498 research outputs found

    Zinc transporter HKE4 as a new target in antihormone resistance of breast cancer

    Get PDF
    Background Oestrogen receptor-positive breast cancers develop resistance to anti-oestrogens by utilising alternative growth factor pathways as observed in our tamoxifen-resistant cell line (TAMR). These include EGFR, IGF1-R and Src signalling as well as increased growth and invasion. Zinc is elevated in breast cancer tissue and has been demonstrated to activate certain growth factor signalling pathways. We have tested the expression level of members of the LIV-1 family of zinc influx transporters and discovered that HKE4 (SLC39A7, ZIP7), previously shown by us capable of increasing the intracellular zinc levels, has increased expression in TAMR. We have therefore investigated whether the development of the more aggressive phenotype observed in our TAMR cells, including activation of these signalling pathways as well as increased growth and invasion, is due to an increase of intracellular zinc and as a direct result of increased expression of HKE4. Methods All nine members of the LIV-1 subfamily of ZIP transporters were measured in our model of tamoxifen-resistant breast cancer using Affymetrix arrays. Zinc-induced activation of growth factor signalling pathway components was investigated by western blot and/or fluorescent microscopy. Short-term (15-min) treatments with 20 μM zinc included ionophore, whereas long-term (hours/days) did not. Recombinant LIV-1 family members with a V5 tag were expressed using pcDNA3.1/V5-His-TOPO vector, and siRNA (Dharmacon smartpools with relevant controls) was used to reduce endogenous expression. Results HKE4 (SLC39A7), a ZIP transporter from the LIV-1 subfamily, was discovered to be elevated in TAMR cells by Affymetrix analysis and confirmed by PCR and western blot. We have observed that our TAMR cells have a twofold increase in intracellular zinc compared with wild-type cells, using the zinc-specific fluorescent dye Newport Green. Short-term zinc treatment of TAMR cells activates the signalling pathways implicated in antihormone-resistant proliferation and is reduced by both the zinc chelator TPEN and the Src kinase inhibitor SU6556. The same effects are observed after longer term (6 days) zinc treatment with additional increases in cell growth and invasion through Matrigel. Since we have previously demonstrated that HKE4 is capable of increasing intracellular zinc in cells and, more recently, that these TAMR have elevated intracellular zinc levels, we have tested the hypothesis that elevated HKE4 expression is directly responsible for the aggressive phenotype observed in our TAMR cells. Reducing HKE4 levels by siRNA demonstrated a role for this molecule in driving the zinc-induced activation of multiple signalling pathways. In the presence of siRNA for HKE4, the previously observed zinc-induced activation of EGFR, Src, and IGF1-R was eradicated and the EGF-stimulated activation was also decreased. Additionally, we have demonstrated the converse by transfecting recombinant HKE4 into wild-type cells and/or treating them with zinc to observe the activation of these signalling pathways and increases in invasive capability. Interestingly, we have observed a similar role of HKE4 in our model of faslodex-resistant breast cancer. Conclusion The presented results propose that HKE4, a member of the LIV-1 subfamily of ZIP transporters, is directly involved in the activation of the aggressive phenotype observed with the development of antihormone resistance, and as such is a potential new target for the prevention of resistance to antihormones in breast cancer progression

    Fulvestrant-induced expression of erbB3 and erbB4 sensitizes ER-positive breast cancer cells to heregulins

    Get PDF
    We have previously reported that induction of EGFR and erbB2 in response to antihormones may provide an early mechanism allowing breast cancer cells to evade the growth inhibitory action of such therapies and ultimately drive resistant growth. More recently, another member of the erbB receptor family, erbB3, has been implicated in antihormone resistance in breast cancer. In the present study we have investigated whether induction of erbB3, and related family member erbB4, may provide an alternative resistance mechanism to antihormonal action in a panel of four ER-positive breast cancer cell lines. MCF-7, T47D, BT474 and MDAMB361 cell lines were exposed to fulvestrant (100 nM) for 7 days, and effects on erbB3/4 signalling and growth were assessed. Effects of the erbB3/4 ligand heregulin-β1 were also examined in the absence and presence of fulvestrant. Fulvestrant potently reduced ER expression and transcriptional activity and significantly inhibited growth in all four cell lines. However, alongside this inhibitory activity, fulvestrant also consistently induced protein expression and activity of erbB4 in the four cell lines and also promoted erbB3, erbB2 and EGFR protein expression and activity in MCF-7 and T47D cells. Consequently, fulvestrant treatment sensitised each cell line to the actions of heregulin-β1 with enhanced erbB3/4-driven signalling activity and significant increases in cell proliferation being observed when compared with untreated cells. Indeed, in T47D and MDAMB361, heregulin-β1 was converted from a ligand having negligible or suppressive growth activity into one that potently promoted cell proliferation. Consequently, fulvestrant-induced growth inhibition was completely overridden by heregulin-β1 in all four cell lines. In conclusion, these findings would suggest that although antihormones, such as fulvestrant, may have potent acute growth inhibitory activity in ER-positive breast cancer cells, their ability to induce and sensitize cells to growth factors, such as heregulins, may serve to reduce and ultimately limit their inhibitory activity

    Microarray studies reveal novel genes associated with endocrine resistance in breast cancer

    Get PDF
    Background Endocrine resistance is a major hurdle in breast cancer management, and determining the underlying factors driving its growth and aggressive behaviour should vastly improve treatment. Methods Microarray technology (BD Atlas Plastic Human 12 K Microarrays; GeneSifter software), verified by PCR, western blotting and immunocytochemisty, was used to identify genes increased in acquired resistant models to tamoxifen (TamR) or faslodex (FasR) as potential predictive/prognostic markers and new therapeutic targets. Results Alongside known breast cancer genes (β-catenin, PEA3, vitronectin, CD44), two novel genes in endocrine resistance were revealed (the latter never previously described in breast cancer): a securin/cell cycle regulator Pituitary Tumour Transforming Gene-1 (PTTG1), and GDNF receptor-alpha 3 (GFRα3) reported to promote cell survival signalling via RET coreceptor. Altered levels of PTTG1, GFRα3, or their associated family members were observed in further endocrine resistant states, including an additional faslodex resistant model that has progressed to a highly-aggressive state (FasR-Lt) and XMCF-7 cells resistant to oestrogen deprivation. PTTG1 and GFRα3 induction were also implicated in limiting response to anti-EGFR agents currently in breast cancer trials, with GFRα3 ligand (artemin) largely overcoming drug response. mRNA studies in clinical disease revealed PTTG1 associated with lymph node spread, high tumour grade and proliferation, while GFRα3 was enriched in ER-negative tumours and those expressing EGFR, profiles implying roles in clinical resistance and aggressive tumour behaviour. Promisingly, PTTG1 or GFRα3 siRNA knockdown promoted cell kill and inhibited proliferation in the resistant models. Conclusion Cumulatively, these data indicate PTTG1 and GFRα3 may provide useful biomarkers, and perhaps clinically relevant therapeutic targets for multiple resistant states

    Overexpression of CD44 in acquired tamoxifen-resistant breast cancer cells augments their migratory response to heregulin beta 1

    Get PDF
    Background Acquired endocrine resistance in breast cancer cells is accompanied by altered growth factor receptor signalling [1] and a highly migratory cell phenotype [2]. Interestingly, in tamoxifen-resistant (TamR) MCF7 cells, our microarray analysis has demonstrated elevated levels of CD44, a transmembrane glycoprotein known to interact with, and modulate the function of, growth factor receptors [3]. Here we have explored the role of CD44 as a modulator of heregulin beta-1-induced migratory signalling in TamR cells. Methods Expression of CD44 (standard and v3 isoforms) were confirmed by RT-PCR and western blotting and their association with erbB family members determined by both immunofluorescence microscopy and immunoprecipitation. Activation of intracellular signalling following heregulin beta 1 treatment (10 ng/ml) in the presence or absence of CD44 (using siRNA-mediated inhibition) was determined by western blotting using phosphospecific antibodies. Cellular migration was determined by seeding cells (control and CD44 siRNA-treated) into fibronectin-coated transwell chambers (8.0 μm pore size) in the presence or absence of heregulin beta 1. After 24 hours, migratory cells were fixed, stained with crystal violet and counted. Results Both standard and v3 isoforms of CD44 were overexpressed in TamR cells at both gene and protein levels (mean fold increase in CD44s protein (TamR versus MCF7): 4.26 ± 1.2, P < 0.05). Moreover, CD44s and v3 colocalised with Her2 and Her3 receptors at the cell surface and were also detectable in Her2/Her3 cellular immunoprecipitates. Treatment of TamR cells with heregulin resulted in phosphorylation of erbB receptors together with a number of downstream signalling intermediates, including Akt, Src and FAK, and resulted in enhanced cellular migration. Significantly, heregulin-induced intracellular signalling was dramatically reduced in cells in which the expression of CD44 was suppressed (via siRNA), with a corresponding loss of heregulin-induced migratory behaviour (mean fold change in cell migration versus untreated control: 6.7 ± 1.1, P < 0.05 (heregulin beta 1); 1.8 ± 0.9 (CD44 siRNA); 1.47 ± 0.6, P < 0.05 (heregulin beta 1 + CD44 siRNA)). Conclusion These data demonstrate a role for CD44 as a modulator of erbB receptor function in endocrine-resistant breast cancer cells, where it augments heregulin beta 1 migratory signalling

    Recruitment of insulin receptor substrate-1 by erbB3 impacts on IGF-IR signalling in oestrogen receptor-positive breast cancer cells

    Get PDF
    Insulin-like growth factor receptor (IGF-IR) signalling classically involves phosphorylation of insulin receptor substrate-1 (IRS-1) to recruit key down-stream signalling pathways effecting breast cancer cell proliferation and survival. Recently, we have shown a further capacity for IRS-1 to associate with the epidermal growth factor receptor (EGFR/erbB1), with activation of EGFR promoting recruitment and phosphorylation of IRS-1 in an oestrogen receptor (ER)-positive tamoxifen-resistant breast cancer cell line. In this study, we examined recruitment of IRS-1 by another member of the erbB receptor family, erbB3, in three ER-positive breast cancer cell lines. Our studies revealed an interaction between erbB3 and IRS-1 in MCF-7, T47D and BT474 cells with HRGβ1 treatment significantly enhancing this recruitment and promoting IRS-1 phosphorylation at tyrosine (Y) 612, a specific phosphoinositide 3-kinase (PI3K) binding site. IRS-1 appears to play a key role in erbB3 signalling in MCF-7 and T47D cells as its knockdown using siRNA greatly impaired HRGβ1 signalling via PI3K/AKT in these cell lines. This novel interaction may have clinical relevance as immunohistochemical analysis of ER-positive breast cancer patient samples revealed IRS-1 Y612 expression positively correlated with total erbB3, p-AKT and Ki67 expression. Importantly, we found that recruitment of IRS-1 by erbB3 impaired IRS-1 recruitment by IGF-IR in both MCF-7 and T47D cells, whilst blockade of IGF-1R enhanced erbB3/IRS-1 interaction and sensitised both cell lines to HRGβ1. Consequently, blockade of erbB3 signalling enhanced the effects of IGF-IR inhibition in these cells. In conclusion, these and previous findings suggest that IRS-1 can be recruited to IGF-1R, EGFR and erbB3 in ER-positive breast cancer cells and this may provide an adaptive resistance mechanism when these receptors are targeted individually. Consequently co-targeting of IGF-IR and erbB receptors may prove to be a more effective strategy for the treatment of ER-positive breast cancer

    Fulvestrant-induced expression of ErbB3 and ErbB4 receptors sensitizes oestrogen receptor-positive breast cancer cells to heregulin β1

    Get PDF
    Introduction We have previously reported that induction of epidermal growth factor receptor and ErbB2 in response to antihormonal agents may provide an early mechanism to allow breast cancer cells to evade the growth-inhibitory action of such therapies and ultimately drive resistant cell growth. More recently, the other two members of the ErbB receptor family, ErbB3 and ErbB4, have been implicated in antihormone resistance in breast cancer. In the present study, we have investigated whether induction of ErbB3 and/or ErbB4 may provide an alternative resistance mechanism to antihormonal action in a panel of four oestrogen receptor (ER)-positive breast cancer cell lines. Methods MCF-7, T47D, BT474 and MDAMB361 cell lines were exposed to fulvestrant (100 nM) for seven days, and effects on ErbB3/4 expression and signalling, as well as on cell growth, were assessed. Effects of heregulin β1 (HRGβ1) were also examined in the absence and presence of fulvestrant to determine the impact of ER blockade on the capacity of this ErbB3/4 ligand to promote signalling and cell proliferation. Results Fulvestrant potently reduced ER expression and transcriptional activity and significantly inhibited growth in MCF-7, T47D, BT474 and MDAMB361 cells. However, alongside this inhibitory activity, fulvestrant also consistently induced protein expression and activity of ErbB3 in MCF-7 and T47D cells and ErbB4 in BT474 and MDAMB361 cell lines. Consequently, fulvestrant treatment sensitised all cell lines to the actions of the ErbB3/4 ligand HRGβ1 with enhanced ErbB3/4-driven signalling activity, reexpression of cyclin D1 and significant increases in cell proliferation being observed when compared to untreated cells. Indeed, in T47D and MDAMB361 HRGβ1 was converted from a ligand having negligible or suppressive growth activity into one that potently promoted cell proliferation. Consequently, fulvestrant-mediated growth inhibition was completely overridden by HRGβ1 in all four cell lines. Conclusions These findings suggest that although antihormones such as fulvestrant may have potent acute growth-inhibitory activity in ER-positive breast cancer cells, their ability to induce and sensitise cells to growth factors may serve to reduce and ultimately limit their inhibitory activity

    GP88 (PC-Cell Derived Growth Factor, progranulin) stimulates proliferation and confers letrozole resistance to aromatase overexpressing breast cancer cells

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Aromatase inhibitors (AI) that inhibit breast cancer cell growth by blocking estrogen synthesis have become the treatment of choice for post-menopausal women with estrogen receptor positive (ER<sup>+</sup>) breast cancer. However, some patients display de novo or acquired resistance to AI. Interactions between estrogen and growth factor signaling pathways have been identified in estrogen-responsive cells as one possible reason for acquisition of resistance. Our laboratory has characterized an autocrine growth factor overexpressed in invasive ductal carcinoma named PC-Cell Derived Growth Factor (GP88), also known as progranulin. In the present study, we investigated the role GP88 on the acquisition of resistance to letrozole in ER<sup>+ </sup>breast cancer cells</p> <p>Methods</p> <p>We used two aromatase overexpressing human breast cancer cell lines MCF-7-CA cells and AC1 cells and their letrozole resistant counterparts as study models. Effect of stimulating or inhibiting GP88 expression on proliferation, anchorage-independent growth, survival and letrozole responsiveness was examined.</p> <p>Results</p> <p>GP88 induced cell proliferation and conferred letrozole resistance in a time- and dose-dependent fashion. Conversely, naturally letrozole resistant breast cancer cells displayed a 10-fold increase in GP88 expression when compared to letrozole sensitive cells. GP88 overexpression, or exogenous addition blocked the inhibitory effect of letrozole on proliferation, and stimulated survival and soft agar colony formation. In letrozole resistant cells, silencing GP88 by siRNA inhibited cell proliferation and restored their sensitivity to letrozole.</p> <p>Conclusion</p> <p>Our findings provide information on the role of an alternate growth and survival factor on the acquisition of aromatase inhibitor resistance in ER<sup>+ </sup>breast cancer.</p
    corecore