA: Phosphoinositide 3-kinase signalling pathways in tumor progression, invasion and angiogenesis. Tumori. 90:2–8. 2004.

9. 

Stambolic V, Suzuki A, de la Pompa JL, et al: Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell. 95:29–39. 1998. View Article : Google Scholar : PubMed/NCBI

10. 

Akca H, Demiray A, Tokgun O and Yokota J: Invasiveness and anchorage independent growth ability augmented by PTEN inactivation through the PI3K/AKT/NFκB pathway in lung cancer cells. Lung Cancer. 73:302–309. 2011.PubMed/NCBI

11. 

Carver BS, Chapinski C, Wongvipat J, et al: Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell. 19:575–586. 2011. View Article : Google Scholar

12. 

Cully M, You H, Levine AJ and Mak TW: Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer. 6:184–192. 2006. View Article : Google Scholar : PubMed/NCBI

13. 

Moasser MM: The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene. 26:6469–6487. 2007. View Article : Google Scholar : PubMed/NCBI

14. 

Baselga J and Swain SM: Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 9:463–475. 2009. View Article : Google Scholar : PubMed/NCBI

15. 

Grille SJ, Bellacosa A, Upson J, et al: The protein kinase Akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 63:2172–2178. 2003.PubMed/NCBI

16. 

Toker A and Yoeli-Lerner M: Akt signaling and cancer: surviving but not moving on. Cancer Res. 66:3963–3966. 2006. View Article : Google Scholar : PubMed/NCBI

17. 

Yoeli-Lerner M and Toker A: Akt/PKB signaling in cancer: a function in cell motility and invasion. Cell Cycle. 5:603–605. 2006. View Article : Google Scholar : PubMed/NCBI

18. 

Gagnon V, St-Germain ME, Parent S and Asselin E: Akt activity in endometrial cancer cells: Regulation of cell survival through cIAP-1. Int J Oncol. 23:803–810. 2003.PubMed/NCBI

19. 

Cross DA, Alessi DR, Cohen P, Andjelkovich M and Hemmings BA: Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature. 378:785–789. 1995. View Article : Google Scholar : PubMed/NCBI

20. 

Luo J: Glycogen synthase kinase 3beta (GSK3beta) in tumorigenesis and cancer chemotherapy. Cancer Lett. 273:194–200. 2009. View Article : Google Scholar : PubMed/NCBI

21. 

Morin PJ: beta-catenin signaling and cancer. Bioessays. 21:1021–1030. 1999. View Article : Google Scholar

22. 

Guturi KK, Mandal T, Chatterjee A, et al: Mechanism of beta-catenin-mediated transcriptional regulation of epidermal growth factor receptor expression in glycogen synthase kinase 3 beta-inactivated prostate cancer cells. J Biol Chem. 287:18287–18296. 2012. View Article : Google Scholar

23. 

Hannigan GE, McDonald PC, Walsh MP and Dedhar S: Integrin-linked kinase: not so ‘pseudo’ after all. Oncogene. 30:4375–4385. 2011.

24. 

Acconcia F, Barnes CJ, Singh RR, Talukder AH and Kumar R: Phosphorylation-dependent regulation of nuclear localization and functions of integrin-linked kinase. Proc Natl Acad Sci USA. 104:6782–6787. 2007. View Article : Google Scholar

25. 

Wani AA, Jafarnejad SM, Zhou J and Li G: Integrin-linked kinase regulates melanoma angiogenesis by activating NF-kappaB/interleukin-6 signaling pathway. Oncogene. 30:2778–2788. 2011. View Article : Google Scholar : PubMed/NCBI

26. 

Tan C, Cruet-Hennequart S, Troussard A, et al: Regulation of tumor angiogenesis by integrin-linked kinase (ILK). Cancer Cell. 5:79–90. 2004. View Article : Google Scholar : PubMed/NCBI

27. 

Taylor CJ, Qiao J, Colon NC, Schlegel C, Josifi E and Chung DH: Integrin-linked kinase regulates phosphatase and tensin homologue activity to promote tumorigenesis in neuroblastoma cells. Surgery. 150:162–168. 2011. View Article : Google Scholar : PubMed/NCBI

28. 

Jung S, Li C, Jeong D, et al: Oncogenic function of p34SEI-1 via NEDD41 mediated PTEN ubiquitination/degradation and activation of the PI3K/AKT pathway. Int J Oncol. 43:1587–1595. 2013.

29. 

Wang X, Trotman LC, Koppie T, et al: NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell. 128:129–139. 2007. View Article : Google Scholar : PubMed/NCBI

30. 

Amodio N, Scrima M, Palaia L, et al: Oncogenic role of the E3 ubiquitin ligase NEDD4-1, a PTEN negative regulator, in non-small-cell lung carcinomas. Am J Pathol. 177:2622–2634. 2010. View Article : Google Scholar : PubMed/NCBI

31. 

Hayashi R, Goto Y, Ikeda R, Yokoyama KK and Yoshida K: CDCA4 is an E2F transcription factor family-induced nuclear factor that regulates E2F-dependent transcriptional activation and cell proliferation. J Biol Chem. 281:35633–35648. 2006. View Article : Google Scholar

32. 

Hsu SI, Yang CM, Sim KG, Hentschel DM, O’Leary E and Bonventre JV: TRIP-Br: a novel family of PHD zinc finger- and bromodomain-interacting proteins that regulate the transcriptional activity of E2F-1/DP-1. EMBO J. 20:2273–2285. 2001. View Article : Google Scholar : PubMed/NCBI

33. 

Hong SW, Kim CJ, Park WS, et al: p34SEI-1 inhibits apoptosis through the stabilization of the X-linked inhibitor of apoptosis protein: p34SEI-1 as a novel target for anti-breast cancer strategies. Cancer Res. 69:741–746. 2009.

34. 

Li Y, Nie CJ, Hu L, et al: Characterization of a novel mechanism of genomic instability involving the SEI1/SET/NM23H1 pathway in esophageal cancers. Cancer Res. 70:5695–5705. 2010. View Article : Google Scholar : PubMed/NCBI

35. 

Tang DJ, Hu L, Xie D, et al: Oncogenic transformation by SEI-1 is associated with chromosomal instability. Cancer Res. 65:6504–6508. 2005. View Article : Google Scholar : PubMed/NCBI

36. 

Van Themsche C, Leblanc V, Parent S and Asselin E: X-linked inhibitor of apoptosis protein (XIAP) regulates PTEN ubiquitination, content, and compartmentalization. J Biol Chem. 284:20462–20466. 2009.PubMed/NCBI

37. 

Mehrotra S, Languino LR, Raskett CM, Mercurio AM, Dohi T and Altieri DC: IAP regulation of metastasis. Cancer Cell. 17:53–64. 2010. View Article : Google Scholar

38. 

Arias AM: Epithelial mesenchymal interactions in cancer and development. Cell. 105:425–431. 2001. View Article : Google Scholar : PubMed/NCBI

39. 

Knuefermann C, Lu Y, Liu B, et al: HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells. Oncogene. 22:3205–3212. 2003. View Article : Google Scholar : PubMed/NCBI

40. 

Xing X, Wang SC, Xia W, et al: The ets protein PEA3 suppresses HER-2/neu overexpression and inhibits tumorigenesis. Nat Med. 6:189–195. 2000. View Article : Google Scholar : PubMed/NCBI

41. 

Yuen HF, McCrudden CM, Chan KK, et al: The role of Pea3 group transcription factors in esophageal squamous cell carcinoma. Am J Pathol. 179:992–1003. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2014
Volume 45 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jung S, Ohk J, Jeong D, Li C, Lee S, Duan J, Kim C, Lim J, Yang Y, Kim K, Kim K, et al: Distinct regulatory effect of the p34SEI-1 oncoprotein on cancer metastasis in HER2/neu-positive and -negative cells. Int J Oncol 45: 189-196, 2014
APA
Jung, S., Ohk, J., Jeong, D., Li, C., Lee, S., Duan, J. ... Lee, M. (2014). Distinct regulatory effect of the p34SEI-1 oncoprotein on cancer metastasis in HER2/neu-positive and -negative cells. International Journal of Oncology, 45, 189-196. https://doi.org/10.3892/ijo.2014.2403
MLA
Jung, S., Ohk, J., Jeong, D., Li, C., Lee, S., Duan, J., Kim, C., Lim, J., Yang, Y., Kim, K., Lee, M."Distinct regulatory effect of the p34SEI-1 oncoprotein on cancer metastasis in HER2/neu-positive and -negative cells". International Journal of Oncology 45.1 (2014): 189-196.
Chicago
Jung, S., Ohk, J., Jeong, D., Li, C., Lee, S., Duan, J., Kim, C., Lim, J., Yang, Y., Kim, K., Lee, M."Distinct regulatory effect of the p34SEI-1 oncoprotein on cancer metastasis in HER2/neu-positive and -negative cells". International Journal of Oncology 45, no. 1 (2014): 189-196. https://doi.org/10.3892/ijo.2014.2403