Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Skin Pharmacol Physiol ; 35(1): 1-12, 2022.
Article in English | MEDLINE | ID: mdl-34237733

ABSTRACT

BACKGROUND: The altered balance between oxidants/antioxidants and inflammation, changes in nitric oxide (NO) release, and mitochondrial function have a role in skin aging through fibroblast modulation. Tocopherol is promising in counteracting the abovementioned events, but the effective mechanism of action needs to be clarified. OBJECTIVE: The aim of this study was to examine the effects of α-tocopherol on cell viability/proliferation, NO release, mitochondrial function, oxidants/antioxidants, and inflammation in human dermal fibroblasts (HDF) subjected to oxidative stress. METHODS: HDF were treated with H2O2 in the presence or absence of 1-10 µM α-tocopherol. Cell viability, reactive oxygen species (ROS), NO release, and mitochondrial membrane potential were measured; glutathione (GSH), superoxide dismutase (SOD)-1 and -2, glutathione peroxidase-1 (GPX-1), inducible NO synthase (iNOS), and Ki-67 were evaluated by RT-PCR and immunofluorescence; cell cycle was analyzed using FACS. Pro- and anti-inflammatory cytokine gene expression was analyzed through qRT-PCR. RESULTS: α-Tocopherol counteracts H2O2, although it remains unclear whether this effect is dose dependent. Improvement of cell viability, mitochondrial membrane potential, Ki-67 expression, and G0/G1 and G2/M phases of the cell cycle was observed. These effects were accompanied by the increase of GSH content and the reduction of SOD-1 and -2, GPX-1, and ROS release. Also, iNOS expression and NO release were inhibited, and pro-inflammatory cytokine gene expression was decreased, confirming the putative role of α-tocopherol against inflammation. CONCLUSION: α-Tocopherol exerts protective effects in HDF which underwent oxidative stress by modulating the redox status, inflammation, iNOS-dependent NO release, and mitochondrial function. These observations have a potential role in the prevention and treatment of photoaging-related skin cancers.


Subject(s)
Nitric Oxide , alpha-Tocopherol , Antioxidants/metabolism , Antioxidants/pharmacology , Fibroblasts/metabolism , Humans , Hydrogen Peroxide , Inflammation/drug therapy , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species , alpha-Tocopherol/pharmacology
3.
J Nat Prod ; 83(6): 1740-1750, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32496797

ABSTRACT

Bitter taste receptors (hTAS2R) are expressed ectopically in various tissues, raising the possibility of a pharmacological exploitation. This seems of particular relevance in airways, since hTAS2Rs are involved in the protection of the aerial tissues from infections and in bronchodilation. The bis-guaianolide absinthin (1), one of the most bitter compounds known, targets the hTAS2R46 bitter receptor. Absinthin (1), an unstable compound, readily turns into anabsinthin (2) with substantial retention of the bitter properties, and this compound was used as a starting material to explore the chemical space around the bis-guaianolide bitter pharmacophore. Capitalizing on the chemoselective opening of the allylic lactone ring, the esters 3 and 4, and the nor-azide 6 were prepared and assayed on human bronchoepithelial (BEAS-2B) cells expressing hTAS2R46. Anti-inflammatory activity was evaluated by measuring the expression of MUC5AC, iNOS, and cytokines, as well as the production of superoxide anion, qualifying the methyl ester 3 as the best candidate for additional studies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bronchi/drug effects , Epithelial Cells/drug effects , Sesquiterpenes, Guaiane/pharmacology , Artemisia/chemistry , Bronchi/cytology , Calcium/metabolism , Cell Line , Cytokines/antagonists & inhibitors , Esters/chemistry , Humans , Molecular Structure , Mucin-5B/drug effects , Nitric Oxide Synthase Type II/drug effects , Receptors, G-Protein-Coupled/drug effects , Superoxides/metabolism , Taste Buds
4.
Cell Physiol Biochem ; 54(3): 401-416, 2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32330379

ABSTRACT

BACKGROUND/AIMS: Oxidative stress and mitochondria dysfunction could be involved in the onset of non-alcoholic fatty liver disease (NAFLD) and in its progression to non-alcoholic steatohepatitis (NASH). Estrogens/phytoestrogens could counteract liver fat deposition with beneficial effects against NAFLD by unclear mechanisms. We aimed to analyze the protective effects elicited by genistein/estradiol in hepatocytes cultured in NAFLD-like medium on cell viability, triglycerides accumulation, mitochondrial function and oxidative stress and the role of NLRP3 inflammasome, toll like receptors 4 (TLR4), Akt and 5' AMP-activated protein kinase (AMPK)α1/2. METHODS: Human primary hepatocytes/hepatoma cell line (Huh7.5 cells) were incubated with a 2 mM mixture of oleate/palmitate in presence/absence of genistein/17ß-estradiol. In some experiments, Huh7.5 cells were exposed to various inhibitors of the above pathways and estrogenic receptors (ERs) and G protein-coupled estrogen receptor (GPER) blockers, before genistein/17ß-estradiol. Cell viability, mitochondrial membrane potential, reactive oxygen species and triglycerides content were examined by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT), 5,51,6,61-tetrachloro-1,11,3,31 tetraethylbenzimidazolyl carbocyanine iodide (JC-1), 2,7-dichlorodihydrofluorescein diacetate (H2DCFDA) and the Triglyceride Colorimetric Assay. The expression/activation of kinases was analyzed by means of Western blot. RESULTS: Genistein/17ß-estradiol protected hepatocytes against NAFLD-like medium, by preventing the loss of cell viability and mitochondrial function, triglycerides accumulation and peroxidation. The blocking of kinases, ERs and GPER was able to reduce the above effects, which were potentiated by NLRP3 inflammasome. CONCLUSION: Our findings suggest novel mechanisms underlying the protective effects elicited by phytoestrogens/estrogens against NAFLD/NASH and open novel therapeutic perspectives in the management of NAFLD in postmenopausal women.


Subject(s)
Cell Survival/drug effects , Estradiol/pharmacology , Genistein/pharmacology , Hepatocytes/drug effects , Inflammasomes/metabolism , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , AMP-Activated Protein Kinase Kinases , Cell Line , Hepatocytes/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects , Phytoestrogens/pharmacology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Triglycerides/metabolism
5.
J Biol Chem ; 294(33): 12472-12482, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31248983

ABSTRACT

Type 2 taste receptors (TAS2R) are G protein-coupled receptors first described in the gustatory system, but have also been shown to have extraoral localizations, including airway smooth muscle (ASM) cells, in which TAS2R have been reported to induce relaxation. TAS2R46 is an unexplored subtype that responds to its highly specific agonist absinthin. Here, we first demonstrate that, unlike other bitter-taste receptor agonists, absinthin alone (1 µm) in ASM cells does not induce Ca2+ signals but reduces histamine-induced cytosolic Ca2+ increases. To investigate this mechanism, we introduced into ASM cells aequorin-based Ca2+ probes targeted to the cytosol, subplasma membrane domain, or the mitochondrial matrix. We show that absinthin reduces cytosolic histamine-induced Ca2+ rises and simultaneously increases Ca2+ influx into mitochondria. We found that this effect is inhibited by the potent human TAS2R46 (hTAS2R46) antagonist 3ß-hydroxydihydrocostunolide and is no longer evident in hTAS2R46-silenced ASM cells, indicating that it is hTAS2R46-dependent. Furthermore, these changes were sensitive to the mitochondrial uncoupler carbonyl cyanide p-(trifluoromethoxy)phenyl-hydrazone (FCCP); the mitochondrial calcium uniporter inhibitor KB-R7943 (carbamimidothioic acid); the cytoskeletal disrupter latrunculin; and an inhibitor of the exchange protein directly activated by cAMP (EPAC), ESI-09. Similarly, the ß2 agonist salbutamol also could induce Ca2+ shuttling from cytoplasm to mitochondria, suggesting that this new mechanism might be generalizable. Moreover, forskolin and an EPAC activator mimicked this effect in HeLa cells. Our findings support the hypothesis that plasma membrane receptors can positively regulate mitochondrial Ca2+ uptake, adding a further facet to the ability of cells to encode complex Ca2+ signals.


Subject(s)
Calcium Signaling/drug effects , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, G-Protein-Coupled/agonists , Respiratory System/metabolism , Sesquiterpenes, Guaiane/pharmacology , Calcium/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Line , Endoplasmic Reticulum/genetics , HeLa Cells , Humans , Mitochondria/genetics , Myocytes, Smooth Muscle/cytology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Respiratory System/cytology , Thiourea/analogs & derivatives , Thiourea/pharmacology
6.
J Nat Prod ; 81(3): 630-633, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29240420

ABSTRACT

Treatment with iodine cleanly converts various p-menthane-type phytocannabinoids and their carboxylated precursors into cannabinol (CBN, 1a). The reaction is superior to previously reported protocols in terms of simplicity and substrate range, which includes not only tricyclic tetrahydrocannabinols such as Δ9-THC (2a) but also bicyclic phytocannabinoids such as cannabidiol (CBD, 3a). Lower homologues from the viridin series (2c and 3c, respectively) afforded cannabivarin (CBV), a non-narcotic compound that, when investigated against a series of ionotropic (thermo-TRPs) biological end-points of phytocannabinoids, retained the submicromolar TRPA1-activating and TRPM8-inhibiting properties of CBN, while also potently activating TRPV2. Treatment with iodine provides an easy access to CBN (1a) from crude extracts and side-cuts of the purification of Δ9-THC and CBD from respectively narcotic Cannabis sativa (marijuana) and fiber hemp, substantially expanding the availability of this compound and, in the case of fiber hemp, dissecting it from narcotic phytocannabinoids.


Subject(s)
Cannabinoid Receptor Agonists/chemistry , Iodine/chemistry , Cannabidiol/chemistry , Cannabidiol/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/chemistry , Cannabinoids/pharmacology , Cannabinol/chemistry , Cannabinol/pharmacology , Cannabis/chemistry , Cell Line , Dronabinol/chemistry , Dronabinol/pharmacology , HEK293 Cells , Humans , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
7.
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
8.
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.

9.
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
10.
J Endocrinol ; 222(1): 137-49, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24860147

ABSTRACT

Adiponectin, the most abundant adipokine released by adipose tissue, appears to play an important role in the regulation of vascular endothelial and cardiac function. To date, however, the physiological effects of human monomeric adiponectin on the coronary vasculature and myocardial systo-diastolic function, as well as on parasympathetic/sympathetic involvement and nitric oxide (NO) release, have not yet been investigated. Thus, we planned to determine the primary in vivo effects of human monomeric adiponectin on coronary blood flow and cardiac contractility/relaxation and the related role of autonomic nervous system, adiponectin receptors, and NO. In 30 anesthetized pigs, human monomeric adiponectin was infused into the left anterior descending coronary artery at constant heart rate and arterial blood pressure, and the effects on coronary blood flow, left ventricular systo-diastolic function, myocardial oxygen metabolism, and NO release were examined. The mechanisms of the observed hemodynamic responses were also analyzed by repeating the highest dose of human monomeric adiponectin infusion after autonomic nervous system and NO blockade, and after specific adiponectin 1 receptor antagonist administration. Intracoronary human monomeric adiponectin caused dose-related increases of coronary blood flow and cardiac function. Those effects were accompanied by increased coronary NO release and coronary adiponectin levels. Moreover, the vascular effects of the peptide were prevented by blockade of ß2-adrenoceptors and NO synthase, whereas all effects of human monomeric adiponectin were prevented by adiponectin 1 receptor inhibitor. In conclusion, human monomeric adiponectin primarily increased coronary blood flow and cardiac systo-diastolic function through the involvement of specific receptors, ß2-adrenoceptors, and NO release.


Subject(s)
Adiponectin/pharmacology , Coronary Vessels/physiology , Heart/drug effects , Heart/physiology , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Unconsciousness , Anesthetics , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Heart Rate/drug effects , Heart Rate/physiology , Humans , Models, Animal , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Nitric Oxide/metabolism , Receptors, Adiponectin/drug effects , Receptors, Adiponectin/physiology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...