Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Efstathia Papageorgiou is active.

Publication


Featured researches published by Efstathia Papageorgiou.


Molecular Medicine | 2009

IGF-1 Expression in Infarcted Myocardium and MGF E Peptide Actions in Rat Cardiomyocytes in Vitro

Anastasia Stavropoulou; Antonios Halapas; Antigone Sourla; Anastassios Philippou; Efstathia Papageorgiou; Apostolos Papalois; Michael Koutsilieris

Insulinlike growth factor-1 (IGF-1) expression is implicated in myocardial pathophysiology, and two IGF-1 mRNA splice variants have been detected in rodents, IGF-1Ea and mechano-growth factor (MGF). We investigated the expression pattern of IGF-1 gene transcripts in rat myocardium from 1 h up to 8 wks after myocardial infarction induced by left anterior descending coronary artery ligation. In addition, we characterized IGF-1 and MGF E peptide action and their respective signaling in H9C2 myocardial-like cells in vitro. IGF-1Ea and MGF expression were significantly increased, both at transcriptional and translational levels, during the late postinfarction period (4 and 8 wks) in infarcted rat myocardium. Measurements of serum IGF-1 levels in infarcted rats were initially decreased (24 h up to 1 wk) but remained unaltered throughout the late experimental phase (4 to 8 wks) compared with sham-operated rats. Furthermore, specific anti-IGF-1R neutralizing antibody failed to block the synthetic MGF E peptide action, whereas it completely blocked IGF-1 action on the proliferation of H9C2 cells. Moreover, this synthetic MGF E peptide did not activate Akt phosphorylation, whereas it activated ERK1/2 in H9C2 rat myocardial cells. These data support the role of IGF-1 expression in the myocardial repair process and suggest that synthetic MGF E peptide actions may be mediated via an IGF-1R independent pathway in rat myocardial cells, as suggested by our in vitro experiments.


Expert Opinion on Therapeutic Targets | 2007

The non-genomic crosstalk between PPAR-γ ligands and ERK1/2 in cancer cell lines

Efstathia Papageorgiou; Nea Pitulis; Pavlos Msaouel; Peter Lembessis; Michael Koutsilieris

Peroxisome proliferator activated receptors (PPARs) are members of the nuclear receptor superfamily acting as transcription factors. PPAR-γ, one of the three PPAR subtypes, is expressed in many malignant and non-malignant cells and tissues. PPAR-γ ligands influence cancer biology via both genomic as well as non-genomic events. The non-genomic action of PPAR-γ ligands, including the activation of MAPK signaling pathways, is under intense investigation. In the presence of PPAR-γ ligands, a rapid phosphorylation of ERK1/2 is observed in many cancer cell lines. Activated ERK1/2 elicits rapid, non-genomic cellular effects and can directly repress PPAR-γ transcriptional activity by phosphorylation. This paper reviews the interrelation of PPAR-γ ligands and activated ERK1/2, in relation to their antineoplastic actions in cancer cell lines, which may offer the potential for improved anticancer therapies.


Molecular Medicine | 2008

Rosiglitazone attenuates insulin-like growth factor 1 receptor survival signaling in PC-3 cells.

Efstathia Papageorgiou; Nea Pitulis; Menelaos N. Manoussakis; Peter Lembessis; Michael Koutsilieris

PPARγ, a member of the peroxisome proliferator-activated receptor family, is overexpressed in prostate cancer. Natural and synthetic ligands of PPARγ via genomic and nongenomic actions promote cell cycle arrest and apoptosis of several prostate cancer cells, in vitro. Insulin-like growth factor 1 (IGF-1) inhibits the adriamycin-induced apoptosis of PC-3 human prostate cancer cells. Therefore, we have analyzed the ability of two PPARγ ligands,15dPGJ2 and rosiglitazone, a natural and a synthetic PPARγ ligand, respectively, to increase the adriamycin-induced cytotoxicity of PC-3 cells and to suppress the IGF-1 survival effect on adriamycin-induced apoptosis of PC-3 cells. Our data revealed that both the PPARγ ligands increased the adriamycin-induced cytostasis of PC-3 cells, however, only rosiglitazone added to the adriamycin-induced apoptosis of PC-3 cells. In addition, rosiglitazone attenuated the type I IGF receptor (IGF-1R) survival signaling on adriamycin-induced apoptosis of PC-3 cells via its nongenomic action on ERK1/2 and AKT phosphorylation. Because the IGF-1R signaling is probably the most important host tissue (bone) metastasis microenvironment-related survival signaling for prostate cancer cells, we conclude that rosiglitazone effects on IGF-1R-mediated activation of ERK1/2 and AKT could have clinical implications for the management of androgen ablation-refractory and chemotherapy-resistant advanced prostate cancer with bone metastasis.


Experimental Biology and Medicine | 2016

Advanced glycation end-products and insulin signaling in granulosa cells:

Evanthia Diamanti-Kandarakis; Antonios Chatzigeorgiou; Efstathia Papageorgiou; Dimitrios Koundouras; Michael Koutsilieris

Advanced glycation end-products (AGEs) may interfere with insulin intracellular signaling and glucose transport in human granulosa cells, potentially affecting ovarian function, follicular growth, linked with diminished fertility. The potential interaction of AGEs with insulin signaling pathways and glucose transport was investigated in human granulosa KGN cells. KGN cells were cultured with variable concentrations of human glycated albumin (HGA, 50–200 µg/mL) or insulin (100 ng/mL). Combined treatments of KGN cells with insulin (100 ng/mL) and HGA (200 µg/mL) were also performed. p-AKT levels and glucose transporter type 4 (Glut-4) translocation analysis were performed by Western blot. Phosphatidylinositol-3-kinase (PI3K)-specific signaling was checked by using the PI3K-inhibitor, LY294002. p-AKT levels were significantly increased following insulin treatment compared to basal levels or HGA exposure. This insulin-mediated AKT-phosphorylation was PI3K-specific and it was inhibited after combined treatment of insulin and HGA. Furthermore, Glut-4 translocation from the cytoplasm to the membrane compartments of KGN cells was remarkably reduced after the combined treatment of insulin and HGA. The present findings support that AGEs interfere with insulin signaling in granulosa cells and prevent Glut-4 membrane translocation suggesting that intra ovarian AGEs accumulation, from endogenous or exogenous sources, may contribute to the pathophysiology of states characterized with anovulation and insulin resistance such as polycystic ovary syndrome.


Molecular Medicine | 2015

Oncogenic Role of the Ec Peptide of the IGF-1Ec Isoform in Prostate Cancer

Athanasios Armakolas; Maria Kaparelou; Andreas Dimakakos; Efstathia Papageorgiou; Nikolaos Armakolas; Athanasios Antonopoulos; Constantina Petraki; Maria Lekarakou; Pavlos Lelovas; Martha Stathaki; Constantinos Psarros; Ismene Donta; Panos S Galanos; Paul Msaouel; Vassilis G. Gorgoulis; Michael Koutsilieris

IGF-1 is one of the key molecules in cancer biology; however, little is known about the role of the preferential expression of the premature IGF-1 isoforms in prostate cancer. We have examined the role of the cleaved COO− terminal peptide (PEc) of the third IGF-1 isoform, IGF-1Ec, in prostate cancer. Our evidence suggests that endogenously produced PEc induces cellular proliferation in the human prostate cancer cells (PC-3) in vitro and in vivo, by activating the ERK1/2 pathway in an autocrine/paracrine manner. PEc overexpressing cells and tumors presented evidence of epithelial to mesenchymal transition, whereas the orthotopic injection of PEc-overexpressing, normal prostate epithelium cells (HPrEC) in SCID mice was associated with increased metastatic rate. In humans, the IGF-1Ec expression was detected in prostate cancer biopsies, where its expression correlates with tumor stage. Our data describes the action of PEc in prostate cancer biology and defines its potential role in tumor growth, progression and metastasis.


Molecular Medicine | 2018

IL-6 is associated to IGF-1Ec upregulation and Ec peptide secretion, from prostate tumors

Athanasios Armakolas; Andreas Dimakakos; C. Loukogiannaki; N. Armakolas; A. Antonopoulos; C. Florou; P. Tsioli; Efstathia Papageorgiou; T. P. Alexandrou; Martha Stathaki; D. Spinos; D. Pektasides; E. Patsouris; Michael Koutsilieris

BackgroundEc peptide (PEc), resulting from the proteolytic cleavage of the IGF-1Ec isoform, is involved in prostate cancer progression and metastasis, whereas in muscle tissue, it is associated with the mobilization of satellite cells prior to repair. Our aim is to determine the physiological conditions associated to the IGF-1Ec upregulation and PEc secretion in prostate tumors, as well as, the effect of tumor PEc on tumor repair.MethodsIGF-1 (mature and isoforms) expression was examined by qRT-PCR, both in prostate cancer cells co-incubated with cells of the immune response (IR) and in tumors. PEc secretion was determined by Multiple Reaction Monitoring.The effect of PEc, on mesenchymal stem cell (MSC) mobilization and repair, was examined using migration and invasion assays, FISH and immunohistochemistry (IHC). The JAK/STAT signaling pathway leading to the IGF1-Ec expression was examined by western blot analysis. Determination of the expression and localization of IL-6 and IGF-1Ec in prostate tumors was examined by qRT-PCR and by IHC.ResultsWe documented that IL-6 secreted by IR cells activates the JAK2 and STAT3 pathway through IL-6 receptor in cancer cells, leading to the IGF-1Ec upregulation and PEc secretion, as well as to the IL-6 expression and secretion. The resulting PEc, apart from its oncogenic role, also mobilizes MSCs towards the tumor, thus promoting tumor repair.ConclusionsIL-6 leads to the PEc secretion from prostate cancer cells. Apart from its oncogenic role, PEc is also involved in the mobilization of MSCs resulting in tumor repair.


Hormones (Greece) | 2016

The human Ec peptide: the active core of a progression growth factor with species-specific mode of action.

Efstathia Papageorgiou; Anastassios Philippou; Athanasios Armakolas; Panagiotis F. Christopoulos; Andreas Dimakakos; Michael Koutsilieris

OBJECTIVE: Preferential IGF-1Ec expression has been firmly associated with skeletal muscle repair mechanisms, post-infarction remodeling of the myocardium, the pathophysiology of endometriosis and prostate cancer biology. Therefore, we have studied the possible biological significance of synthetic Ec peptide, a putative cleavage product of IGF-1Ec in PC-3 cells and C2C12 myoblasts. DESIGN: We had previously designed and synthesized commercially peptides corresponding to the human Ec and its mouse igf1 counterpart as well as synthetic peptides that correspond to parts of the hEc. Using proliferation and mitogenic signaling assays, we tested their effect on PC-3 cells and C2C12 myoblasts at different doses and in different culture conditions. RESULTS: Human Ec, hEc, was documented as exerting progression but not competence growth factor actions, activating ERK1/2 without affecting Akt phosphorylation in PC-3 cells. A narrow concentration range of hEc (5–50nM) stimulated the growth of PC-3 cells grown in culture media supplemented with 10% FBS. hEc did not stimulate the growth of PC-3 cells cultured with media containing 0.5% FBS or in mouse C2C12 myoblasts under any culture conditions. The activity of hEc was blocked by a neutralizing anti-human IGF-1Ec antibody but not by a neutralizing anti-human IGF-1 receptor antibody. The synthetic mouse Ec was inactive in human PC-3 cells; however, it stimulated significantly the proliferation of mouse C2C12. By analyzing the bioactivity of synthetic hEc fragments, we documented that hEc’s active core is located in the last 4aa of its C-terminal end. CONCLUSION: The hEc peptide is an important progression factor for human PC-3 prostate cancer cells.


Anticancer Research | 2009

The glutamatergic system expression in human PC-3 and LNCaP prostate cancer cells.

Nikolaos Pissimissis; Efstathia Papageorgiou; Peter Lembessis; Athanasios Armakolas; Michael Koutsilieris


Anticancer Research | 2009

IL-6 and PPARγ Signalling in Human PC-3 Prostate Cancer Cells

Nea Pitulis; Efstathia Papageorgiou; Roxane Tenta; Peter Lembessis; Michael Koutsilieris


Experimental Biology and Medicine | 2018

Advanced glycation end products interfere in luteinizing hormone and follicle stimulating hormone signaling in human granulosa KGN cells

Eleni Kandaraki; Antonios Chatzigeorgiou; Efstathia Papageorgiou; Christina Piperi; Christos Adamopoulos; Athanasios G. Papavassiliou; Michael Koutsilieris; Evanthia Diamanti-Kandarakis

Collaboration


Dive into the Efstathia Papageorgiou's collaboration.

Top Co-Authors

Avatar

Michael Koutsilieris

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Athanasios Armakolas

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Peter Lembessis

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Andreas Dimakakos

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Evanthia Diamanti-Kandarakis

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Nea Pitulis

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Anastassios Philippou

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Christina Piperi

National and Kapodistrian University of Athens

View shared research outputs
Top Co-Authors

Avatar

Eleni Kandaraki

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Martha Stathaki

National and Kapodistrian University of Athens

View shared research outputs
Researchain Logo
Decentralizing Knowledge