Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
PLoS One ; 17(7): e0272292, 2022.
Article in English | MEDLINE | ID: mdl-35905079

ABSTRACT

Research and innovation play a key role in generating smart and sustainable economic growth. By producing new knowledge, the research contributes to the development of new and innovative products, processes, and services, which in turn lead to increased productivity, industrial competitiveness, and, ultimately, the prosperity of the community as a whole. However, all research, development and innovation activities depend on the financial resources made available, as specific financing accelerates the production and dissemination of the best ideas and practices, as well as their role in meeting the challenges our society deals with nowadays. Our study aims to identify the determining factors for the researcher's participation and success rates in research funding competitions. The goal of the research is to understand how variables such as age, gender, main field, affiliation, and scientific rank can affect the access to funding opportunities available for research and innovation. The study relies on a questionnaire-based survey conducted with 243 early-career and senior researchers from many state universities across Romania. For an in-depth analysis of the factors that influence the success rate in research competitions, in the present approach, we used both graphical and econometric methods. A binary logistic regression modelling was performed in order to explain the relationships between variables. Among other considerations, our findings revealed that in all main research fields, scientific rank and gender are important features for raising the participation and success rate in research funding competitions.


Subject(s)
Efficiency , Research Personnel , Humans , Occupations , Romania
2.
Spectrochim Acta A Mol Biomol Spectrosc ; 190: 208-214, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-28926772

ABSTRACT

The interaction between chlorin e6 (Ce6) and human serum albumin (HSA) in the presence and absence of ultrasound have been investigated by ultraviolet-visible absorption spectroscopy and fluorescence spectroscopy. Ce6 is found to bind strongly to HSA at or near physiological pH conditions, but the strength of the binding is significantly weakened at lower pHs. The intrinsic fluorescence of HSA is incrementally quenched with increasing concentration of Ce6, and the quenching is enhanced after exposure to high-frequency ultrasound. Our experimental results suggest that Ce6-induced sonodynamic oxidation of HSA is mainly mediated by singlet oxygen. The formulation of Ce6 by high molecular weight polyvinylpyrrolidone (PVP) increased its stability in aqueous solutions and its quantum yield of singlet oxygen under ultrasound irradiation.


Subject(s)
Drug Compounding , Porphyrins/metabolism , Serum Albumin, Human/metabolism , Singlet Oxygen/chemistry , Ultrasonics , Humans , Hydrogen-Ion Concentration , Povidone/chemistry , Sodium Azide/chemistry , Spectrometry, Fluorescence
3.
Rom J Morphol Embryol ; 56(2 Suppl): 691-6, 2015.
Article in English | MEDLINE | ID: mdl-26429160

ABSTRACT

Iron oxide nanoparticles are promising candidates for theranostics in cancer, that aims to achieve in one-step precise tumor imaging by magnetic resonance, and targeted therapy through surface attached anti-cancer drugs. The aim of this study was to investigate in preclinical setting the biocompatibility of new iron oxide-based nanoparticles that were coated with L-DOPA for improved dispersion in biological media. These nanostructures (NPs) were designed for biomedical applications as contrast agents and/or drug carriers. We investigated the effect exerted in vitro by NPs and L-DOPA on the viability and proliferation of normal mouse L929 fibroblasts. NPs exhibited good biocompatibility against these cells. Moreover, L-DOPA contained in NPs sustained fibroblasts proliferation and/or limited anti-proliferative effects of naked nanoparticles. In the animal study, C57BL/6 mice were injected intraperitoneally with a single dose of NPs (approximately 125 mg/kg body weight). We followed up hematological and histological parameters for one, three and seven days after NPs administration. Results indicated that NPs possibly induced local inflammation and consequent recruitment of peripheral lymphocytes, whilst the decrease of platelet counts may reflect tissue lesions caused by NPs. The histopathological study showed mild to moderate alterations in the hepatocytes, splenic and renal cells, while the brain parenchyma only presented nonspecific congestive changes. Taken altogether, the preclinical study indicated that the new iron oxide nanoparticles coated with L-DOPA were biocompatible against fibroblasts and had a convenient toxicological profile when administered intraperitoneally in a single dose to C57BL/6 mice. Accordingly, the proposed nanostructure is a promising candidate for imaging and treating dispersed peritoneal tumors.


Subject(s)
Ferric Compounds/chemistry , Infusions, Parenteral/methods , Levodopa/chemistry , Metal Nanoparticles/chemistry , Nanomedicine/methods , Animals , Biocompatible Materials/chemistry , Cell Line , Cell Proliferation , Cell Survival , Coated Materials, Biocompatible , Contrast Media/chemistry , Drug Carriers/chemistry , Fibroblasts/metabolism , Hepatocytes/cytology , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nanostructures/chemistry , Spleen/metabolism
4.
Toxins (Basel) ; 7(2): 593-603, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25690693

ABSTRACT

Contamination of crops used for food and feed production with Fusarium mycotoxins, such as deoxynivalenol (DON), raise important health and economic issues all along the food chain. Acute exposure to high DON concentrations can alter the intestinal barrier, while chronic exposure to lower doses may exert more subtle effects on signal transduction pathways, leading to disturbances in cellular homeostasis. Using real-time cellular impedance measurements, we studied the effects exerted in vitro by low concentrations of DON (0.37-1.50 µM), relevant for mycotoxin-contaminated food, on the proliferation of undifferentiated Caco-2 cells presenting a tumorigenic phenotype. A 1.5 µM concentration of DON maintained cell adherence of non-proliferating Caco-2 cells, whilst arresting the growth of actively proliferating cells compared with control Caco-2 cells in vitro. At 0.37 µM, DON enhanced Caco-2 cell metabolism, thereby triggering a moderate increase in cell proliferation. The results of the current study suggested that low concentrations of DON commonly detected in food may either limit or sustain the proliferation of colon cancer cells, depending on their proliferation status and on DON concentration. Soluble factors released by Lactobacillus strains can partially counteract the inhibitory action of DON on actively proliferating colon cancer cells. The study also emphasized that real-time cellular impedance measurements were a valuable tool for investigating the dynamics of cellular responses to xenobiotics.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Environmental Pollutants/toxicity , Trichothecenes/toxicity , Caco-2 Cells , Cell Culture Techniques , Culture Media, Conditioned , Dose-Response Relationship, Drug , Electric Impedance , Humans , Inactivation, Metabolic , Lactobacillus/growth & development
5.
Adv Exp Med Biol ; 806: 525-44, 2014.
Article in English | MEDLINE | ID: mdl-24952201

ABSTRACT

Mass spectrometry (MS) has been increasingly used to study central nervous system disorders, including autism spectrum disorders (ASDs). The first studies of ASD using MS focused on the identification of external toxins, but current research is more directed at understanding endogenous protein changes that occur in ASD (ASD proteomics). This chapter focuses on how MS has been used to study ASDs, with particular focus on proteomic analysis. Other neurodevelopmental disorders have been investigated using this technique, including genetic syndromes associated with autism such as fragile X syndrome and Smith-Lemli-Opitz syndrome.


Subject(s)
Autistic Disorder/metabolism , Fragile X Syndrome/metabolism , Mass Spectrometry/methods , Proteomics/methods , Smith-Lemli-Opitz Syndrome/metabolism , Animals , Humans
6.
Cardiovasc Diabetol ; 12: 154, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24148218

ABSTRACT

BACKGROUND: The dipeptidyl peptidase-4 (DPP-4) inhibitors Sitagliptin and Vildagliptin lower blood glucose by augmenting endogenous levels of glucagon-like peptide-1 (GLP-1), an incretin which also confers cardioprotection. As such, we hypothesized that treatment with DPP-4 inhibitors are also cardioprotective. METHODS: In ex vivo experiments: Male Sprague-Dawley rats were randomized to receive by oral gavage either Vildagliptin (20 mg/kg/day), Sitagliptin (100 mg/kg/day), or water for 2 weeks. Excised hearts were Langendorff-perfused with buffer containing either 5 mmol/L or 11 mmol/L glucose and subjected to 35 minutes ischaemia/120 minutes reperfusion. In in vivo experiments: Male young Wistar and Sprague-Dawley rats, middle aged Wistar and Goto-Kakizaki diabetic rats were randomized to receive by oral gavage either Sitagliptin (100 mg/kg/day), or water for 2 weeks. Rats were then subjected to 30 minutes ischaemia/120 minutes reperfusion and infarct size ascertained. RESULTS: Two weeks pre-treatment with either Vildagliptin or Sitagliptin reduced ex vivo myocardial infarction (MI) size in hearts perfused with buffer containing 11 mmol/L glucose but not 5 mmol/L glucose. This effect was abolished by Exendin 9-39 (GLP-1 receptor antagonist) and H-89 (PKA antagonist). Treatment of perfused hearts with native GLP-1 was also glucose-sensitive, reducing MI size, at glucose concentrations 7, 9, and 11 mmol/L but not at 5 mmol/L. Finally, Sitagliptin reduced in vivo MI size in middle aged Wistar (7-8 mmol/L glucose) and Goto-Kakizaki (9-10 mmol/L glucose) rats where blood glucose was elevated, but not in young Wistar (5 mmol/L glucose) or Sprague-Dawley (5 mmol/L glucose) rats, where blood glucose was normal. CONCLUSIONS: We find that chronic treatment with DPP-4 inhibitors reduced MI size, via the GLP-1 receptor-PKA pathway, in a glucose-dependent manner. Glucose-sensitive cardioprotection of endogenous GLP-1 in diabetic patients may in part explain why intensive control of serum glucose levels has been associated with increased cardiovascular risk.


Subject(s)
Adamantane/analogs & derivatives , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1/drug effects , Glucose/metabolism , Heart/drug effects , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Nitriles/pharmacology , Pyrazines/pharmacology , Pyrrolidines/pharmacology , Triazoles/pharmacology , Adamantane/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide-1 Receptor , Male , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Glucagon/antagonists & inhibitors , Severity of Illness Index , Signal Transduction , Sitagliptin Phosphate , Vildagliptin
7.
PLoS Biol ; 11(9): e1001666, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24086110

ABSTRACT

Cardiomyocytes are vulnerable to hypoxia in the adult, but adapted to hypoxia in utero. Current understanding of endogenous cardiac oxygen sensing pathways is limited. Myocardial oxygen consumption is determined by regulation of energy metabolism, which shifts from glycolysis to lipid oxidation soon after birth, and is reversed in failing adult hearts, accompanying re-expression of several "fetal" genes whose role in disease phenotypes remains unknown. Here we show that hypoxia-controlled expression of the transcription factor Hand1 determines oxygen consumption by inhibition of lipid metabolism in the fetal and adult cardiomyocyte, leading to downregulation of mitochondrial energy generation. Hand1 is under direct transcriptional control by HIF1α. Transgenic mice prolonging cardiac Hand1 expression die immediately following birth, failing to activate the neonatal lipid metabolising gene expression programme. Deletion of Hand1 in embryonic cardiomyocytes results in premature expression of these genes. Using metabolic flux analysis, we show that Hand1 expression controls cardiomyocyte oxygen consumption by direct transcriptional repression of lipid metabolising genes. This leads, in turn, to increased production of lactate from glucose, decreased lipid oxidation, reduced inner mitochondrial membrane potential, and mitochondrial ATP generation. We found that this pathway is active in adult cardiomyocytes. Up-regulation of Hand1 is protective in a mouse model of myocardial ischaemia. We propose that Hand1 is part of a novel regulatory pathway linking cardiac oxygen levels with oxygen consumption. Understanding hypoxia adaptation in the fetal heart may allow development of strategies to protect cardiomyocytes vulnerable to ischaemia, for example during cardiac ischaemia or surgery.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Energy Metabolism/genetics , Lipid Metabolism/genetics , Myocardium/metabolism , Oxygen Consumption/genetics , Adenosine Triphosphate/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia/genetics , Cell Line , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Transgenic , Mitochondria/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Transcriptional Activation
8.
Cardiovasc Res ; 99(4): 694-704, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23723063

ABSTRACT

AIMS: Old age and diabetes are risk factors that often coexist increasing the vulnerability of the heart to the lethal effects of ischaemia-reperfusion injury (IRI). However, to our knowledge, no investigations have examined IRI and cardioprotective signalling in animal models bearing these co-morbidities concomitantly. The ability of the heart to recover following IRI is greatly dependent on its innate cardioprotective potential, in which a central role is played by Akt. We aimed to investigate in an aging diabetic rat model, the susceptibility of the heart to IRI, the achievability of ischaemic preconditioning (IPC) against this lethal event, and the changes in Akt signalling, as the main prosurvival intracellular pathway. METHODS AND RESULTS: Our data showed that the isolated hearts of aged, diabetic Goto-Kakizaki rats were more susceptible to sub-lethal injury and not amenable to cardioprotection via IPC, compared with younger diabetic rat hearts. Western blot analysis of the heart tissue suggested a chronic up-regulation of Akt phosphorylation, and reduced expression of the mitochondrial regulator PGC-1α and of the anti-oxidant enzyme catalase, potentially due to the Akt up-regulation. Moreover, no further activation of Akt could be achieved following IPC. CONCLUSION: An increased susceptibility to IRI in the aged, diabetic heart could be a consequence of impaired Akt signalling due to chronic Akt phosphorylation. Additional Akt phosphorylation required for IPC protection may therefore not be possible in the aged, diabetic rat heart and may explain why this cardioprotective manoeuvre cannot be achieved in these hearts.


Subject(s)
Aging/physiology , Diabetes Mellitus/physiopathology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/etiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Animals , Blood Glucose/analysis , Glycated Hemoglobin/analysis , Humans , Male , Myocardial Infarction/etiology , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Phosphorylation , Rats , Rats, Wistar , Transcription Factors/physiology
9.
PLoS One ; 8(4): e62400, 2013.
Article in English | MEDLINE | ID: mdl-23638067

ABSTRACT

OBJECTIVES: Mutations in PTEN inducible kinase-1 (PINK1) induce mitochondrial dysfunction in dopaminergic neurons resulting in an inherited form of Parkinson's disease. Although PINK1 is present in the heart its exact role there is unclear. We hypothesized that PINK1 protects the heart against acute ischemia reperfusion injury (IRI) by preventing mitochondrial dysfunction. METHODS AND RESULTS: Over-expressing PINK1 in HL-1 cardiac cells reduced cell death following simulated IRI (29.2±5.2% PINK1 versus 49.0±2.4% control; N = 320 cells/group P<0.05), and delayed the onset of mitochondrial permeability transition pore (MPTP) opening (by 1.3 fold; P<0.05). Hearts excised from PINK1+/+, PINK1+/- and PINK1-/- mice were subjected to 35 minutes regional ischemia followed by 30 minutes reperfusion. Interestingly, myocardial infarct size was increased in PINK1-/- hearts compared to PINK1+/+ hearts with an intermediate infarct size in PINK1+/- hearts (25.1±2.0% PINK1+/+, 38.9±3.4% PINK1+/- versus 51.5±4.3% PINK1-/- hearts; N>5 animals/group; P<0.05). Cardiomyocytes isolated from PINK1-/- hearts had a lower resting mitochondrial membrane potential, had inhibited mitochondrial respiration, generated more oxidative stress during simulated IRI, and underwent rigor contracture more rapidly in response to an uncoupler when compared to PINK1+/+ cells suggesting mitochondrial dysfunction in hearts deficient in PINK1. CONCLUSIONS: We show that the loss of PINK1 increases the heart's vulnerability to ischemia-reperfusion injury. This may be due, in part, to increased mitochondrial dysfunction. These findings implicate PINK1 as a novel target for cardioprotection.


Subject(s)
Myocardium/metabolism , Protein Kinases/deficiency , Reperfusion Injury/enzymology , Animals , Cell Line , Disease Susceptibility , Gene Knockout Techniques , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress , Oxygen/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
10.
Physiol Genomics ; 45(7): 243-55, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23386204

ABSTRACT

We have previously identified inbred rat strains differing in survival time to a severe controlled hemorrhage (StaH). In efforts to identify cellular mechanisms and ultimately genes that are important contributors to enhanced STaH, we conducted a study to characterize potential differences in cardiac mitochondrial proteins in these rats. Inbred rats from three strains [Brown Norway/Medical College of Wisconsin (BN); Dark Agouti (DA), and Fawn Hooded Hypertensive (FHH)] with different StaH (DA = FHH > BN) were assigned to one of three treatment groups (n = 4/strain): nonoperated controls, surgically catheterized rats, or rats surgically catheterized and hemorrhaged 24 h postsurgery. Rats were euthanized 30 min after handling or 30 min after initiation of a 26 min hemorrhage. After euthanasia, hearts were removed and mitochondria isolated. Differential protein expression was determined using 2D DIGE-based Quantitative Intact Proteomics and proteins identified by MALDI/TOF mass spectrometry. Hundreds of proteins (791) differed among inbred rat strains (P ≤ 0.038), and of these 81 were identified. Thirty-eight were unique proteins and 43 were apparent isoforms. For DA rats (longest STaH), 36 proteins increased and 30 decreased compared with BN (shortest STaH). These 81 proteins were associated with lipid (e.g., acyl CoA dehydrogenase) and carbohydrate (e.g., fumarase) metabolism, oxidative phosphorylation (e.g., ubiquinol-cytochrome C reductase), ATP synthesis (F1 ATPase), and H2S synthesis (3-mercaptopyruvate sulfurtransferase). Although we cannot make associations between these identified mitochondrial proteins and StaH, our data do provide evidence for future candidate proteins with which to consider such associations.


Subject(s)
Hemorrhage/metabolism , Mitochondria, Heart/metabolism , Proteome/analysis , Animals , Male , Mitochondria, Heart/chemistry , Mitochondrial Proteins/analysis , Mitochondrial Proteins/metabolism , Proteome/metabolism , Proteomics , Rats , Rats, Inbred BN , Rats, Inbred Strains , Time Factors , Two-Dimensional Difference Gel Electrophoresis
11.
J Cardiovasc Pharmacol Ther ; 18(3): 263-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23263382

ABSTRACT

AIMS: The diabetic heart is resistant to the myocardial infarct-limiting effects of ischemic preconditioning (IPC). This may be in part due to the downregulation of the phosphatidylinositol 3'-kinase-Akt pathway, an essential component of IPC protection. We hypothesized that treating the diabetic heart with the sulfonylurea, glimepiride, which has been reported to activate Akt, may lower the threshold required to protect the diabetic heart by IPC. METHODS: Goto-Kakizaki rats (a type II lean model of diabetes) received glimepiride (20 mg/kg per d, by oral gavage) or vehicle for (a) 3 months (chronic treatment) or (b) 24 hours (subacute treatment). In the third group, glimepiride (10 µmol/L) was administered only to the isolated hearts on the Langendorff apparatus (acute treatment). All hearts were subjected to 35 minutes ischemia and 120 minutes reperfusion ex vivo, at the end of which infarct size was determined by tetrazolium staining. Preconditioning treatment comprised 1 (IPC-1) or 3 (IPC-3) cycles of 5 minutes global ischemia and 10 minutes reperfusion. RESULTS: The diabetic heart was found to be resistant to IPC such that 3-IPC cycles, instead of the usual 1-IPC cycle, were required for cardioprotection. However, pretreatment with glimepiride lowered the threshold for IPC such that both 1 and 3 cycles of IPC elicited cardioprotection: chronic glimepiride treatment (IPC-1 31.9% ± 3.8% and IPC-3 33.5% ± 2.4% vs 43.9% ± 1.4% control, P < .05; N > 6 per group); subacute glimepiride treatment (IPC-1 31.1% ± 3.0% and IPC-3 29.3% ± 3.3% vs 42.2% ± 2.3% control, P < .05 N > 6 per group); and acute glimepiride treatment (IPC-1 28.2% ± 3.7% and IPC-3 24.6% ± 5.4% vs 41.9% ± 5.4% control, P < .05; N > 6 per group). This effect of glimepiride was independent of changes in blood glucose. CONCLUSIONS: We report for the first time that glimepiride treatment facilitates the cardioprotective effect elicited by IPC in the diabetic heart.


Subject(s)
Cardiotonic Agents/therapeutic use , Diabetic Cardiomyopathies/drug therapy , Hypoglycemic Agents/therapeutic use , Ischemic Preconditioning, Myocardial , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Sulfonylurea Compounds/therapeutic use , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Combined Modality Therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/therapy , Enzyme Activators/administration & dosage , Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Glycated Hemoglobin/analysis , Heart/drug effects , Hyperglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/enzymology , Myocardium/metabolism , Myocardium/pathology , Perfusion , Proto-Oncogene Proteins c-akt/agonists , Random Allocation , Rats , Rats, Inbred Strains , Sulfonylurea Compounds/administration & dosage , Sulfonylurea Compounds/pharmacology
12.
Cardiovasc Drugs Ther ; 27(1): 5-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23192487

ABSTRACT

PURPOSE: Clinical and experimental investigations demonstrated that metformin, a widely used anti-diabetic drug, exhibits cardioprotective properties against myocardial infarction. Interestingly, metformin was previously shown to increase the expression of PGC-1α a key controller of energy metabolism in skeletal muscle, which is down-regulated in diabetic conditions. We hypothesized that chronic treatment with metformin could protect the aged, diabetic heart against ischemia-reperfusion injury (IRI) by up-regulating PGC-1α and improving the impaired functionality of diabetic mitochondria. METHODS: Following 4 weeks of metformin (300 mg/kg) administered in the drinking water, 12 month-old diabetic Goto Kakizaki and non-diabetic Wistar rat hearts were assigned for infarct measurement following 35 min ischemia and 60 min reperfusion or for electron microscopy (EM) and Western blotting (WB) investigations. RESULTS: Metformin elicited a cardioprotective effect in both non-diabetic and diabetic hearts. In contrast with the diabetic non-treated hearts, the diabetic hearts treated with metformin showed more organized and elongated mitochondria and demonstrated a significant increase in phosphorylated AMPK and in PGC-1α expression. CONCLUSIONS: In summary these results show for the first time that chronic metformin treatment augments myocardial resistance to ischemia-reperfusion injury, by an alternative mechanism in addition to the lowering of blood glucose. This consisted of a positive effect on mitochondrial structure possibly via a pathway involving AMPK activation and PGC-1α. Thus, metformin prescribed chronically to patients may lead to a basal state of cardioprotection thereby potentially limiting the occurrence of myocardial damage by cardiovascular events.


Subject(s)
Blood Glucose/metabolism , Cardiotonic Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Myocardial Infarction/drug therapy , AMP-Activated Protein Kinase Kinases , Aging/blood , Aging/metabolism , Aging/pathology , Animals , Blotting, Western , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Male , Metformin/administration & dosage , Metformin/pharmacology , Microscopy, Electron , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Myocardial Infarction/enzymology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/enzymology , Myocardium/metabolism , Myocardium/ultrastructure , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinases/metabolism , RNA-Binding Proteins/biosynthesis , Rats , Rats, Wistar , Transcription Factors/biosynthesis
13.
Rev Med Chir Soc Med Nat Iasi ; 117(3): 605-9, 2013.
Article in English | MEDLINE | ID: mdl-24502023

ABSTRACT

UNLABELLED: The AIM of our study was to evaluate gastric, duodenal and gallbladder motility disorders in patients with gastroesophageal reflux disease (GERD) and metabolic syndrome. MATERIAL AND METHODS: We studied 128 patients with GERD divided into two groups: first group with metabolic syndrome and the second without metabolic syndrome. By abdominal ultrasound we monitored our patients for the gastric emptying rate, the duodenal motility and the nonlithiasic pathology of gallbladder (cholesterolosis). RESULTS: We found that patients with metabolic syndrome had three kind of abnormal motility disorders including stomach, duodenum, and gallbladder. The patients without metabolic syndrome we found only two abnormal motility disorders: of the duodenum and gallbladder. Hyperglycemia and high serum cholesterol level in the first group were correlated with stomach, duodenum and gallbladder abnormal motilities. In our opinion metabolic syndrome can aggravate gastroesophageal reflux disease due to these metabolic abnormalities. CONCLUSIONS: We consider that treatment of reflux disease in these particular cases must also involve measures to correct metabolic disorders.


Subject(s)
Biomarkers/blood , Gastric Emptying , Gastroesophageal Reflux/blood , Gastroesophageal Reflux/diagnostic imaging , Metabolic Syndrome/blood , Metabolic Syndrome/diagnostic imaging , Adult , Blood Glucose/metabolism , Body Mass Index , Case-Control Studies , Cholesterol/blood , Cholesterol, HDL/blood , Duodenum/diagnostic imaging , Female , Gallbladder/diagnostic imaging , Gastroesophageal Reflux/complications , Humans , Male , Metabolic Syndrome/complications , Triglycerides/blood , Ultrasonography
14.
Int J Mol Sci ; 13(12): 16718-36, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23222638

ABSTRACT

UVA affects epidermal cell physiology in a complex manner, but the harmful effects have been studied mainly in terms of DNA damage, mutagenesis and carcinogenesis. We investigated UVA effects on membrane integrity and antioxidant defense of dysplastic keratinocytes after one and two hours of irradiation, both immediately after exposure, and 24 h post-irradiation. To determine the UVA oxidative stress on cell membrane, lipid peroxidation was correlated with changes in fatty acid levels. Membrane permeability and integrity were assessed by propidium iodide staining and lactate dehydrogenase release. The effects on keratinocyte antioxidant protection were investigated in terms of catalase activity and expression. Lipid peroxidation increased in an exposure time-dependent manner. UVA exposure decreased the level of polyunsaturated fatty acids, which gradually returned to its initial value. Lactate dehydrogenase release showed a dramatic loss in membrane integrity after 2 h minimum of exposure. The cell ability to restore membrane permeability was noted at 24 h post-irradiation (for one hour exposure). Catalase activity decreased in an exposure time-dependent manner. UVA-irradiated dysplastic keratinocytes developed mechanisms leading to cell protection and survival, following a non-lethal exposure. The surviving cells gained an increased resistance to apoptosis, suggesting that their pre-malignant status harbors an abnormal ability to control their fate.


Subject(s)
Antioxidants/metabolism , Cell Membrane Permeability/radiation effects , Keratinocytes/metabolism , Lipid Peroxidation/radiation effects , Oxidative Stress/radiation effects , Ultraviolet Rays/adverse effects , Catalase/biosynthesis , Cell Line , Cell Membrane/metabolism , Cell Survival/radiation effects , Gene Expression Regulation, Enzymologic/radiation effects , Humans , Keratinocytes/pathology
15.
Electrophoresis ; 33(24): 3745-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23161654

ABSTRACT

Apolipoprotein E (ApoE) is a major lipid carrier protein. In humans, ApoE is expressed in three polymorphic isoforms, which are encoded by three different alleles APOE2, APOE3, and APOE4. In the brains of Alzheimer's disease (AD) patients, each one of these three allelic isoforms is found in several "isoelectric" protein isoforms (qPI), i.e. protein isoforms resulting from PTMs altering the net charge (q) of the polypeptide. AD is a complex disease in which multiple causes and several risk factors affect the onset and disease outcome. A major risk factor for AD is ApoE4; therefore, it is important to characterize the different ApoE qPIs. We have implemented a detergent-based method for isolation and quantitation of protein isoforms, and we found differences in the solubility of protein isoforms depending on the type of solvent used. In this manuscript, we describe these methods and applied them to young human-ApoE targeted replacement mice. Our results indicate that there are no significant differences in the hippocampus proteome of these mice as a function of the APOE genotype.


Subject(s)
Apolipoprotein E3/biosynthesis , Apolipoprotein E4/biosynthesis , Proteome/analysis , Analysis of Variance , Animals , Apolipoprotein E3/analysis , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/analysis , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Creatine Kinase/analysis , Creatine Kinase/chemistry , Creatine Kinase/metabolism , Electrophoresis, Gel, Two-Dimensional , Genotype , Hippocampus/chemistry , Hippocampus/metabolism , Humans , Mice , Mice, Transgenic , Phosphopyruvate Hydratase/analysis , Phosphopyruvate Hydratase/chemistry , Phosphopyruvate Hydratase/metabolism , Protein Isoforms , Proteome/chemistry , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Solubility
16.
Eur J Med Chem ; 53: 380-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22516425

ABSTRACT

Six new Cu(II), Ni(II), and VO(II) complexes (1-6) with Schiff base 1-phenyl-2,3-dimethyl-4-(1H-indole-3-carboxaldehyde)-3-pyrazolin-5-one (HL) were synthesized. The Schiff base was prepared through the condensation of 1-phenyl-2,3-dimethyl-4-amino-3-pyrazolin-5-one (antipyrine) with 1H-indole-3-carboxaldehyde. The new obtained compounds were characterized by (1)H NMR, (13)C NMR, UV-VIS, IR, EPR spectroscopy, elemental analysis, molar electric conductibility, magnetic susceptibility and thermal gravimetric analysis. In addition, the structure of the ligand HL has been determined by X-ray diffraction methods. The biological activity of complex compounds was investigated in terms of antibacterial effect on prokaryotic cells, by using paper disc diffusion technique, and for antiproliferative effect on eukaryotic cells, by monitoring mitotic activity in timelapse videomicroscopy experiments. The compounds were screened for their antibacterial activity against gram-positive bacteria (Staphylococcus aureus var. Oxford 6538, Klebsielle pneumoniae ATCC 100131 and Legionella monocytogenes ATCC 35182), gram-negative bacteria (Escherichia coli ATCC 10536, Pseudomonas aeruginosa ATCC 9027 and Salmonella typhimurium ATCC 14028) and anti-fungal activity (Candida albicans and Aspergillus flavus) using paper disc diffusion technique. The minimum inhibitory concentrations (MICs) of the compounds were also determined by agar streak dilution method. Compounds 3 and 4 proved to be the most effective as antibacterial agents. The antiproliferative activity was investigated by counting the number of mitoses for HeLa, and MCF7 cells. No significant antiproliferative effect was noted for HL and complex 2, for both used cell types. For complexes 1 and 3 complete inhibition of cell proliferation was observed in the case of HeLa cells, while the effects on MCF7 cell proliferation were lower. In conclusion, six new complex compounds were synthesized, and their biological activity investigated on both prokaryotic and eukaryotic cells, proving that some of them could be putative therapeutic substances.


Subject(s)
Bacteria/drug effects , Fungi/drug effects , Indoles/chemistry , Metals, Heavy/chemistry , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Copper/chemistry , HeLa Cells , Humans , Nickel/chemistry , Organometallic Compounds/chemistry , Schiff Bases/chemistry , Vanadium/chemistry
17.
Cardiol Res Pract ; 2012: 845698, 2012.
Article in English | MEDLINE | ID: mdl-22462028

ABSTRACT

Diabetes mellitus is a major risk factor for ischemic heart disease (IHD). Patients with diabetes and IHD experience worse clinical outcomes, suggesting that the diabetic heart may be more susceptible to ischemia-reperfusion injury (IRI). In contrast, the animal data suggests that the diabetic heart may be either more, equally, or even less susceptible to IRI. The conflicting animal data may be due to the choice of diabetic and/or IRI animal model. Ischemic conditioning, a phenomenon in which the heart is protected against IRI by one or more brief nonlethal periods of ischemia and reperfusion, may provide a novel cardioprotective strategy for the diabetic heart. Whether the diabetic heart is amenable to ischemic conditioning remains to be determined using relevant animal models of IRI and/or diabetes. In this paper, we review the limitations of the current experimental models used to investigate IRI and cardioprotection in the diabetic heart.

18.
Methods Mol Biol ; 854: 47-66, 2012.
Article in English | MEDLINE | ID: mdl-22311753

ABSTRACT

Difference gel electrophoresis (DIGE) is a common technique for characterizing differential protein expression in quantitative proteomics. Usually a combination of enzymatic digestion and peptide analysis by mass spectrometry is used to identify differentially expressed proteins following separation and statistical analysis by DIGE. In this chapter, methods for gel spot picking, enzymatic digestion, and matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS) for protein identification of DIGE-analyzed proteins are discussed. Two examples are given: first, a specific protein is used to test the sensitivity of the 2D DIGE/MALDI MS combination for protein quantification and identification, and second, several proteins with and without the labels typically used in DIGE are identified to demonstrate that these labels do not alter MS-based protein identification. Technical variations of protein gel spot preparation, in-gel digestion, and mass spectral protein identification are discussed.


Subject(s)
Proteins/analysis , Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Two-Dimensional Difference Gel Electrophoresis/methods , Humans , Peptide Fragments/analysis , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Proteins/isolation & purification , Proteins/metabolism , Proteomics , Trypsin/metabolism
19.
Methods Mol Biol ; 854: 129-43, 2012.
Article in English | MEDLINE | ID: mdl-22311757

ABSTRACT

Difference gel electrophoresis (DIGE) is most often used to assess relative changes in the expression levels of individual proteins in multiple complex samples, and this information is valuable in making inferences about relative protein activity. However, a protein's activity is not solely dependent upon its expression level. A change in activity may also be influenced by myriad posttranslational modifications (PTMs), including palmitoylation, ubiquitination, oxidation, and phosphorylation. In this chapter, we describe the use of DIGE to determine specific PTMs by introducing specific labels or changes in pI and/or molecular weight.


Subject(s)
Protein Processing, Post-Translational , Proteins/metabolism , Proteomics/methods , Two-Dimensional Difference Gel Electrophoresis/methods , Analytic Sample Preparation Methods , Isoelectric Focusing , Lipoylation , Molecular Weight , Oxidation-Reduction , Phosphorylation , Proteins/chemistry , Proteins/isolation & purification , Staining and Labeling , Substrate Specificity , Ubiquitination
20.
J Cell Mol Med ; 16(8): 1739-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22117619

ABSTRACT

Phosphatidyl-inositol-3-kinase (PI3K)-Akt pathway is essential for conferring cardioprotection in response to ischaemic preconditioning (IPC) stimulus. However, the role of the individual Akt isoforms expressed in the heart in mediating the protective response to IPC is unknown. In this study, we investigated the specific contribution of Akt1 and Akt2 in cardioprotection against ischaemia-reperfusion (I-R) injury. Mice deficient in Akt1 or Akt2 were subjected to in vivo regional myocardial ischaemia for 30 min. followed by reperfusion for 2 hrs with or without a prior IPC stimulus. Our results show that mice deficient in Akt1 were resistant to protection with either one or three cycles of IPC stimulus (42.7 ± 6.5% control versus 38.5 ± 1.9% 1 χ IPC, N = 6, NS; 41.4 ± 6.3% control versus 32.4 ± 3.2% 3 χ IPC, N = 10, NS). Western blot analysis, performed on heart samples taken from Akt1(-/-) mice subjected to IPC, revealed an impaired phosphorylation of GSK-3ß, a downstream effector of Akt, as well as Erk1/2, the parallel component of the reperfusion injury salvage kinase pathway. Akt2(-/-) mice, which exhibit a diabetic phenotype, however, were amenable to protection with three but not one cycle of IPC (46.4 ± 5.6% control versus 35.9 ± 5.0% in 1 χ IPC, N = 6, NS; 47.0 ± 6.0% control versus 30.8 ± 3.3% in 3 χ IPC, N = 6; *P = 0.039). Akt1 but not Akt2 is essential for mediating a protective response to an IPC stimulus. Impaired activation of GSK-3ß and Erk1/2 might be responsible for the lack of protective response to IPC in Akt1(-/-) mice. The rise in threshold for protection in Akt2(-/-) mice might be due to their diabetic phenotype.


Subject(s)
Ischemic Preconditioning, Myocardial , Proto-Oncogene Proteins c-akt/metabolism , Aging/pathology , Animals , Hemodynamics , Hyperglycemia/enzymology , Hyperglycemia/pathology , Immunoblotting , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/enzymology , Myocardium/pathology , Proto-Oncogene Proteins c-akt/deficiency , Signal Transduction , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...