Evangelos Balafas
Academy of Athens
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Evangelos Balafas.
Journal of Proteome Research | 2009
Alexandros P. Siskos; Theodora Katsila; Evangelos Balafas; Nikolaos Kostomitsopoulos; Constantin Tamvakopoulos
The incretin hormone Glucose-dependent Insulinotropic Polypeptide GIP1-42 (approximately 5 kDa), is released postprandially, and rapidly degraded by Dipeptidyl Peptidase IV (DP-IV) to yield the inactive GIP3-42. Methods for the quantification of the pair of GIP peptides include combinations of immunoassays; however, mass spectrometry based approaches can offer the improved selectivity required for the distinction between the active and inactive forms. In this study, we report an LC/ESI-MS/MS approach for the simultaneous absolute quantification of GIP1-42 and GIP3-42 via the corresponding surrogate proteolytic peptide fragments, GIP1-16 and GIP3-16. These surrogate peptides afford approximately 250-fold improvement in lower limits of quantification (LLOQ) compared to the precursor proteins. The LLOQ of the reported method was 5 ng/mL (5-1000 ng/mL) for GIP1-42 and 10 ng/mL (10-1000 ng/mL) for GIP3-42, using 100 microL of mouse plasma. This is the first reported study in which the GIP1-42 and GIP3-42 polypeptides are quantified simultaneously with LC/ESI-MS/MS via their tryptic surrogate peptides. The approach is suitable for both preclinical and clinical pharmacokinetic studies due to the low volume required for the analysis. The described methodology was applied to a pharmacokinetic study, in which enhanced stability of exogenously administered GIP1-42 was demonstrated in mice treated with a DP-IV inhibitor.
Journal of Pharmacology and Experimental Therapeutics | 2011
Theodora Katsila; Evangelos Balafas; George Liapakis; Patrizia Limonta; Marina Montagnani Marelli; Konstantinos Gkountelias; Theodore Tselios; Nikolaos Kostomitsopoulos; John Matsoukas; Constantin Tamvakopoulos
Gonadotropin-releasing hormone (GnRH) receptor agonists have wide clinical applications including the treatment of prostate cancer and endocrine disorders. However, such agonists are characterized by poor pharmacokinetic properties, often requiring repeated administration or special formulations. Therefore, the development of novel peptide analogs with enhanced in vivo stability could potentially provide therapeutic alternatives. The pharmacological evaluation of a bioactive peptide [Des-Gly10,Tyr5(OMe),d-Leu6,Aze-NHEt9]GnRH, analog 1, is presented herein and compared with leuprolide. Peptide stability was evaluated using mouse kidney membrane preparations, followed by a liquid chromatography-tandem mass spectrometry-based approach that afforded identification and quantification of its major metabolites. The analog was significantly more stable in vitro in comparison with leuprolide. In vitro and in vivo stability results correlated well, encouraging us to develop a clinically relevant pharmacokinetic mouse model, which facilitated efficacy measurements using testosterone as a biomarker. Analog 1, an agonist of the GnRH receptor with a binding affinity in the nanomolar range, caused testosterone release in mice that was acutely dose-dependent, an effect blocked by the GnRH receptor antagonist cetrorelix. Repeated dosing studies in mice demonstrated that analog 1 was well tolerated and had potency similar to that of leuprolide, based on plasma and testis testosterone reduction and histopathological findings. Analog 1 also shared with leuprolide similar significant antiproliferative activity on androgen-dependent prostate cancer (LNCaP) cells. On the basis of pharmacokinetic advantages, we expect that analog 1 or analogs based on this new design will be therapeutically advantageous for the treatment of cancer and endocrine disorders.
Brain | 2016
Evangelia Emmanouilidou; Georgia Minakaki; Maria V. Keramioti; Mary Xylaki; Evangelos Balafas; Margarita Chrysanthou-Piterou; Ismini Kloukina; Kostas Vekrellis
α-Synuclein is readily released in human and mouse brain parenchyma, even though the normal function of the secreted protein has not been yet elucidated. Under pathological conditions, such as in Parkinsons disease, pathologically relevant species of α-synuclein have been shown to propagate between neurons in a prion-like manner, although the mechanism by which α-synuclein transfer induces degeneration remains to be identified. Due to this evidence extracellular α-synuclein is now considered a critical target to hinder disease progression in Parkinsons disease. Given the importance of extracellular α-synuclein levels, we have now investigated the molecular pathway of α-synuclein secretion in mouse brain. To this end, we have identified a novel synaptic network that regulates α-synuclein release in mouse striatum. In this brain area, the majority of α-synuclein is localized in corticostriatal glutamatergic terminals. Absence of α-synuclein from the lumen of brain-isolated synaptic vesicles suggested that they are unlikely to mediate its release. To dissect the mechanism of α-synuclein release, we have used reverse microdialysis to locally administer reagents that locally target specific cellular pathways. Using this approach, we show that α-synuclein secretion in vivo is a calcium-regulated process that depends on the activation of sulfonylurea receptor 1-sensitive ATP-regulated potassium channels. Sulfonylurea receptor 1 is distributed in the cytoplasm of GABAergic neurons from where the ATP-dependent channel regulates GABA release. Using a combination of specific agonists and antagonists, we were able to show that, in the striatum, modulation of GABA release through the sulfonylurea receptor 1-regulated ATP-dependent potassium channels located on GABAergic neurons controls α-synuclein release from the glutamatergic terminals through activation of the presynaptic GABAB receptors. Considering that sulfonylurea receptors can be selectively targeted, our study highlights the potential use of the key molecules in the α-synuclein secretory pathway to aid the discovery of novel therapeutic interventions for Parkinsons disease.
International Journal of Pharmaceutics | 2015
Eirini Christodoulou; Irene-Ariadne Kechagia; Stavros Tzimas; Evangelos Balafas; Nikolaos Kostomitsopoulos; Helen Archontaki; Aristides Dokoumetzidis; Georgia Valsami
Silibinin, the main active component of Silybum marianum is a hepatoprotective and antioxidant agent with antitumor effect, exhibiting very low aqueous solubility and oral bioavailability limiting its use in therapeutics. We characterized serum and tissue pharmacokinetics of SLB, calculated its absolute bioavailability and developed an open loop physiologically based pharmacokinetic (PBPK) model, after oral (per os, p.o) and intravenous (i.v.) administration in mice as water-soluble silibinin-hydroxypropyl-beta-cyclodextrin (SLB-HP-β-CD) lyophilized product. 60 C57Bl/6J mice were divided into groups of 5, each group representing one sampling time point. SLB-HP-β-CD lyophilized product was administered orally (50mg/kg) and i.v. (20mg/kg) after reconstitution with water for injection. Blood and tissue samples were collected at selected time points after animal sacrificed, properly treated and analyzed with HPLC-PDA for non-metabolized and total SLB. NONMEM pharmacokinetic analysis revealed a 2-compartment PK model to describe serum SLB pharmacokinetics, with zero order absorption after oral administration and was applied as forcing function to an open loop PBPK model incorporating heart, liver, kidneys and lungs. Tissue/plasma Kp values were estimated using i.v. data and can be used to predict tissue SLB distribution after oral administration. Absolute oral bioavailability of SLB from the lyophilized SLB-HP-β-CD product was 10 times higher than after administration of pure SLB.
Molecular Cancer | 2015
Eumorphia G. Konstantakou; Gerassimos E. Voutsinas; Athanassios D. Velentzas; Aggeliki-Stefania Basogianni; Efthimios Paronis; Evangelos Balafas; Nikolaos Kostomitsopoulos; Konstantinos Syrigos; Ema Anastasiadou; Dimitrios J. Stravopodis
BackgroundUrinary bladder cancer is one of the most fatal and expensive diseases of industrialized world. Despite the strenuous efforts, no seminal advances have been achieved for its clinical management. Given the importance of metabolic reprogramming in cancer cell survival and growth, we have herein employed 3-BrPA, a halogenated derivative of pyruvate and historically considered inhibitor of glycolysis, to eliminate bladder cancer cells with highly oncogenic molecular signatures.MethodsBladder cancer cells were exposed to 3-BrPA in the absence or presence of several specific inhibitors. Cell viability was determined by MTT and flow-cytometry assays; cell death, signaling activity and metabolic integrity by Western blotting and immunofluorescence; mutant-gene profiling by DNA sequencing; and gene expression by RT-sqPCR.Results3-BrPA could activate dose-dependent apoptosis (type 1 PCD) and regulated necrosis (type 3 PCD) of T24 (grade III; H-RasG12V; p53ΔY126), but not RT4 (grade I), cells, with PARP, MLKL, Drp1 and Nec-7-targeted components critically orchestrating necrotic death. However, similarly to RIPK1 and CypD, p53 presented with non-essential contribution to 3-BrPA-induced cellular collapse, while reactivation of mutant p53 with PRIMA-1 resulted in strong synergism of the two agents. Given the reduced expression of MPC components (likely imposing mitochondrial dysfunction) in T24 cells, the suppression of constitutive autophagy (required by cells carrying oncogenic Ras; also, type 2 PCD) and derangement of glucose-homeostasis determinants by 3-BrPA critically contribute to drug-directed depletion of ATP cellular stores. This bioenergetic crisis is translated to severe dysregulation of Akt/FoxO/GSK-3, mTOR/S6, AMPK and MAPK (p44/42, p38 and SAPK/JNK) signaling pathways in 3-BrPA-treated T24 cells. Sensitivity to 3-BrPA (and tolerance to glucose deprivation) does not rely on B-RafV600E or K-RasG13D mutant oncogenic proteins, but partly depends on aberrant signaling activities of Akt, MAPK and AMPK kinases. Interestingly, MCT1- and macropinocytosis-mediated influx of 3-BrPA in T24 represents the principal mechanism that regulates cellular responsiveness to the drug. Besides its capacity to affect transcription in gene-dependent manner, 3-BrPA can also induce GLUT4-specific splicing silencing in both sensitive and resistant cells, thus dictating alternative routes of drug trafficking.ConclusionsAltogether, it seems that 3-BrPA represents a promising agent for bladder cancer targeted therapy.
Hormones (Greece) | 2013
Angeliki Krikri; Vasileios Alexopoulos; Emmanouil Zoumakis; Paraskevas Katsaronis; Evangelos Balafas; Gregory Kouraklis; Panagiotis E. Karayannacos; George P. Chrousos; Gregory Skalkeas
OBJECTIVE: Minimally invasive operations, such as laparoscopic cholecystectomy and adrenalectomy, result in a more rapid recovery of normal function, less physiological disturbances and less stress to the organism than similar open operations. The purpose of this study was to determine the stress response associated with minimally invasive abdominal surgery compared to conventional small or large incision laparotomy. METHODS: We compared the responses of the stress hormones cortisol and the catecholamines adrenaline and noradrenaline to elective conventional and laparoscopic cholecystectomy and unilateral adrenalectomy in male pigs. Blood samples were taken from all animals at the same time, one day before surgery, at the beginning of the operation, every 15 minutes during surgery and on the first postoperative morning. RESULTS: Plasma adrenaline and noradrenaline concentrations were significantly lower in both cholecystectomies (p<0.05) and adrenalectomies (p<0.01) during laparoscopic than during open surgery. Plasma cortisol levels were significantly lower in laparoscopic than in open adrenalectomies both during surgery and on postoperative day one (p<0.05), while no major differences in cortisol levels were observed between laparoscopic and open cholecystectomies. Thus, the stress-related benefit of laparoscopic surgery depended on the size of the surgical incision in the conventional operation. CONCLUSION: Laparoscopic surgery was associated with less surgical stress than open surgery and this difference was accentuated as the surgical abdominal wall trauma increased.
Ultrasound in Medicine and Biology | 2014
Marianna Stasinopoulou; George Mantziaras; Euthimios Paronis; Evangelos Balafas; Pavlos Lelovas; Athina Samara; Nikolaos Kostomitsopoulos
n Abstractn n Researchers sometimes face difficulties in the diagnosis of pregnancy and assessment of embryonic development. Ultrasonography (US) is a non-invasive imaging method with minimal side effects on the subjects or operators. It provides real-time evaluation of the physiology of rapidly moving structures (i.e., heart) and facilitates evaluation of fetal tissue development. US discerns tissues based on composition, making it the imaging method of choice for abdominal examination. In this study we used real-time US as an alternative method for early diagnosis of pregnancy in rats. Sixty-four Wistar rats aged 16–20xa0wk were examined, and day 8 was the earliest point at which pregnancy could be detected. We constructed a detailed timeline of embryonic features detectable by US on days 8 to 19. We trust this index will be a valuable tool. More refined work toward a more detailed “atlas” will help to reduce animal sacrifice during embryonic development studies.n n
Frontiers in Physiology | 2017
Ioanna Andreadou; Panagiotis Efentakis; Evangelos Balafas; Gabriele Togliatto; Constantinos H. Davos; Aimilia Varela; Constantinos A. Dimitriou; Panagiota-Efstathia Nikolaou; Eirini Maratou; Vaia Lambadiari; Ignatios Ikonomidis; Nikolaos Kostomitsopoulos; Maria Felice Brizzi; George Dimitriadis; Efstathios K. Iliodromitis
Empagliflozin (EMPA), a drug approved for type 2 diabetes management, reduced cardiovascular death but is unknown if it reduces myocardial infarction. We sought to investigate: (i) the effect of EMPA on myocardial function and infarct size after ischemia/reperfusion in mice fed with western diet (WD), (ii) the underlying signaling pathways, (iii) its effects on cell survival in rat embryonic-heart-derived cardiomyoblasts (H9C2) and endothelial cells (ECs). To facilitate the aforementioned aims, mice were initially randomized in Control and EMPA groups and were subjected to 30 min ischemia and 2 h reperfusion. EMPA reduced body weight, blood glucose levels, and mean arterial pressure. Cholesterol, triglyceride, and AGEs remained unchanged. Left ventricular fractional shortening was improved (43.97 ± 0.92 vs. 40.75 ± 0.61%) and infarct size reduced (33.2 ± 0.01 vs. 17.6 ± 0.02%). In a second series of experiments, mice were subjected to the above interventions up to the 10th min of reperfusion and myocardial biopsies were obtained for assessment of the signaling cascade. STAT3 was increased in parallel with reduced levels of malondialdehyde (MDA) and reduced expression of myocardial iNOS and interleukin-6. Cell viability and ATP content were increased in H9C2 and in ECs. While, STAT3 phosphorylation is known to bestow infarct sparing properties through interaction with mitochondria, we observed that EMPA did not directly alter the mitochondrial calcium retention capacity (CRC); therefore, its effect in reducing myocardial infarction is STAT3 dependent. In conclusion, EMPA improves myocardial function and reduces infarct size as well as improves redox regulation by decreasing iNOS expression and subsequently lipid peroxidation as shown by its surrogate marker MDA. The mechanisms of action implicate the activation of STAT3 anti-oxidant and anti-inflammatory properties.
Lab Animal | 2014
Elda Koutroli; Paul Alexakos; Zacharias Kakazanis; Irene Symeon; Evangelos Balafas; Chrysa Voyiatzaki; Nikolaos Kostomitsopoulos
Embryo transfer is a surgical technique that is widely used in reproductive biotechnology. Despite the ethical obligation to relieve animals post-operative pain, analgesia is not routinely provided after embryo transfer surgery because it has been suggested that analgesics may be detrimental to embryo survival. Studies suggest, however, that the potential for adverse effects varies depending on the type of analgesic used and the timing of its administration. The authors carried out a study to determine whether pre-operatively administered tramadol, a synthetic analogue of codeine, influenced birth rate, litter survival or the post-operative body weights of surrogate dams. Compared with controls that were not given any analgesic, surrogate dams given tramadol had similar birth rates and similar body weights at all time points. The tramadol-treated surrogate dams showed a statistically significant increase in the number of offspring that survived to weaning. The authors conclude that pre-operatively administered tramadol does not harm the success rate of embryo transfer surgery and even may improve litter survival.
Brain and behavior | 2018
Adamantia F. Fragopoulou; Alexandros Polyzos; Maria-Despoina Papadopoulou; Anna Sansone; Areti K. Manta; Evangelos Balafas; Nikolaos Kostomitsopoulos; Aikaterini Skouroliakou; Chryssostomos Chatgilialoglu; Alexandros G. Georgakilas; Dimitrios J. Stravopodis; Carla Ferreri; Dimitris Thanos; Lukas H. Margaritis
The widespread use of wireless devices during the last decades is raising concerns about adverse health effects of the radiofrequency electromagnetic radiation (RF‐EMR) emitted from these devices. Recent research is focusing on unraveling the underlying mechanisms of RF‐EMR and potential cellular targets. The “omics” high‐throughput approaches are powerful tools to investigate the global effects of RF‐EMR on cellular physiology.