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.
PLoS One ; 13(3): e0193779, 2018.
Article in English | MEDLINE | ID: mdl-29590154

ABSTRACT

BACKGROUND: The Na+/K+-ATPase (NKP) is an important ion transporter also involved in signal transduction. Its expression profile is altered in various tumours including that of the breast. We studied the effect of inhibiting NKP activity in non-tumorigenic breast cell line and in estrogen receptor positive and negative breast cancer cells. METHODS: Expression and localization of NKP and downstream signaling molecules were determined by RT-PCR, western blotting and immunofluorescence. Cell proliferation, apoptosis and cell cycle stage were determined using MTT, annexin V and flow cytometry. Cell motility and invasion were determined using wound healing and matrigel assays. Total matrix metalloproteinase (MMP) was determined by a fluorescence-based assay. RESULTS: NKP was mainly localized on the cell membrane. Its baseline expression and activity were enhanced in breast cancer compared to the non-tumorigenic breast cell line. Ouabain and 3,4,5,6-tetrahydroxyxanthone (TTX) treatment significantly inhibited NKP activity, which significantly reduced cell proliferation, motility, invasion and pH-induced membrane blebbing. EGF stimulation induced internalization of NKP from the cell membrane to the cytoplasm. Ouabain inhibited EGF-induced phosphorylation of Rac/cdc42, profillin, ERK1/2 and P70S6K. CONCLUSIONS: The NKP may offer a novel therapeutic target in breast cancer patients who have developed metastasis, aiming to improve therapeutic outcomes and enhance survival rate.


Subject(s)
Breast Neoplasms/enzymology , Cell Movement/physiology , Neoplasm Invasiveness/physiopathology , Sodium-Potassium-Exchanging ATPase/metabolism , Actins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/pathology , Cell Movement/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/metabolism , Gene Expression/drug effects , Humans , Hydrogen-Ion Concentration , Ouabain/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Xanthones/pharmacology
2.
Oncol Res ; 25(8): 1283-1295, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28276317

ABSTRACT

Current mainstream pharmacological options for the treatment of endocrine-resistant breast cancer have limitations in terms of their side effect profile and lack of discrimination between normal and cancer cells. In the current study, we assessed the responses of normal breast epithelial cells MCF10A, estrogen receptor-positive (ER+) MCF-7, and ER-silenced pII breast cancer cells to inhibitors (either individually or in combination) of downstream signaling molecules. The expression/activity of ERK1/2, p38 MAPK, and Akt was determined by Western blotting. Cell proliferation, motility, and invasion were determined using MTT, wound healing, and Matrigel assays, respectively. Morphological changes in response to variation in external pH were assessed by light microscopy. Our results demonstrated that the inhibitors of ERK1/2 (PD0325901), p38 MAPK (SB203580), and PI3K (LY294002) preferentially reduce breast cancer cell proliferation. In pII cells, they also reduced motility, invasion, and bleb formation induced by alkaline conditions. Combination treatment with lower concentrations of inhibitors was significantly more effective than single agents and was more effective against the cancer cell lines than the normal MCF10A. In contrast, the commonly used cytotoxic agent paclitaxel did not sufficiently discriminate between the MCF10A and the cancer cells. We concluded that combination therapy using ERK1/2 inhibitor and either p38 MAPK or PI3K inhibitor may provide a greater therapeutic benefit in treating breast cancer by specifically targeting cancer cells with lower doses of each drug than needed individually, potentially reducing unwanted side effects.


Subject(s)
Breast Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , MCF-7 Cells
3.
Mol Med Rep ; 13(4): 3311-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26936341

ABSTRACT

In the face of increasing resistance to the existing antibiotics, oxazolidinones (exemplified by linezolid) have been developed as promising antibacterial agents, but may have other useful actions. In the present study, a series of 5­(1H­1,2,3­triazoly) l­methyl­, 5­acetamidomethyl­morpholino and N­substituted­piperazino oxazolidinone derivatives were investigated to determine whether they are active against eukaryotic cells. An MTT assay, validated by cell counting, was used to assess the effect of nine oxazolidinone derivatives (concentrations 100 nM­10 µM) on the proliferation of MCF7 human breast cancer cells. The three most active compounds were then tested on MDA231 breast cancer cells. Cytotoxicity of the selected derivatives was determined by assessing the extent of apoptosis by flow cytometry. The antimetastatic potential of these compounds was assessed on MDA231 cells using wound healing and agarose invasion assays. The 5­triazolylmethyl piperazino­oxazolidinone derivatives containing 4­N­(2­chlorocinnamoyl), 4­N­(4­nitrobenzoyl) and 4­N­methylsulfonyl moieties exhibited the most potent cytostatic activity against cancer, inhibiting proliferation by up to 70%, in the same order as their reported antibacterial activity against Staphylococcus aureus, but at higher concentrations. Unexpectedly, several derivatives stimulated proliferation at 100 nM, well below their antibacterial minimum inhibitory concentrations. Certain compounds also retarded the motility and invasion of MDA231 cells. Three of the tested derivatives had no effect on the eukaryotic cell lines, demonstrating their preferential activity against bacteria. Two compounds actually stimulated eukaryotic cell proliferation. The remaining three exhibited potent cytostatic activity against and cancer cells, displaying differences in response at low and high concentrations, which may suggest multiple targets on eukaryotic cells. These latter compounds may be useful as anticancer agents.


Subject(s)
Oxazolidinones/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Flow Cytometry , Humans , MCF-7 Cells , Microbial Sensitivity Tests , Oxazolidinones/chemistry , Staphylococcus aureus/drug effects
4.
Int J Oncol ; 46(4): 1685-98, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25672508

ABSTRACT

De novo and acquired resistance to endocrine-based therapies in breast cancer occurs in parallel with epithelial to mesenchymal transition (EMT), which is associated with enhanced proliferative and metastatic potential, and poor clinical outcome. We have established several endocrine insensitive breast cancer lines by shRNA-induced depletion of estrogen receptor (ER) by transfection of MCF7 cells. All of these exhibit EMT. We have previously reported that brief exposure of specifically ER- breast cancer cells, to extracellular alkaline pH, results in cell rounding and segregation, and leads to enhanced invasive potential. In this study we describe more detailed morphological changes and compare these with cell exposure to acidic pH. Morphological changes and localization of various molecules critical for cell adhesion and motility, associated with pH effects, were assessed by live cell microscopy, electron microscopy, and immunofluorescence. Exposure of either ER- or ER+ breast cancer cells to extracellular acidic pH did not induce significant changes in morphological appearance. Conversely, brief exposure of specifically ER silenced cells, to alkaline pH, resulted in cell contractolation and formation of bleb-like actin-rich structures which were evenly distributed on the outer membrane. Integrin α2, FAK, and JAM-1 were found in the cytoplasm streaming into the newly formed blebs. These blebs appear to be related to cell polarity and movement. Pre-treatment with cytochalasin-D or inhibitors of Rho or MLCK prevented both contractolation and bleb formation. Our data suggest that the effect of pH on the microenvironment of endocrine resistant breast cancer cells needs to be more extensively investigated. Alkaline, rather than acidic pH, appears to induce dramatic morphological changes, and enhances their invasive capabilities, through re-organization of cortical actin.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Receptors, Estrogen/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Cell Polarity , Cytochalasin D/pharmacology , Female , Focal Adhesion Kinase 1/metabolism , Gene Silencing , Humans , Hydrogen-Ion Concentration , Integrin alpha2/metabolism , MCF-7 Cells , Receptors, Cell Surface/metabolism
5.
PLoS One ; 8(10): e76327, 2013.
Article in English | MEDLINE | ID: mdl-24098477

ABSTRACT

INTRODUCTION: Endocrine resistance in breast cancer is associated with enhanced metastatic potential and poor clinical outcome, presenting a significant therapeutic challenge. We have established several endocrine insensitive breast cancer lines by shRNA induced depletion of estrogen receptor (ER) by transfection of MCF-7 cells which all exhibit enhanced expression profile of mesenchymal markers with reduction of epithelial markers, indicating an epithelial to mesenchymal transition. In this study we describe their behaviour in response to change in extracellular pH, an important factor controlling cell motility and metastasis. METHODS: Morphological changes associated with cell exposure to extracellular alkaline pH were assessed by live cell microscopy and the effect of various ion pumps on this behavior was investigated by pretreatment with chemical inhibitors. The activity and expression profile of key signaling molecules was assessed by western blotting. Cell motility and invasion were examined by scratch and under-agarose assays respectively. Total matrix metalloproteinase (MMP) activity and specifically of MMP2/9 was assessed in conditioned medium in response to brief alkaline pH exposure. RESULTS: Exposure of ER -ve but not ER +ve breast cancer cells to extracellular alkaline pH resulted in cell shrinkage and spherical appearance (termed contractolation); this was reversed by returning the pH back to 7.4. Contractolation was blocked by targeting the Na(+)/K(+) and Na(+)/H(+) pumps with specific chemical inhibitors. The activity and expression profile of key signaling molecules critical for cell adhesion were modulated by the exposure to alkaline pH. Brief exposure to alkaline pH enhanced MMP2/9 activity and the invasive potential of ER -ve cells in response to serum components and epithelial growth factor stimulation without affecting unhindered motility. CONCLUSIONS: Endocrine resistant breast cancer cells behave very differently to estrogen responsive cells in alkaline pH, with enhanced invasive potential; these studies emphasise the crucial influence of extracellular pH and caution against indiscriminate application of alkalinising drug therapy.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Gene Silencing , Receptors, Estrogen/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Enzyme Activation , Extracellular Space/metabolism , Female , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Ion Pumps/antagonists & inhibitors , Ion Pumps/metabolism , MCF-7 Cells , Matrix Metalloproteinases/metabolism , Neoplasm Metastasis , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
6.
PLoS One ; 8(1): e54361, 2013.
Article in English | MEDLINE | ID: mdl-23335999

ABSTRACT

PURPOSE: To examine whether (99m)Tc(V)-DMSA could be used as a non-invasive measure of cancer cell proliferation. METHODS: Human breast cancer MCF-7, MDA-MB-231 and pII, and prostate cancer PC-3 cell lines were grown to 30, 50 and 100% confluency and pulsed with (99m)Tc(V)-DMSA in media for 60 min at 37°C. DNA synthesis was analysed by quantification of the S phase using flow cytometry, [methyl-(3)H]thymidine incorporation and expression of proliferation markers PCNA and Ki-67 using realtime PCR. One way ANOVA was used to compare groups. RESULTS: In all cell lines rates of (99m)Tc(V)-DMSA uptake were inversely related to cell density. This was paralleled by similar trends in S phase proportions, [methyl-(3)H]thymidine incorporation and expression of PCNA and Ki-67. CONCLUSION: Rates of (99m)Tc(V)-DMSA uptake into different types of tumour cells correlate well with cell density that is useful as a non-invasive measure of tumour cellular proliferation in vivo.


Subject(s)
Radiopharmaceuticals/metabolism , Technetium Tc 99m Dimercaptosuccinic Acid/metabolism , Benzamides/pharmacology , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Fluorodeoxyglucose F18/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , MCF-7 Cells , Neoplasms/genetics , Neoplasms/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , S Phase/drug effects
7.
PLoS One ; 7(7): e41847, 2012.
Article in English | MEDLINE | ID: mdl-22860018

ABSTRACT

We have established several breast cancer cell lines that exhibit a permanent ER-depleted phenotype, induced by shRNA transfection of MCF-7 cells, which afford a useful model for studying acquired endocrine resistance. Previously we showed that MDA-231 as well as ER-silenced cells could invade through simulated extracellular matrix components. However, the contribution of individual serum components responsible for cell invasion was not determined. In the present study, an under-agarose gel assay was used to quantitatively assess the invasive movement of two ER-silenced cell lines (pII and YS2.5) in comparison to the parental MCF-7, the ER negative MDA-231, and normal HBL100 cells, as well as a line that was ER-shRNA transfected but failed to exhibit ER down-regulation (YS1.2). We also examined the effect of the growth factors EGF, IGF-1, TGFß, PDGFC and RANTES on pII cell invasion and proliferation. All breast cancer cell lines which had reduced ER expression exhibited a serum-dependent invasive ability related to the degree of induced ER loss. TGFß treatment inhibited pII cell proliferation and enhanced their invasive ability but at a relatively high dose. IGF-1 and EGF enhanced pII cell proliferation, with the latter playing the major role in promoting cell invasion. PDGFC did not affect either process although it is highly expressed in pII cells. Differential effects were observed on activation of Akt and ERK1/2 suggesting their involvement as intracellular mediators of EGF induced invasion, in part through the regulation of matrix metalloproteinase activity. Targeting EGF receptor tyrosine kinase activity by erlotinib resulted in significant inhibition of both pII cell proliferation and directional invasion towards EGF suggesting that this drug has potential therapeutic usefulness for preventing spread of particularly endocrine resistant breast cancer.


Subject(s)
Cell Movement , Cell Proliferation , Chemokine CCL5/physiology , Epidermal Growth Factor/physiology , Insulin-Like Growth Factor I/physiology , Lymphokines/physiology , Platelet-Derived Growth Factor/physiology , Transforming Growth Factor beta/physiology , Benzamides/pharmacology , Breast Neoplasms , Chromones/pharmacology , Collagenases/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , Morpholines/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , RNA, Small Interfering/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...