Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Int J Mol Med ; 44(6): 2298-2310, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31638174

ABSTRACT

Phytoestrogens exert protective effects on the cardiovascular system through mechanisms that have yet to be clearly demonstrated. The aim of this study was to evaluate the protective effects exerted by genistein on cardiomyoblasts (H9C2) against oxidative stress, nitric oxide (NO) release, viability, proliferation/migration and mitochondrial function. H9C2 cultured in physiological or peroxidative conditions, were treated with genistein in the absence or presence of estrogen receptors (ERs), G protein­â€‹coupled­estrogenic­receptors (GPER), Akt, extracellular­â€‹signal­regulated kinases 1/2 (ERK1/2) and p38 mitogen activated protein kinase (p38MAPK) blockers. Cell viability, proliferation, migration, mitochondrial membrane potential, mitochondrial oxygen consumption and oxidant/antioxidant system, were measured by specific assays. Western blot assay was used for the analysis of NO synthase (NOS) subtypes' and expression and activation of various kinases. In all experiments 17ß­estradiol was used for comparison. The results showed that phytoestrogens and estrogens can increase cell viability, proliferation/migration and improve mitochondrial membrane potential and oxygen consumption of H9C2. Furthermore, NO release was modulated by genistein and 17ß­estradiol. These effects were reduced or abolished by the pre­treatment with ERs, GPER, Akt, ERK1/2 and p38MAPK blockers. Finally, a reduction of reactive oxygen species production and an increase of glutathione content was found in response to the two agents. In H9C2 cultured in physiological conditions, genistein induced endothelial NOS­dependent NO production through the involvement of estrogenic receptors and by the modulation of intracellular signalling related to Akt, ERK1/2, and p38MAPK. Moreover, estrogens and phytoestrogens protected H9C2 against oxidative stress by reducing inducible NOS expression and through the modulation of the antioxidant system and mitochondrial functioning.


Subject(s)
Cell Proliferation/drug effects , Cell Survival/drug effects , Genistein/pharmacology , Myoblasts, Cardiac/drug effects , Animals , Apoptosis/drug effects , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/genetics , Mitochondria/drug effects , Nitric Oxide Synthase/genetics , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects
2.
PLoS One ; 14(6): e0218437, 2019.
Article in English | MEDLINE | ID: mdl-31206561

ABSTRACT

BACKGROUND: Oxidative stress is involved in the pathogenesis and maintenance of pregnancy-related disorders, such as intrauterine growth restriction (IUGR) and preeclampsia (PE). Human umbilical cord mesenchymal stem cells (hUMSCs) have been suggested as a possible therapeutic tool for the treatment of pregnancy-related disorders in view of their paracrine actions on trophoblast cells. OBJECTIVES: To quantify the plasma markers of peroxidation in patients affected by PE and IUGR and to examine the role of oxidative stress in the pathophysiology of PE and IUGR in vitro by using hUMSCs from physiological and pathological pregnancies and a trophoblast cell line (HTR-8/SVneo). STUDY DESIGN: In pathological and physiological pregnancies the plasma markers of oxidative stress, arterial blood pressure, serum uric acid, 24h proteinuria, weight gain and body mass index (BMI) were examined. Furthermore, the pulsatility index (PI) of uterine and umbilical arteries, and of fetal middle cerebral artery was measured. In vitro, the different responses of hUMSCs, taken from physiological and pathological pregnancies, and of HTR-8/SVneo to pregnancy-related hormones in terms of viability and nitric oxide (NO) release were investigated. In some experiments, the above measurements were performed on co-cultures between HTR-8/SVneo and hUMSCs. RESULTS: The results obtained have shown that in pathological pregnancies, body mass index, serum acid uric, pulsatility index in uterine and umbilical arteries and markers of oxidative stress were higher than those found in physiological ones. Moreover, in PE and IUGR, a relation was observed between laboratory and clinical findings and the increased levels of oxidative stress. HTR-8/SVneo and hUMSCs showed reduced viability and increased NO production when stressed with H2O2. Finally, HTR-8/SVneo cultured in cross-talk with hUMSCs from pathological pregnancies showed a deterioration of cell viability and NO release when treated with pregnancy-related hormones. CONCLUSION: Our findings support that hUMSCs taken from patients affected by PE and IUGR have significant features in comparison with those from physiologic pregnancies. Moreover, the cross-talk between hUMSCs and trophoblast cells might be involved in the etiopathology of IUGR and PE secondary to oxidative stress.


Subject(s)
Cell Communication , Fetal Growth Retardation/etiology , Mesenchymal Stem Cells/physiology , Pre-Eclampsia/etiology , Trophoblasts/physiology , Adult , Cell Survival , Female , Humans , Nitric Oxide/metabolism , Oxidative Stress , Paracrine Communication/physiology , Pregnancy , Pregnancy Complications/etiology , Umbilical Cord/cytology
3.
Crit Rev Oncol Hematol ; 138: 241-254, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31121392

ABSTRACT

BACKGROUND: Effectiveness of music-based interventions (MI) on cancer patients' anxiety, depression, pain and quality of life (QoL) is a current research theme. MI are highly variable, making it challenging to compare studies. OBJECTIVE AND METHODS: To summarize the evidence on MI in cancer patients, 40 studies were reviewed following the PRISMA statement. Studies were included if assessing at least one outcome among anxiety, depression, QoL and pain in patients aged ≥ 18, with an active oncological/onco-haematological diagnosis, participating to any kind of Music Therapy (MT), during/after surgery, chemotherapy or radiotherapy. RESULTS: A positive effect of MI on the outcomes measured was supported. Greater reductions of anxiety and depression were observed in breast cancer patients. MI involving patients admitted to a hospital ward were less effective on QoL. CONCLUSION: The increasing evidence about MI effectiveness, tolerability, feasibility and appreciation, supports the need of MI implementation in Oncology, Radiotherapy and Surgery wards, and promotion of knowledge among health operators.


Subject(s)
Music Therapy/methods , Neoplasms/psychology , Anxiety/etiology , Anxiety/therapy , Depression/etiology , Depression/therapy , Humans , Quality of Life
4.
Acta Ophthalmol ; 97(4): e559-e567, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30585429

ABSTRACT

PURPOSE: Subthreshold micropulse laser (SMPL) has been increasingly used for the treatment of different retinal and choroidal macular disorders. However, the exact mechanisms of action have not yet been clearly defined. Therefore, we aimed to examine the role of SMPL treatment in the modulation of oxidant/antioxidant systems, apoptosis and autophagy in the mice eyes. METHODS: A specific laser contact lens for retina was positioned on the cornea of 40 mice (20 young and 20 old) in order to focus the laser on the eye fundus for SMPL treatment. Within 6 months, 20 animals received one treatment only, whereas the others were treated three times. Eye specimens underwent histological analysis and were used for thiobarbituric acid reactive substances (TBARS) and glutathione (GSH) quantification, as well as for the superoxide dismutase 1 (SOD1) and the selenoprotein thioredoxin reductase 1 (TrxR1) expression evaluation. Western blot was performed for nitric oxide synthase (NOS) subtypes detection and to examine changes in apoptotic/autophagy proteins expression. RESULTS: SMPL treatment reduced TBARS and increased GSH and SOD1 in the mice eyes. It also reduced cytochrome c, caspase 3 expression and activity and cleaved caspase 9, and increased Beclin 1, p62 and LC3ß. The effects were more relevant in the elderly animals. CONCLUSION: Our results showed that SMPL therapy restored the oxidant/antioxidant balance within retinal layers and modulated programmed forms of cell death. Further studies may confirm these data and could evaluate their relevance in clinical practice.


Subject(s)
Antioxidants/metabolism , Laser Coagulation/methods , Lasers, Semiconductor/therapeutic use , Oxidants/metabolism , Oxidative Stress , Retina/surgery , Retinal Diseases/surgery , Animals , Autophagy , Cell Death , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Retina/metabolism , Retina/pathology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Tomography, Optical Coherence/methods
5.
Nutrients ; 10(8)2018 Jul 27.
Article in English | MEDLINE | ID: mdl-30060502

ABSTRACT

Obesity can lead to pathological growth of adipocytes by inducing inflammation and oxidative stress. Genistein could be a potential candidate for the treatment of obesity due to its antioxidant properties. Specific kits were used to examine the effects of genistein vs adiponectin on human visceral pre-adipocytes differentiation, cell viability, mitochondrial membrane potential, and oxidative stress in pre-adipocytes and in white/brown adipocytes. Western Blot was performed to examine changes in protein activation/expression. Genistein increased human visceral pre-adipocytes differentiation and browning, and caused a dose-related improvement of cell viability and mitochondrial membrane potential. Similar effects were observed in brown adipocytes and in white adipocytes, although in white cells the increase of cell viability was inversely related to the dose. Moreover, genistein potentiated AMP-activated protein kinase (AMPK)/mitofusin2 activation/expression in pre-adipocytes and white/brown adipocytes and protected them from the effects of hydrogen peroxide. The effects caused by genistein were similar to those of adiponectin. The results obtained showed that genistein increases human visceral pre-adipocytes differentiation and browning, protected against oxidative stress in pre-adipocytes and white/brown adipocytes through mechanisms related to AMPK-signalling and the keeping of mitochondrial function.


Subject(s)
Adipocytes/drug effects , Antioxidants/pharmacology , Genistein/pharmacology , Glycine max/chemistry , Intra-Abdominal Fat/drug effects , Obesity, Abdominal/metabolism , Oxidative Stress/drug effects , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Adiponectin/metabolism , Animals , Cell Differentiation , Cell Survival , GTP Phosphohydrolases/metabolism , Humans , Hydrogen Peroxide/metabolism , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/metabolism , Mice , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Obesity, Abdominal/prevention & control , Phytotherapy , Plant Extracts/pharmacology , Signal Transduction , Uncoupling Protein 1/metabolism
6.
J Dermatol Sci ; 92(1): 62-77, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30104108

ABSTRACT

BACKGROUND: Estrogens and phytoestrogens can hinder the aging process through mechanisms related to estrogen receptors (ERs), guanine nucleotide-binding protein-coupled receptor (GPER30), mitochondria function and nitric oxide (NO) release. Up to date, however, the above issues are a matter of debate. OBJECTIVE: To examine the effects elicited by 17 ß-estradiol and genistein against peroxidation in human keratinocytes/fibroblasts and evaluate the role played by ERs, GPER30, mitochondria and NO. METHODS: Human fibroblasts/keratinocytes, either subjected to peroxidation or not, were exposed to 17 ß-estradiol/genistein in the absence or presence of the NO synthase (NOS) inhibitor, the ERs and GPER30 blockers, fulvestrant and G15, the phosphatidyl-inositol-3-kinase (PI3K-Akt), the p38 mitogen-activated protein (MAP) kinase and the extracellular signal-regulated kinases (ERK) 1/2 inhibitors. Specific kits were used for cell viability, NO, ROS and glutathione (GSH) detection and mitochondrial membrane potential measurement. Western Blot analysis was performed for kinases expression/activation detection. RESULTS: In physiological and peroxidative conditions, 17 ß-estradiol/genistein respectively increased and reduced NO release by fibroblasts/keratinocytes. Moreover, both agents prevented the ROS release and the fall of cell viability and mitochondrial membrane potential, while increasing GSH levels and the proliferation rate. Fulvestrant and G15 counteracted all above responses. Also, the NOS, and the kinases blockers reduced the protection exerted by 17 ß-estradiol/genistein on cell viability/mitochondria function. The involvement of PI3K-Akt and p38-MAPK was confirmed by Western blot. CONCLUSION: 17 ß-estradiol/genistein protected fibroblasts/keratinocytes against peroxidation by modulating oxidant/antioxidant system and mitochondria membrane potential, through mechanisms related to ERs and GPER30 and kinases activation.


Subject(s)
Antioxidants/pharmacology , Estradiol/pharmacology , Fibroblasts/drug effects , Genistein/pharmacology , Keratinocytes/drug effects , Oxidative Stress/drug effects , Skin/drug effects , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Nitric Oxide/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Skin/metabolism , Skin/pathology
7.
Cell Physiol Biochem ; 42(5): 1725-1738, 2017.
Article in English | MEDLINE | ID: mdl-28743128

ABSTRACT

BACKGROUND/AIMS: the anti-vascular endothelial growth factors (VEGF), Aflibercept and Ranibizumab, are used for the treatment of macular degeneration. Here we examined the involvement of nitric oxide (NO), mitochondria function and of apoptosis/autophagy in their antioxidant effects in human retinal pigment epithelium cells (RPE). METHODS: RPE were exposed to Ranibizumab/Aflibercept in the absence or presence of NO synthase (NOS) inhibitor and of autophagy activator/blocker, rapamicyn/3-methyladenine. Specific kits were used for cell viability, NO and reactive oxygen species detection and mitochondrial membrane potential measurement, whereas Western Blot was performed for apoptosis/ autophagy markers and other kinases detection. RESULTS: In RPE cultured in physiological conditions, Aflibercept/Ranibizumab increased NO release in a dose and time-dependent way. Opposite results were obtained in RPE pretreated with hydrogen peroxide. Moreover, both the anti-VEGF agents were able to prevent the fall of cell viability and of mitochondrial membrane potential. Those effects were reduced by the NOS inhibitor and 3-methyladenine and were potentiated by rapamycin. Finally, Aflibercept and Ranibizumab counteracted the changes of apoptosis/autophagy markers, NOS, Phosphatidylinositol-3-Kinase/Protein Kinase B and Extracellular signal-regulated kinases 1/2 caused by peroxidation. CONCLUSION: Aflibercept and Ranibizumab protect RPE against peroxidation through the modulation of NO release, apoptosis and autophagy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Autophagy/drug effects , Nitric Oxide/metabolism , Ranibizumab/pharmacology , Recombinant Fusion Proteins/pharmacology , Adenine/analogs & derivatives , Adenine/toxicity , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Hydrogen Peroxide/toxicity , Membrane Potential, Mitochondrial/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Vascular Endothelial Growth Factor , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Sirolimus/toxicity , Swine
8.
Cell Physiol Biochem ; 42(3): 1051-1062, 2017.
Article in English | MEDLINE | ID: mdl-28662498

ABSTRACT

BACKGROUND/AIMS: estrogens and phytoestrogens exert hepatoprotection through mechanisms not clearly examined yet. Here, we investigated the protective effects exerted by 17ß-estradiol and genistein against oxidative stress in hepatocytes and hepatic stellate cells (HSCs) and the involvement of specific receptors and the intracellular signalling. METHODS: Huh7.5 and LX-2, alone or in co-culture with Huh7.5, were treated with 17ß-estradiol and genistein alone or in the presence of menadione and of estrogen receptors (ERs) and G protein-coupled-estrogenic-receptors (GPER) blockers. Cell viability, mitochondrial membrane potential and oxidant/antioxidant system were measured by specific kits. Western Blot was used for the analysis of Akt and p38-mitogen-activated-protein kinases (MAPK) activation and α-smooth-muscle actin expression. RESULTS: In Huh7.5, 17ß-estradiol and genistein prevented the effects of peroxidation by modulating Akt and p38MAPK activation. Similar antioxidant and protective findings were obtained in LX-2 of co-culture experiments, only. ERs and GPER blockers were able to prevent the effects of 17ß-estradiol and genistein. CONCLUSION: In Huh7.5 and LX-2, 17ß-estradiol and genistein counteract the effects of peroxidation through the involvement of ERs and GPER and by an intracellular signalling related to Akt and p38MAPK. As concerning LX-2, paracrine factors released by Huh7.5 play a key role in protection against oxidative stress.


Subject(s)
Antioxidants/pharmacology , Estradiol/pharmacology , Genistein/pharmacology , Hepatocytes/drug effects , Oxidative Stress/drug effects , Phytoestrogens/pharmacology , Cell Line , Cell Survival/drug effects , Hepatocytes/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Data Brief ; 8: 1381-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27583345

ABSTRACT

We found that monomeric adiponectin was able to increase cell viability in porcine aortic endothelial cells (PAE) cultured both in normal and high glucose condition. Moreover, in normal glucose condition monomeric adiponectin increased p38MAPK, Akt, ERK1/2 and eNOS phosphorylation in a dose- and time-dependent way. Also in high glucose condition monomeric adiponectin increased eNOS and above kinases phosphorylation with similar patterns but at lower extent. For interpretation of the data presented in this article, please see the research article "Monomeric adiponectin modulates nitric oxide release and calcium movements in porcine aortic endothelial cells in normal/high glucose conditions" (Grossini et al., in press) [1].

10.
Life Sci ; 161: 1-9, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27469459

ABSTRACT

AIMS: Perivascular adipose tissue can be involved in the process of cardiovascular pathology through the release of adipokines, namely adiponectins. Monomeric adiponectin has been shown to increase coronary blood flow in anesthetized pigs through increased nitric oxide (NO) release and the involvement of adiponectin receptor 1 (AdipoR1). The present study was therefore planned to examine the effects of monomeric adiponectin on NO release and Ca(2+) transients in porcine aortic endothelial cells (PAEs) in normal/high glucose conditions and the related mechanisms. MAIN METHODS: PAEs were treated with monomeric adiponectin alone or in the presence of intracellular kinases blocker, AdipoR1 and Ca(2+)-ATPase pump inhibitors. The role of Na(+)/Ca(2+) exchanger was examined in experiments performed in zero Na(+) medium. NO release and intracellular Ca(2+) were measured through specific probes. KEY FINDINGS: In PAE cultured in normal glucose conditions, monomeric adiponectin elevated NO production and [Ca(2+)]c. Similar effects were observed in high glucose conditions, although the response was lower and not transient. The Ca(2+) mobilized by monomeric adiponectin originated from an intracellular pool thapsigargin- and ATP-sensitive and from the extracellular space. Moreover, the effects of monomeric adiponectin were prevented by kinase blockers and AdipoR1 inhibitor. Finally, in normal glucose condition, a role for Na(+)/Ca(2+) exchanger and Ca(2+)-ATPase pump in restoring Ca(2+) was found. SIGNIFICANCE: Our results add new information about the control of endothelial function elicited by monomeric adiponectin, which would be achieved by modulation of NO release and Ca(2+) transients. A signalling related to Akt, ERK1/2 and p38MAPK downstream AdipoR1 would be involved.


Subject(s)
Adiponectin/pharmacology , Aorta/drug effects , Calcium/metabolism , Endothelium, Vascular/drug effects , Glucose/metabolism , Nitric Oxide/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Ion Transport , Swine
11.
Endocrinology ; 157(6): 2403-15, 2016 06.
Article in English | MEDLINE | ID: mdl-27100620

ABSTRACT

Des-acyl ghrelin (DAG), the most abundant form of ghrelin in humans, has been found to reduce arterial blood pressure and prevent cardiac and endothelial cell apoptosis. Despite this, data regarding its direct effect on cardiac function and coronary blood flow, as well as the related involvement of autonomic nervous system and nitric oxide (NO), are scarce. We therefore examined these issues using both in vivo and in vitro studies. In 20 anesthetized pigs, intracoronary 100 pmol/mL DAG infusion with a constant heart rate and aortic blood pressure, increased coronary blood flow and NO release, whereas reducing coronary vascular resistances (P < .05). Dose responses to DAG were evaluated in five pigs. No effects on cardiac contractility/relaxation or myocardial oxygen consumption were observed. Moreover, whereas the blockade of muscarinic cholinoceptors (n = 5) or α- and ß-adrenoceptors (n = 5 each) did not abolish the observed responses, NO synthase inhibition (n = 5) prevented the effects of DAG on coronary blood flow and NO release. In coronary artery endothelial cells, DAG dose dependently increased NO release through cAMP signaling and ERK1/2, Akt, and p38 MAPK involvement as well as the phosphorylation of endothelial NO synthase. In conclusion, in anesthetized pigs, DAG primarily increased cardiac perfusion through the involvement of NO release. Moreover, the phosphorylation of ERK1/2 and Akt appears to play roles in eliciting the observed NO production in coronary artery endothelial cells.


Subject(s)
Ghrelin/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Animals , Coronary Circulation/drug effects , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Female , Ghrelin/administration & dosage , Heart/drug effects , Heart Rate/drug effects , Hemodynamics/drug effects , MAP Kinase Signaling System/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Swine , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Cell Physiol Biochem ; 36(6): 2108-20, 2015.
Article in English | MEDLINE | ID: mdl-26279419

ABSTRACT

BACKGROUND/AIM: Previous reports have made it hypothetically possible that human chorionic gonadotropin (hCG) could protect against the onset of pregnancy-related pathological conditions by acting as an antioxidant. In the present study we planned to examine the effects of hCG against oxidative stress in human umbilical vein endothelial cells (HUVEC). METHODS: HUVEC were subjected to peroxidation by hydrogen peroxide. The modulation of nitric oxide (NO) release by hCG and its effects on cell viability, glutathione (GSH) levels, mitochondrial membrane potential and mitochondrial transition pore opening (MPTP) were examined by specific dyes. Endothelial and inducible NO synthase (eNOS and iNOS), Akt and extracellular -signal-regulated kinases 1/2 (ERK1/2) activation and markers of apoptosis were analyzed by Western Blot. RESULTS: In HUVEC, hCG reduced NO release by modulating eNOS and iNOS. Moreover, hCG protected HUVEC against oxidative stress by preventing GSH reduction and apoptosis, by maintaining Akt and ERK1/2 activation and by keeping mitochondrial function. CONCLUSION: The present results have for the first time shown protective effects exerted by hCG on vascular endothelial function, which would be achieved by modulation of NO release, antioxidant and antiapoptotic actions and activation of cell survival signalling. These findings could have clinical implications in the management of pregnancy-related disorders.


Subject(s)
Apoptosis/drug effects , Chorionic Gonadotropin/pharmacology , Mitochondria/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Signal Transduction/drug effects , Antioxidants/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Survival/drug effects , Cytochromes c/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/pharmacology , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , bcl-2-Associated X Protein/metabolism
13.
Phytother Res ; 29(9): 1339-1348, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26032176

ABSTRACT

Artemetin is one of the main components of Achillea millefolium L. and Artemisia absinthium, which have long been used for the treatment of various diseases. To date, however, available information about protective effects of their extracts on the cardiovascular system is scarce. Therefore, we planned to analyze the effects of artemetin on nitric oxide (NO) release and the protection exerted against oxidation in porcine aortic endothelial (PAE) cells. In PAE, we examined the modulation of NO release caused by artemetin and the involvement of muscarinic receptors, ß2-adrenoreceptors, estrogenic receptors (ER), protein-kinase A, phospholipase-C, endothelial-NO-synthase (eNOS), Akt, extracellular-signal-regulated kinases 1/2 (ERK1/2) and p38 mitogen activated protein kinase (p38 MAPK). Moreover, in cells treated with hydrogen peroxide, the effects of artemetin were examined on cell survival, glutathione (GSH) levels, apoptosis, mitochondrial membrane potential and transition pore opening. Artemetin increased eNOS-dependent NO production by the involvement of muscarinic receptors, ß2-adrenoreceptors, ER and all the aforementioned kinases. Furthermore, artemetin improved cell viability in PAE that were subjected to peroxidation by counteracting GSH depletion and apoptosis and through the modulation of mitochondrial function. In conclusion, artemetin protected endothelial function by acting as antioxidant and antiapoptotic agent and through the activation of ERK1/2 and Akt. Copyright © 2015 John Wiley & Sons, Ltd.

14.
PLoS One ; 10(4): e0124742, 2015.
Article in English | MEDLINE | ID: mdl-25880552

ABSTRACT

BACKGROUND: Levosimendan protects rat liver against peroxidative injuries through mechanisms related to nitric oxide (NO) production and mitochondrial ATP-dependent K (mitoKATP) channels opening. However, whether levosimendan could modulate the cross-talk between apoptosis and autophagy in the liver is still a matter of debate. Thus, the aim of this study was to examine the role of levosimendan as a modulator of the apoptosis/autophagy interplay in liver cells subjected to peroxidation and the related involvement of NO and mitoKATP. METHODS AND FINDINGS: In primary rat hepatocytes that have been subjected to oxidative stress, Western blot was performed to examine endothelial and inducible NO synthase isoforms (eNOS, iNOS) activation, apoptosis/autophagy and survival signalling detection in response to levosimendan. In addition, NO release, cell viability, mitochondrial membrane potential and mitochondrial permeability transition pore opening (MPTP) were examined through specific dyes. Some of those evaluations were also performed in human hepatic stellate cells (HSC). Pre-treatment of hepatocytes with levosimendan dose-dependently counteracted the injuries caused by oxidative stress and reduced NO release by modulating eNOS/iNOS activation. In hepatocytes, while the autophagic inhibition reduced the effects of levosimendan, after the pan-caspases inhibition, cell survival and autophagy in response to levosimendan were increased. Finally, all protective effects were prevented by both mitoKATP channels inhibition and NOS blocking. In HSC, levosimendan was able to modulate the oxidative balance and inhibit autophagy without improving cell viability and apoptosis. CONCLUSIONS: Levosimendan protects hepatocytes against oxidative injuries by autophagic-dependent inhibition of apoptosis and the activation of survival signalling. Such effects would involve mitoKATP channels opening and the modulation of NO release by the different NOS isoforms. In HSC, levosimendan would also play a role in cell activation and possible evolution toward fibrosis. These findings highlight the potential of levosimendan as a therapeutic agent for the treatment or prevention of liver ischemia/reperfusion injuries.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Hepatocytes/drug effects , Hydrazones/pharmacology , Lipid Peroxidation/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Oxidative Stress/drug effects , Pyridazines/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Blotting, Western , Hepatocytes/cytology , Humans , Male , Mitochondrial Permeability Transition Pore , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Simendan
SELECTION OF CITATIONS
SEARCH DETAIL
...