Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Neoplasia ; 22(11): 576-589, 2020 11.
Article in English | MEDLINE | ID: mdl-32980776

ABSTRACT

There are data to suggest that some ductal carcinoma in situ (DCIS) may evolve through an evolutionary bottleneck, where minor clones susceptible to the imposed selective pressure drive disease progression. Here, we tested the hypothesis that an impact of the inflammatory environment on DCIS evolution is HER2-dependent, conferring proliferative dominance of HER2-negative cells. In tissue samples, density of tumour-infiltrating immune cells (TIICs) was associated only with high tumour nuclear grade, but in 9% of predominantly HER2-negative cases, the presence of tumoral foci ('hot-spots') of basal-like cells with HIF1-α activity adjacent to the areas of dense stromal infiltration was noted. Results of in vitro analyses further demonstrated that IL-1ß and TNF-α as well as macrophage-conditioned medium triggered phosphorylation of NF-κB and subsequent upregulation of COX2 and HIF1-α, exclusively in HER2-negative cells. Treatment with both IL-1ß and TNF-α resulted in growth stimulation and inhibition of HER2-negative and HER2-positive cells, respectively. Moreover, ectopic overexpression of HIF1-α rescued HER2-positive cells from the negative effect of IL-1ß and TNF-α on cell growth. Our data provide novel insight into the molecular basis of HER2-dependent proliferation of DCIS cells and indicate the NF-κB/COX2 → HIF1-α signalling axis as a dominant mechanism of DCIS evolution induced by inflammatory microenvironment. Presented findings also highlight the clinical significance of heterogeneity of DCIS tumours and suggest that HIF1-α might be considered as a predictive marker of disease progression.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/etiology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Cyclooxygenase 2/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , NF-kappa B/metabolism , Biomarkers , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Cytokines/metabolism , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Inflammation Mediators , Neoplasm Grading , Neoplasm Staging , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
2.
Transl Res ; 207: 44-55, 2019 05.
Article in English | MEDLINE | ID: mdl-30639369

ABSTRACT

CD151/Tspan24 (SFS-1, PETA3) is one of the best characterized members of the tetraspanin family, whose involvement in breast cancer (BCa) progression was demonstrated both in vitro and in vivo. We have recently reported that in ErbB2-overexpressing BCa cells grown in 3D laminin-rich extracellular matrix, CD151 regulated basal phosphorylation and homodimerization of ErbB2 and sensitized the cells to Herceptin (trastuzumab). Following from these data, we have here analyzed an involvement of CD151 in regulation of ErbB2/ErbB3 heterodimerization and its impact on cell response to Herceptin. CD151 was found to: (1) impair ErbB2/ErbB3 heterodimerization, (2) inhibit heregulin-dependent cell growth in 3D and signaling, and (3) counteract the protective effect of heregulin on Herceptin-mediated growth inhibition. Analysis of tissue samples demonstrated for the first time clinical significance of CD151 in patients with ErbB2-overexpressing BCa undergone trastuzumab-based therapy. Consistent with in vitro results, CD151 impact on disease outcome was ErbB3-dependent. In patients with ErbB3-negative tumors, CD151 significantly improved both overall survival (OS) (hazard ratio [HR] = 0.19, P = 0.034) and progression-free survival (PFS) (HR = 0.36, P = 0.043), while in ErbB3-positive cases it had no significant effect on patient survival (OS: HR = 3.33, P = 0.283; PFS: HR = 2.40, P = 0.208). These results support previous findings and show that CD151 acts as an important component of ErbB2 signaling axis in BCa cells, affecting their sensitivity to ErbB2-targeting therapy.


Subject(s)
Breast Neoplasms/metabolism , Protein Multimerization , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Tetraspanin 24/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease-Free Survival , Female , Humans , Middle Aged , Neuregulin-1/pharmacology , Protein Multimerization/drug effects , Signal Transduction/drug effects , Trastuzumab/pharmacology
3.
Oncol Lett ; 15(2): 1817-1822, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29434878

ABSTRACT

Breast cancer (BCa) is the most common cancer affecting women worldwide. Overexpression of human epidermal growth factor receptor 2 (HER2) occurs in ~20-25% of invasive ductal breast carcinomas and is associated with the more aggressive phenotype. Herceptin, a humanized antibody against HER2, is a standard therapy in HER2-overexpressing cases. Approximately one-third of patients relapse despite treatment. Therefore numerous studies have investigated the molecular mechanisms associated with Herceptin resistance. An interaction between HER2 signalling and steroid hormone receptor signalling pathways has been previously investigated, but the effect of this relationship on Herceptin resistance has never been studied. The present study analysed an impact of the steroid hormone, progesterone (PG), on Herceptin-dependent cell growth inhibition. Results indicated that Herceptin-inhibited proliferation of breast cancer cell lines overexpressing HER2 (BT474 and MCF/HER2) in 3D culture is abolished by PG. Furthermore, results demonstrated that PG led to the activation of HER2/HER3-mediated signalling. Moreover, PG treatment induced a shift of Herceptin-dependent cell cycle arrest in G1 phase towards S and G2 phases with concomitant upregulation of cyclin-dependent kinase 2 (CDK2) and downregulation of CDK inhibitor p27Kip1. These results demonstrate for the first time PG involvement in the failure of Herceptin treatment in vitro. The present observations suggest that cross-talk between PG- and HRG/HER2-initiated signalling pathways may lead to the acquisition of resistance to Herceptin in patients with BCa.

4.
Neoplasia ; 19(10): 791-804, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28869838

ABSTRACT

Signaling mediated by growth factors receptors has long been suggested as one of the key factors responsible for failure of endocrine treatment in breast cancer (BCa). Herein we present that in the presence of tamoxifen, FGFs (Fibroblast Growth Factors) promote BCa cell growth with the strongest effect being produced by FGF7. FGFR2 was identified as a mediator of FGF7 action and the FGFR2-induced signaling was found to underlie cancer-associated fibroblasts-dependent resistance to tamoxifen. FGF7/FGFR2-triggered pathway was shown to induce ER phosphorylation, ubiquitination and subsequent ER proteasomal degradation which counteracted tamoxifen-promoted ER stabilization. We also identified activation of PI3K/AKT signaling targeting ER-Ser167 and regulation of Bcl-2 expression as a mediator of FGFR2-promoted resistance to tamoxifen. Analysis of tissue samples from patients with invasive ductal carcinoma revealed an inversed correlation between expression of FGFR2 and ER, thus supporting our in vitro data. These results unveil the complexity of ER regulation by FGFR2-mediated signaling likely to be associated with BCa resistance to endocrine therapy.


Subject(s)
Estrogen Receptor alpha/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction/drug effects , Tamoxifen/pharmacology , Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fibroblast Growth Factors/metabolism , Fibroblasts/drug effects , Gene Expression , Gene Knockout Techniques , Humans , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Proteolysis , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics
5.
Oncotarget ; 7(52): 86011-86025, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27852068

ABSTRACT

We have recently demonstrated that, fibroblast growth factor 2 (FGFR2), signalling via ribosomal S6 kinase 2 (RSK2), promotes progression of breast cancer (BCa). Loss of progesterone receptor (PR), whose activity in BCa cells can be stimulated by growth factor receptors (GFRs), is associated with poor patient outcome. Here we showed that FGF7/FGFR2 triggered phosphorylation of PR at Ser294, PR ubiquitination and subsequent receptor`s degradation via the 26S proteasome pathway in BCa cells. We further demonstrated that RSK2 mediated FGF7/FGFR2-induced PR downregulation. In addition, a strong synergistic effect of FGF7 and progesterone (Pg), reflected in the enhanced anchorage-independent growth and cell migration, was observed. Analysis of clinical material demonstrated that expression of PR inversely correlated with activated RSK (RSK-P) (p = 0.016). Patients with RSK-P(+)/PR(-) tumours had 3.629-fold higher risk of recurrence (p = 0.002), when compared with the rest of the cohort. Moreover, RSK-P(+)/PR(-) phenotype was shown as an independent prognostic factor (p = 0.006). These results indicate that the FGF7/FGFR2-RSK2 axis promotes PR turnover and activity, which may sensitize BCa cells to stromal stimuli and contribute to the progression toward steroid hormone negative BCa.


Subject(s)
Breast Neoplasms/metabolism , Fibroblast Growth Factor 7/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Receptors, Progesterone/metabolism , Signal Transduction/physiology , Breast Neoplasms/mortality , Cell Line, Tumor , Female , Humans , Proteasome Endopeptidase Complex/physiology , Ribosomal Protein S6 Kinases, 90-kDa/physiology
6.
Br J Cancer ; 113(9): 1350-7, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26418423

ABSTRACT

BACKGROUND: The proposed involvement of CD151 in breast cancer (BCa) progression is based on findings from studies in invasive ductal carcinoma (IDC). The IDC and invasive lobular carcinoma (ILC) represent distinct disease entities. Here we evaluated clinical significance of CD151 alone and in association with integrin α3ß1 in patients with ILC in context of the data of our recent IDC study. METHODS: Expression of CD151 and/or integrin α3ß1 was evaluated in ILC samples (N=117) using immunohistochemistry. The findings were analysed in relation to our results from an IDC cohort (N=182) demonstrating a prognostic value of an expression of CD151/integrin α3ß1 complex in patients with HER2-negative tumours. RESULTS: Unlike in the IDCs, neither CD151 nor CD151/α3ß1 complex showed any correlation with any of the ILC characteristics. Lack of both CD151 and α3ß1 was significantly correlated with poor survival (P=0.034) in lymph node-negative ILC N(-) cases. The CD151(-)/α3ß1(-) patients had 3.12-fold higher risk of death from BCa in comparison with the rest of the ILC N(-) patients. CONCLUSIONS: Biological role of CD151/α3ß1 varies between ILC and IDC. Assessment of CD151/α3ß1 might help to identify ILC N(-) patients with increased risk of distant metastases.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Lobular/metabolism , Integrin alpha3beta1/metabolism , Lymph Nodes/pathology , Tetraspanin 24/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Female , Humans , Immunohistochemistry/methods , Middle Aged , Prognosis , Receptor, ErbB-2/metabolism
7.
Biochim Biophys Acta ; 1843(11): 2461-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25014166

ABSTRACT

The members of p90 ribosomal S6 kinase (RSK) family of Ser/Thr kinases are downstream effectors of MAPK/ERK pathway that regulate diverse cellular processes including cell growth, proliferation and survival. In carcinogenesis, RSKs are thought to modulate cell motility, invasion and metastasis. Herein, we have studied an involvement of RSKs in FGF2/FGFR2-driven behaviours of mammary epithelial and breast cancer cells. We found that both silencing and inhibiting of FGFR2 attenuated phosphorylation of RSKs, whereas FGFR2 overexpression and/or its stimulation with FGF2 enhanced RSKs activity. Moreover, treatment with ERK, Src and p38 inhibitors revealed that p38 kinase acts as an upstream RSK2 regulator. We demonstrate for the first time that in FGF2/FGFR2 signalling, p38 but not MEK/ERK, indirectly activated RSK2 at Tyr529, which facilitated phosphorylation of its other residues (Thr359/Ser363, Thr573 and Ser380). In contrast to FGF2-triggered signalling, inhibition of p38 in the EGF pathway affected only RSK2-Tyr529, without any impact on the remaining RSK phosphorylation sites. p38-mediated phosphorylation of RSK2-Tyr529 was crucial for the transactivation of residues located at kinase C-terminal domain and linker-region, specifically, in the FGF2/FGFR2 signalling pathway. Furthermore, we show that FGF2 promoted anchorage-independent cell proliferation, formation of focal adhesions and cell migration, which was effectively abolished by treatment with RSKs inhibitor (FMK). These indicate that RSK2 activity is indispensable for FGF2/FGFR2-mediated cellular effects. Our findings identified a new FGF2/FGFR2-p38-RSK2 pathway, which may play a significant role in the pathogenesis and progression of breast cancer and, hence, may present a novel therapeutic target in the treatment of FGFR2-expressing tumours.

SELECTION OF CITATIONS
SEARCH DETAIL
...