Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Mol Med Rep ; 29(1)2024 Jan.
Article in English | MEDLINE | ID: mdl-37997856

ABSTRACT

Lactate dehydrogenase (LDH) is being increasingly recognized as a major factor in the progression of breast cancer. It was previously shown that short interfering RNA­mediated knockdown of either LDH­A or ­B isoform resulted in inhibition of cell motility due to reduced lactate levels in the extracellular environment. The aim of the present study was to determine the use of pharmacological LDH inhibitors to reduce aggressive behavior of breast cancer cells. The effect of LDH inhibitors was investigated in both estrogen receptor (ER)+ and ER­ breast cancer cell lines and in normal breast epithelial cells. Cell proliferation, motility and invasion were measured using MTT, wound healing and cultrex assays, respectively. Changes in several key mediators of mitogenic signaling important in breast cancer cells were determined using western blotting. Treatment with various inhibitors reported to block LDH activity resulted in significant reduction in extracellular lactate level, cell proliferation, motility and invasion. This was associated with changes in the levels of vimentin, E­cadherin, p38 MAPK, ERK1/2 and AKT. A couple of these inhibitors such as quercetin and lonidamine showed preferential inhibition of cancer cell proliferation compared with normal epithelial cell inhibition. These data extend initial findings, further underlining the importance of lactate as a major factor in breast cancer progression and indicate the practical use of various commercially available LDH inhibitors as promising therapeutic agents to oppose the processes leading to cancer progression.


Subject(s)
Breast Neoplasms , L-Lactate Dehydrogenase , Humans , Female , L-Lactate Dehydrogenase/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Lactic Acid , Cell Proliferation , MCF-7 Cells , Receptors, Estrogen/metabolism , Cell Line, Tumor , Cell Movement
2.
PLoS One ; 18(7): e0289098, 2023.
Article in English | MEDLINE | ID: mdl-37490473

ABSTRACT

Chronic smoking is a primary risk factor for breast cancer due to the presence of various toxins and carcinogens within tobacco products. Nicotine is the primary addictive component of tobacco products and has been shown to promote breast cancer cell proliferation and metastases. Nicotine activates nicotinic acetylcholine receptors (nAChRs) that are expressed in cancer cell lines. Here, we examine the role of the α7 nAChR in coupling to heterotrimeric G proteins within breast cancer MCF-7 cells. Pharmacological activation of the α7 nAChR using choline or nicotine was found to increase proliferation, motility, and calcium signaling in MCF-7 cells. This effect of α7 nAChR on cell proliferation was abolished by application of Gαi/o and Gαq protein blockers. Specifically, application of the Gαi/o inhibitor pertussis toxin was found to abolish choline-mediated cell proliferation and intracellular calcium transient response. These findings were corroborated by expression of a G protein binding dominant negative nAChR subunit (α7345-348A), which resulted in significantly attenuating calcium signaling and cellular proliferation in response to choline. Our study shows a new role for G protein signaling in the mechanism of α7 nAChR-associated breast cancer growth.


Subject(s)
Breast Neoplasms , Heterotrimeric GTP-Binding Proteins , Receptors, Nicotinic , Humans , Female , Nicotine/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Calcium Signaling , Receptors, Nicotinic/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Cell Proliferation , Choline/pharmacology , Calcium/metabolism
3.
PLoS One ; 17(8): e0272449, 2022.
Article in English | MEDLINE | ID: mdl-35917304

ABSTRACT

BACKGROUND: Breast cancer chemotherapy with high dose alkylating agents is severely limited by their collateral toxicity to crucial normal tissues such as immune and gut cells. Taking advantage of the selective dependence of cancer cells on high glucose and combining glucose deprivation with these agents could produce therapeutic synergy. METHODS: In this study we examined the effect of glucose as well as its deprivation, and antagonism using the non-metabolized analogue 2-deoxy glucose, on the proliferation of several breast cancer cell lines MCF7, MDA-MB-231, YS1.2 and pII and one normal breast cell line, using the MTT assay. Motility was quantitatively assessed using the wound healing assay. Lactate, as the end product of anaerobic glucose metabolism, secreted into culture medium was measured by a biochemical assay. The effect of paclitaxel and doxorubicin on cell proliferation was tested in the absence and presence of low concentrations of glucose using MTT assay. RESULTS: In all cell lines, glucose supplementation enhanced while glucose deprivation reduced both their proliferation and motility. Lactate added to the medium could substitute for glucose. The inhibitory effects of paclitaxel and doxorubicin were significantly enhanced when glucose concentration was decreased in the culture medium, requiring 1000-fold lesser concentration to achieve a similar degree of inhibition to that seen in glucose-containing medium. CONCLUSION: Our data show that a synergy was obtained by combining paclitaxel and doxorubicin with glucose reduction to inhibit cancer cell growth, which in vivo, might be achieved by applying a carbohydrate-restricted diet during the limited phase of application of chemotherapy; this could permit a dose reduction of the cytotoxic agents, resulting in greater tolerance and lesser side effects.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Female , Glucose/metabolism , Humans , Lactates/pharmacology , Paclitaxel
4.
Biochem Biophys Rep ; 31: 101316, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35879960

ABSTRACT

Background: MicroRNAs (miRs) regulate gene expression through translation inhibition of target mRNAs. One of the most promising approaches for cancer therapy is through mimicking or antagonizing the action of miRs. In this report, we analyzed the miRnome profile of several human breast cancer cell lines to determine the influence of estrogen receptor (ER) silencing previously shown to result in epithelial to mesenchymal transition (EMT) and enhanced tumor invasion. Methods: MicroRNA extracted from MDA-MB-231 (de novo ER-) and ER-silenced (acquired ER-) pII and IM-26 or ER-expressing (YS1.2) siRNA transfected derivatives of MCF7 cells was deep sequenced on Illumina NextSeq500. Respective miRnomes were compared with edgeR package in R and Venny2.1 and target prediction performed with miRTarBase. Mimics and inhibitors of selected differentially expressed miRs associated with EMT mediators (miR-200c-3p targeting ZEB1, miR-449a targeting δ-catenin and miR-29a-3p) were transfected into pII cells and mRNA targets, as well as E-cadherin and keratin 19 (epithelial and mesenchymal markers respectively) were measured using taqman PCR. Results: Each cell line expressed about 20% of the total known human miRnome; There was a high degree of similarity between the 3 tested ER-lines. Out of these expressed miRs, 50-60% were significantly differentially expressed between ER- and ER + lines. Transfection of miR-200c-3p mimic into pII cells down regulated ZEB1 and vimentin, and increased E-cadherin and keratin 19 with accompanying morphological changes, and reduced cell motility, reflecting a reversal back into an epithelial phenotype. On the other hand, transfecting pII with miR-449a inhibitor reduced cell invasion but did not induce EMT. Transfecting pII cell line with the mimic or inhibitor of miR-29a-3p showed no change in EMT markers or cell invasion suggesting that the EMT induced by loss of ER function can be reversed by blocking some but not just any random EMT-associated genes. Conclusions: These data suggest that differences in miR expression can be exploited not only as mediators (using mimics) and targets (using miR antagonists) for general cancer therapies aimed at regulating either individual or multiple mRNAs, but also to re-sensitize endocrine resistant breast cancers by turning them back into a type that will be susceptible to endocrine agents.

5.
Med Princ Pract ; 31(1): 1-10, 2022.
Article in English | MEDLINE | ID: mdl-34662884

ABSTRACT

In 1 year, COVID-19 spread rapidly worldwide affecting all societies and most age-groups. It has taken not only a toll of human lives (approaching 220 million people infected with 4.55 million reported deaths at time of writing) but also decimated every economy as countries struggle to control infection rates by introducing draconian lockdown and social distancing measures, bringing great suffering well beyond medical effects of the disease. A parallel pandemic has resulted in a deluge of information emanating from both scientific as well as international news media including social media platforms. Fact and fiction, reality, and perception have become entangled; the only realistic solution, both medically as well as politically, is concerted global vaccination (which is currently underway) to reduce further infection by introducing universal immunity. However, public controversy rages due to widespread apprehension regarding necessity, immediate risks, and long-term safety of what is perceived as "fast-tracked" medication. While some concerns may be justified, much is due to misconception and misunderstanding. This review highlights some of the issues concerning the handling of the COVID-19 crisis by governments worldwide, the medical and scientific communities, and the media and how this may have laid the foundations for a far greater medical, social, and economic burden in the coming years. We present comparative data to challenge current conceptions of this disease in the more general context of human health to provide a perspective that seems to have been lost in the general panic. We need more rational approaches to the handling of a disease which is unlikely to disappear from our spectrum of afflictions even after the magnifying glass has been removed.


Subject(s)
COVID-19 , Communicable Disease Control , Pandemics/prevention & control , Social Media , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines , Humans , SARS-CoV-2
6.
Front Pharmacol ; 12: 747001, 2021.
Article in English | MEDLINE | ID: mdl-34744727

ABSTRACT

Background: Lactate dehydrogenase (LDH) plays an important role in cancer pathogenesis and enhanced expression/activity of this enzyme has been correlated with poor prognosis. In this study we determined the expression profile of LDH-A and B in normal as well as in endocrine-resistant and -responsive breast cancer cells and the effect of their knockdown on LDH activity, lactate production, proliferation and cell motility. Methods: Knockdown experiments were performed using siRNA and shRNA. The expression profile of LDH and signaling molecules was determined using PCR and western blotting. Intracellular LDH activity and extracellular lactate levels were measured by a biochemical assay. Cell motility was determined using wound healing, while proliferation was determined using MTT assay. Results: LDH-A was expressed in all of the tested cell lines, while LDH-B was specifically expressed only in normal and endocrine-resistant breast cancer cells. This was correlated with significantly enhanced LDH activity and lactate production in endocrine resistant breast cancer cells when compared to normal or endocrine responsive cancer cells. LDH-A or -B knockdown significantly reduced LDH activity and lactate production, which led to reduced cell motility. Exogenous lactate supplementation enhanced cell motility co-incident with enhanced phosphorylation of ERK1/2 and reduced E-cadherin expression. Also, LDH-A or -B knockdown reduced ERK 1/2 phosphorylation. Conclusion: Enhanced cell motility in endocrine resistant breast cancer cells is at least in part mediated by enhanced extracellular lactate levels, and LDH inhibition might be a promising therapeutic target to inhibit cancer cell motility.

7.
PLoS One ; 16(6): e0252820, 2021.
Article in English | MEDLINE | ID: mdl-34086816

ABSTRACT

Three scarce terpenes, psiadin, plectranthone and saudinolide, were obtained after chromatographic isolation and purification from the aerial parts of the respective plants. Their identities were established based on their spectral data. Their anticancer effects against two human colorectal carcinoma cell lines, CCL233 and CCL235, along with the potential molecular mechanisms of action, were explored. Psiadin and plectranthone exhibited marked growth inhibition on both cell lines in a time- and dose-dependent manner with minimal cytotoxicity against normal breast cells (HB2). The terpenes even showed superior activities to the tested standards. Flow cytometry showed apoptosis induction and alteration in the cell cycle in colorectal cancer cells treated with both compounds. Nevertheless, it was also found that both compounds inhibited NF-κB transcriptional activity, induced mitochondrial membrane potential depolarization and increased the percentage of reactive oxygen species in the treated cancer cells in a dose-dependent manner as well. Since the anticancer effect of psiadin on cancer cells was higher than that produced by plectranthone, only psiadin was tested to determine its possible targets. The results suggested a high degree of specificity of action affecting particular cellular processes in both cancer cells. In conclusion, both terpenes, in particular psiadin, showed significant discriminative therapeutic potential between cancer and normal cells, a value that is missing in current chemotherapies.


Subject(s)
Apoptosis , Colorectal Neoplasms , Cyclins , Diterpenes , Sesquiterpenes , Humans , Membrane Potential, Mitochondrial , Reactive Oxygen Species/metabolism
9.
Int J Oncol ; 56(4): 1014-1024, 2020 04.
Article in English | MEDLINE | ID: mdl-32319574

ABSTRACT

Estrogen receptor (ER)­silenced breast cancer cell lines exhibit endocrine resistance and morphological changes from an epithelial to a mesenchymal phenotype. These cells also display increased motility and invasive properties that are further accentuated by exposure to an alkaline pH, exhibiting dynamic plasma membrane blebbing and cytoplasmic streaming. These latter morphological changes are hypothesized to involve substantial water movement across the plasma membrane, contributing to bleb formation; this may involve aquaporin channel proteins (AQPs). AQP 1, 3, 4 and 5 expression/localization was examined via reverse transcription­quantitative PCR, western blotting and confocal microscopy in endocrine­sensitive (YS1.2) and ­resistant (pII and MDA­MB­231) breast cancer cells, as well as normal breast epithelial cells (MCF10A). The effects of osmotic changes on bleb formation were examined via live cell imaging. AQP3 protein expression was knocked down by small interfering RNA (siRNA) transfection, and the effect of its reduced expression on bleb formation, cell motility and invasion were determined via immunofluorescence, scratch and Cultrex assays, respectively. Expression of the four AQPs varied across the different cell lines, and exhibited nuclear, cytoplasmic and membranous localization. Osmotic changes affected the formation of blebs. In pII cells exposed to alkaline pH, AQP3 was observed to be redistributed from the nucleus into the newly formed blebs. siRNA­mediated knockdown of AQP3 in pII cells significantly reduced cellular blebbing induced by alkaline pH, as well as motility and invasion. These data suggested that AQP3, and potentially other aquaporins, may participate in the processes leading to blebbing of endocrine­resistant cells which is proposed to be a mechanism that drives tumor metastasis.


Subject(s)
Aquaporin 3/metabolism , Breast Neoplasms/pathology , Cell Membrane/pathology , Cell Movement , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Aquaporin 3/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation , Female , Humans , Neoplasm Invasiveness
10.
Sci Rep ; 10(1): 1127, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980706

ABSTRACT

The response of cancer cells to hypoxic conditions found within the interior of a tumor mass is mediated through the hypoxia inducible factor (HIF) cascade and is thought to promote metastasis. However, given their distant proximity from blood vessels as compared to normoxic cells at the vascularised tumor periphery, it is uncertain if these cells can migrate through the tumor mass to gain access. Hypoxia was simulated by exposure to cobalt chloride or deferoxamine in normal (MCF10A) and cancerous [estrogen receptor (ER)-ve (pII), and ER +ve (YS1.2/ EII)] cells. In this report, HIF1α expression and localization was measured using western blotting, ELISA, and immunofluorescence, cell proliferation by MTT assay, motility and invasion by wound healing, live cell imaging, matrigel and co-culture in chambered slides. We found that the expression and nuclear translocation of HIF1α was significantly elevated by hypoxia, which inhibited cell proliferation, but significantly increased motility of pII cells and their penetration into and through a dense layer of adjacent EII cells, as well as their selective emergence out of a co-culture. These data suggest that endocrine resistant pII cancer cells, having undergone epithelial to mesenchymal transition are able to penetrate through other cell layers, with possible enhancement in response to hypoxia.


Subject(s)
Cell Hypoxia , Epithelial-Mesenchymal Transition , MCF-7 Cells/physiology , Basement Membrane , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cobalt/pharmacology , Coculture Techniques , Deferoxamine/pharmacology , Estrogens , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MCF-7 Cells/drug effects , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Estrogen/analysis , Sepharose
11.
Int J Mol Sci ; 20(14)2019 Jul 19.
Article in English | MEDLINE | ID: mdl-31330984

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are important regulators of metabolism, making their receptors (GLP-1R and GIPR) attractive targets in the treatment of type 2 diabetes mellitus (T2DM). GLP-1R agonists are used clinically to treat T2DM but the use of GIPR agonists remains controversial. Recent studies suggest that simultaneous activation of GLP-1R and GIPR with a single peptide provides superior glycemic control with fewer adverse effects than activation of GLP-1R alone. We investigated the signaling properties of a recently reported dual-incretin receptor agonist (P18). GLP-1R, GIPR, and the closely related glucagon receptor (GCGR) were expressed in HEK-293 cells. Activation of adenylate cyclase via Gαs was monitored using a luciferase-linked reporter gene (CRE-Luc) assay. Arrestin recruitment was monitored using a bioluminescence resonance energy transfer (BRET) assay. GLP-1, GIP, and glucagon displayed exquisite selectivity for their receptors in the CRE-Luc assay. P18 activated GLP-1R with similar potency to GLP-1 and GIPR with higher potency than GIP. Interestingly, P18 was less effective than GLP-1 at recruiting arrestin to GLP-1R and was inactive at GCGR. These data suggest that P18 can act as both a dual-incretin receptor agonist, and as a G protein-biased agonist at GLP-1R.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon/metabolism , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Amino Acid Sequence , Arrestin/metabolism , Arrestin/pharmacology , Bioluminescence Resonance Energy Transfer Techniques , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ligands , Peptides/chemistry , Peptides/pharmacology , Receptors, Glucagon/antagonists & inhibitors
12.
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
13.
Dev Comp Immunol ; 74: 200-208, 2017 09.
Article in English | MEDLINE | ID: mdl-28487234

ABSTRACT

We recently demonstrated Ang 1-7 reduced inflammation in the dextran sulfate sodium (DSS) colitis model. In this study we examined the effect of Ang 1-7 on modulation of plasma levels of selected cytokines and chemokines and immune cell effector functions (apoptosis, chemotaxis and superoxide release) in vitro. The degree of neutrophil recruitment to the colon was assessed by immunofluorescence and myeloperoxidase activity. Daily Ang 1-7 treatment at 0.01 mg/kg dose which previously ameliorated colitis severity, showed a significant reduction in circulating levels of several cytokines and chemokines, and neutrophil recruitment to the colonic tissue. It also significantly enhanced immune cell apoptosis, and reduced neutrophil chemotaxis and superoxide release in vitro. In contrast, daily administration of the Ang 1-7R antagonist A779 which previously worsened colitis severity showed significant up-regulation of specific mediators. Our results demonstrate a novel anti-inflammatory action of Ang 1-7 through modulation of plasma levels of cytokines/chemokines and immune cell activity.


Subject(s)
Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Colitis/therapy , Colon/immunology , Neutrophils/immunology , Peptide Fragments/therapeutic use , Angiotensin I/antagonists & inhibitors , Angiotensin II/administration & dosage , Angiotensin II/analogs & derivatives , Animals , Apoptosis , Cell Movement , Chemokines/blood , Chemotaxis , Colitis/immunology , Cytokines/blood , Dextran Sulfate/immunology , Immunity, Cellular , Immunomodulation , Mice , Mice, Inbred BALB C , Models, Animal , Peptide Fragments/administration & dosage , Peptide Fragments/antagonists & inhibitors , Renin-Angiotensin System , Superoxides/metabolism
14.
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
15.
PLoS One ; 11(3): e0150861, 2016.
Article in English | MEDLINE | ID: mdl-26963721

ABSTRACT

BACKGROUND: There is evidence to support a role for angiotensin (Ang) 1-7 in reducing the activity of inflammatory signaling molecules such as MAPK, PKC and SRC. Enhanced angiotensin converting enzyme 2 (ACE2) expression has been observed in patients with inflammatory bowel disease (IBD) suggesting a role in its pathogenesis, prompting this study. METHODS: The colonic expression/activity profile of ACE2, Ang 1-7, MAS1-receptor (MAS1-R), MAPK family and Akt were determined by western blot and immunofluorescence. The effect of either exogenous administration of Ang 1-7 or pharmacological inhibition of its function (by A779 treatment) was determined using the mouse dextran sulfate sodium model. RESULTS: Enhanced colonic expression of ACE2, Ang1-7 and MAS1-R was observed post-colitis induction. Daily Ang 1-7 treatment (0.01-0.06 mg/kg) resulted in significant amelioration of DSS-induced colitis. In contrast, daily administration of A779 significantly worsened features of colitis. Colitis-associated phosphorylation of p38, ERK1/2 and Akt was reduced by Ang 1-7 treatment. CONCLUSION: Our results indicate important anti-inflammatory actions of Ang 1-7 in the pathogenesis of IBD, which may provide a future therapeutic strategy to control the disease progression.


Subject(s)
Angiotensin I/pharmacology , Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , MAP Kinase Signaling System/drug effects , Peptide Fragments/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Male , Mice , Mice, Inbred BALB C , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/metabolism
16.
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
17.
Med Princ Pract ; 25 Suppl 2: 28-40, 2016.
Article in English | MEDLINE | ID: mdl-26849149

ABSTRACT

In the majority of women, breast cancer progresses through increased transcriptional activity due to over-expressed oestrogen receptors (ER). Therapeutic strategies include: (i) reduction of circulating ovarian oestrogens or of peripherally produced oestrogen (in postmenopausal women) with aromatase inhibitors and (ii) application of selective ER modulators for receptor blockade. The success of these interventions is limited by the variable but persistent onset of acquired resistance and by an intrinsic refractiveness which manifests despite adequate levels of ER in about 50% of patients with advanced metastatic disease. Loss of functional ER leads to endocrine insensitivity, loss of cellular adhesion and polarity, and increased migratory potential due to trans-differentiation of the epithelial cancer cells into a mesenchymal-like phenotype (epithelial-mesenchymal transition; EMT). Multiple mechanisms contributing to therapeutic failure have been proposed: (i) loss or modification of ER expression including epigenetic mechanisms, (ii) agonistic actions of selective ER modulators that may be enhanced through an increased expression of co-activators, (iii) attenuation of the tamoxifen metabolism through expression of genetic variants of P450 cytochromes which leads to more or less active metabolites and (iv) increased growth factor signalling particularly through epidermal growth factor receptor activation of pathways involving keratinocyte growth factor, platelet-derived growth factor, and nuclear factor x03BA;B. In addition, the small non-coding microRNAs, recently recognized as critical gene regulators, exhibit differential expression in tamoxifen-sensitive versus resistant cell lines. Several studies suggest the potential of using these either as targets or as therapeutic agents to modulate EMT regulators as a means of reversing the aggressive metastatic phenotype by reversal of the EMT, with the added benefit of re-sensitization to anti-oestrogens.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Receptors, Estrogen , Antineoplastic Agents, Hormonal/pharmacology , Biomarkers, Tumor/blood , Breast Neoplasms/blood , Breast Neoplasms/pathology , Disease Progression , Female , Fibroblast Growth Factor 7 , Humans , MicroRNAs/blood , MicroRNAs/drug effects , MicroRNAs/metabolism , Prognosis , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
18.
Int J Oncol ; 48(1): 73-83, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26718772

ABSTRACT

Voltage-gated Na+ channels (VGSCs) are membrane proteins which are normally expressed in excitable cells but have also been detected in cancer cells, where they are thought to be involved in malignancy progression. In this study we examined the ion current and expression profile of VGSC (Nav1.5) in estrogen receptor (ER)-positive (MCF-7) and silenced (pII) breast cancer cells and its possible influence on their proliferation, motility and invasion. VGSC currents were analysed by whole cell patch clamp recording. Nav1.5 expression and localization, in response to EGF stimulation, was examined by western blotting and immunofluorescence respectively. Cell invasion (under-agarose and Matrigel assays), motility (wound healing assay) and proliferation (MTT assay) were assessed in pII cells in response to VGSC blockers, phenytoin (PHT) and tetrodotoxin (TTX), or by siRNA knockdown of Nav1.5. The effect of PHT and TTX on modulating EGF-induced phosphorylation of Akt and ERK1/2 was determined by western blotting. Total matrix metalloproteinase (MMP) was determined using a fluorometric-based activity assay. The level of various human proteases was detected by using proteome profiler array kit. VGSC currents were detected in pII cells, but were absent in MCF-7. Nav1.5 showed cytoplasmic and perinuclear expression in both MCF-7 and pII cells, with enhanced expression upon EGF stimulation. Treatment of pII cells with PHT, TTX or siRNA significantly reduced invasion towards serum components and EGF, in part through reduction of P-ERK1/2 and proteases such as cathepsin E, kallikrein-10 and MMP-7, as well as total MMP activity. At high concentrations, PHT inhibited motility while TTX reduced cell proliferation. Pharmacological or genetic blockade of Nav1.5 may serve as a potential anti-metastatic therapy for breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Receptor alpha/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Neoplasm Invasiveness/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , NAV1.5 Voltage-Gated Sodium Channel/drug effects , Patch-Clamp Techniques , Phenytoin/administration & dosage , Tetrodotoxin/administration & dosage
19.
Med Princ Pract ; 25 Suppl 2: 18-27, 2016.
Article in English | MEDLINE | ID: mdl-26488882

ABSTRACT

Cellular blebbing is a unique form of dynamic protrusion emanating from the plasma membrane which can be either apoptotic or nonapoptotic in nature. Blebs have been observed in a wide variety of cell types and in response to multiple mechanical and chemical stimuli. They have been linked to various physiological and pathological processes including tumor motility and invasion, as well as to various immunological disorders. They can form and retract extremely rapidly in seconds or minutes, or slowly over hours or days. This review focuses on recent evidence regarding the role of blebbing in cell locomotion with particular emphasis on its role in tumor metastasis, indicating the role of specific causative molecules. The phenomenon of blebbing has been observed in endocrine-resistant breast cancer cells in response to brief exposure to extracellular alkaline pH, which leads to enhanced invasive capacity. Genetic or pharmacological targeting of cellular blebs could serve as a potential therapeutic option to control tumor metastasis.


Subject(s)
Breast Neoplasms/metabolism , Cell Membrane/physiology , Animals , Apoptosis , Blister , Breast Neoplasms/pathology , Cell Membrane/drug effects , Cell Movement , Death-Associated Protein Kinases , Female , Humans , Immune System Diseases , Neoplasm Metastasis/prevention & control , Proto-Oncogenes , rhoA GTP-Binding Protein , src-Family Kinases
20.
Carcinogenesis ; 36 Suppl 1: S128-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106135

ABSTRACT

The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.


Subject(s)
Carcinogens, Environmental/adverse effects , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Animals , Disease Progression , Environmental Exposure/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Humans
SELECTION OF CITATIONS
SEARCH DETAIL