Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 918: 174778, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35093322

ABSTRACT

Quercetin, a flavonoid abundantly present in the Mediterranean diet, is considered a vasodilator despite its recognized capability to stimulate vascular CaV1.2 channel current (ICa1.2). The present study was undertaken to assess its possible vasocontractile activity. Functional and electrophysiology experiments were performed in vitro on rat aorta rings and tail artery myocytes along with an in-depth molecular modelling analysis. The CaV1.2 channel stimulator (S)-(-)-methyl-1,4-dihydro-2,6-dimethyl-3-nitro-4-(2-trifluoromethylphenyl) pyridine-5-carboxylate (Bay K 8644) was used as reference compound. Quercetin and Bay K 8644 caused a significant leftward shift of KCl concentration-response curve. Neither agent affected basal muscle tone, though in rings pre-treated with thapsigargin or 15 mM KCl they caused a strong, concentration-dependent contraction. Both quercetin and Bay K 8644 potentiated the response to Ca2+ in weakly depolarised rings. At high KCl concentrations, however, quercetin caused vasorelaxation. While Bay K 8644 stimulated ICa1.2, this effect being sustained with time, quercetin-induced stimulation was transient, although the molecule in solution underwent only marginal oxidation. Quercetin transient stimulation was not affected by pre-treatment with isoprenaline, sodium nitroprusside, or dephostatin; however, it converted to a sustained one in myocytes pre-incubated with Gö6976. Classical molecular dynamics simulations revealed that quercetin and Bay K 8644 formed hydrogen bonds with target sensing residues of CaV1.2 channel favouring the inactivated conformation. In conclusion, quercetin-induced stimulation of ICa1.2 promoted vasocontraction when Ca2+ buffering function of sarcoplasmic reticulum was impaired and/or smooth muscle cell membrane was moderately depolarised, as it may occur under certain pathological conditions.


Subject(s)
3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Arteries , Calcium Channels, L-Type/metabolism , Muscle Contraction/drug effects , Muscle, Smooth, Vascular , Quercetin/pharmacology , Vasodilation/drug effects , Animals , Antioxidants/pharmacology , Arteries/drug effects , Arteries/pathology , Arteries/physiology , Calcium Channel Agonists/pharmacology , Electrophysiological Phenomena/drug effects , Molecular Dynamics Simulation , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Rats , Vasodilator Agents/pharmacology
2.
Eur J Pharmacol ; 899: 174030, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33727059

ABSTRACT

The cardiac action potential is regulated by several ion channels. Drugs capable to block these channels, in particular the human ether-à-go-go-related gene (hERG) channel, also known as KV11.1 channel, may lead to a potentially lethal ventricular tachyarrhythmia called "Torsades de Pointes". Thus, evaluation of the hERG channel off-target activity of novel chemical entities is nowadays required to safeguard patients as well as to avoid attrition in drug development. Flavonoids, a large class of natural compounds abundantly present in food, beverages, herbal medicines, and dietary food supplements, generally escape this assessment, though consumed in consistent amounts. Continuously growing evidence indicates that these compounds may interact with the hERG channel and block it. The present review, by examining numerous studies, summarizes the state-of-the-art in this field, describing the most significant examples of direct and indirect inhibition of the hERG channel current operated by flavonoids. A description of the molecular interactions between a few of these natural molecules and the Rattus norvegicus channel protein, achieved by an in silico approach, is also presented.


Subject(s)
ERG1 Potassium Channel/antagonists & inhibitors , Flavonoids/toxicity , Heart Rate/drug effects , Long QT Syndrome/chemically induced , Myocytes, Cardiac/drug effects , Potassium Channel Blockers/toxicity , Torsades de Pointes/chemically induced , Action Potentials , Animals , ERG1 Potassium Channel/chemistry , ERG1 Potassium Channel/metabolism , Humans , Long QT Syndrome/metabolism , Long QT Syndrome/physiopathology , Myocytes, Cardiac/metabolism , Protein Conformation , Risk Assessment , Risk Factors , Structure-Activity Relationship , Torsades de Pointes/metabolism , Torsades de Pointes/physiopathology
3.
Acta Pharmacol Sin ; 41(9): 1158-1166, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32132658

ABSTRACT

CaV1.2 channel blockers or 5-HT2 receptor antagonists constitute effective therapy for Raynaud's syndrome. A functional link between the inhibition of 5-HT2 receptors and CaV1.2 channel blockade in arterial smooth muscles has been hypothesized. Therefore, the effects of ritanserin, a nonselective 5-HT2 receptor antagonist, on vascular CaV1.2 channels were investigated through electrophysiological, functional, and computational studies. Ritanserin blocked CaV1.2 channel currents (ICa1.2) in a concentration-dependent manner (Kr = 3.61 µM); ICa1.2 inhibition was antagonized by Bay K 8644 and partially reverted upon washout. Conversely, the ritanserin analog ketanserin (100 µM) inhibited ICa1.2 by ~50%. Ritanserin concentration-dependently shifted the voltage dependence of the steady-state inactivation curve to more negative potentials (Ki = 1.58 µM) without affecting the slope of inactivation and the activation curve, and decreased ICa1.2 progressively during repetitive (1 Hz) step depolarizations (use-dependent block). The addition of ritanserin caused the contraction of single myocytes not yet dialyzed with the conventional method. Furthermore, in depolarized rings, ritanserin, and to a lesser extent, ketanserin, caused a concentration-dependent relaxation, which was antagonized by Bay K 8644. Ritanserin and ketanserin were docked at a region of the CaV1.2 α1C subunit nearby that of Bay K 8644; however, only ritanserin and Bay K 8644 formed a hydrogen bond with key residue Tyr-1489. In conclusion, ritanserin caused in vitro vasodilation, accomplished through the blockade of CaV1.2 channels, which was achieved preferentially in the inactivated and/or resting state of the channel. This novel activity encourages the development of ritanserin derivatives for their potential use in the treatment of Raynaud's syndrome.


Subject(s)
Calcium Channels, L-Type/metabolism , Electrophysiological Phenomena/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Ritanserin/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Arteries/cytology , Binding Sites , Calcium Channels, L-Type/chemistry , Ketanserin/metabolism , Ketanserin/pharmacology , Male , Molecular Docking Simulation , Muscle, Smooth, Vascular/cytology , Protein Binding , Rats, Wistar , Ritanserin/metabolism , Serotonin 5-HT2 Receptor Antagonists/metabolism , Vasoconstriction/drug effects
4.
Planta Med ; 86(4): 284-293, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31994147

ABSTRACT

Dalbergia species heartwood, widely used in traditional medicine to treat various cardiovascular diseases, might represent a rich source of vasoactive agents. In Vietnam, Dalbergia tonkinensis is an endemic tree. Therefore, the aim of the present work was to investigate the vascular activity of R-(-)-3'-hydroxy-2,4,5-trimethoxydalbergiquinol isolated from the heartwood of D. tonkinensis and to provide circular dichroism features of its R absolute configuration. The vascular effects of R-(-)-3'-hydroxy-2,4,5-trimethoxydalbergiquinol were assessed on the in vitro mechanical activity of rat aorta rings, under isometric conditions, and on whole-cell Ba2+ currents through CaV1.2 channels (IBa1.2) recorded in single, rat tail main artery myocytes by means of the patch-clamp technique. R-(-)-3'-Hydroxy-2,4,5-trimethoxydalbergiquinol showed concentration-dependent, vasorelaxant activity on both endothelium-deprived and endothelium intact rings precontracted with the α 1 receptor agonist phenylephrine. Neither the NO (nitric oxide) synthase inhibitor Nω-nitro-L-arginine methyl ester nor the cyclooxygenase inhibitor indomethacin affected its spasmolytic activity. R-(-)-3'-Hydroxy-2,4,5-trimethoxydalbergiquinol-induced vasorelaxation was antagonized by (S)-(-)-Bay K 8644 and unaffected by tetraethylammonium plus glibenclamide. In patch-clamp experiments, R-(-)-3'-hydroxy-2,4,5-trimethoxydalbergiquinol inhibited IBa1.2 in a concentration-dependent manner and significantly decreased the time constant of current inactivation. R-(-)-3'-Hydroxy-2,4,5-trimethoxydalbergiquinol likely stabilized the channel in its closed state, as suggested by molecular modelling and docking simulation to the CaV1.2 channel α 1c subunit. In conclusion, D. tonkinensis species may represent a source of agents potentially useful for the development of novel antihypertensive drugs.


Subject(s)
Dalbergia , Vasodilation , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester , Animals , Aorta, Thoracic , Endothelium, Vascular , Rats , Vasodilator Agents , Vietnam
5.
Biochem Pharmacol ; 155: 434-443, 2018 09.
Article in English | MEDLINE | ID: mdl-30036502

ABSTRACT

3,5-Di-t-butyl-4-hydroxyanisole (DTBHA) is considered as an activator of the skeletal muscle sarcoplasmic reticulum (SR) Ca2+-uptake, endowed with antioxidant and L-type Ca2+ channel blocking activities. In this study we assessed the cardiac effects of DTBHA on Langendorff perfused rat hearts, isolated rat atria and rat cardiac SR membrane vesicles, as well as on several SERCA isoforms of membrane preparations. Moreover, in order to clarify its molecular mechanism of action Ca2+ imaging experiments were carried out on HEK293 cells transiently transfected with RyR2 channel. Docking of DTBHA at the rat RyR2 protein was investigated in silico. In Langendorff perfused rat hearts, DTBHA significantly increased, in a concentration-dependent manner, left ventricular pressure and diastole duration, while reducing heart rate and the time-constant of isovolumic relaxation, leaving unaltered coronary perfusion pressure. At the maximum concentration tested (30 µM), it significantly prolonged PQ interval, but left the corrected QT intervals unaffected. In spontaneously beating atria, DTBHA decreased sinus rate in a concentration-dependent manner. DTBHA, at concentrations higher than 10 µM, increased Ca2+ uptake in cardiac SR without affecting Ca2+-dependent ATPase activity assayed on several SERCA isoforms. Moreover, DTBHA antagonized thapsigargin-stimulated Ca2+ leak in cardiac SR and reduced caffeine-induced, RyR2-activated Ca2+ release in RyR2 expressing HEK293 cells. Using computational approaches, DTBHA showed a good affinity outline into binding sites of RyR2 protein. In conclusion, DTBHA behaved like a negative chronotropic, a positive inotropic and a lusitropic agent on rat heart preparations and improved cardiac SR Ca2+ uptake by lowering SR Ca2+ leak.


Subject(s)
Butylated Hydroxyanisole/analogs & derivatives , Calcium/metabolism , Heart Rate/drug effects , Myocardial Contraction/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Butylated Hydroxyanisole/metabolism , Butylated Hydroxyanisole/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Heart Rate/physiology , Humans , Isolated Heart Preparation/methods , Male , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Rats , Rats, Wistar
6.
Toxicol In Vitro ; 47: 89-93, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29158021

ABSTRACT

This study was designed to unveil effects of 5-nm sized, polyvinylpyrrolidone-coated gold nanoparticles (AuNPs) on vascular CaV1.2 and CaV3.1 channels. Ba2+ currents through both channels (IBa1.2 and IBa3.1) were recorded in single myocytes isolated from the rat tail main artery by means of the whole-cell patch-clamp method. AuNPs increased IBa1.2 and IBa3.1 amplitude in a concentration- and Vh-dependent manner. Neither the voltage dependence of inactivation and activation curves nor inactivation and activation kinetics were affected by AuNPs. In conclusion, these findings warrant further investigation to clarify whether different types of NPs possess the same stimulatory activity and may represent a toxic hazard to humans.


Subject(s)
Arteries/drug effects , Calcium Channels, L-Type/metabolism , Calcium Channels/metabolism , Environmental Pollutants/toxicity , Gold/toxicity , Metal Nanoparticles/toxicity , Muscle, Smooth, Vascular/drug effects , Animals , Arteries/cytology , Arteries/metabolism , Barium/metabolism , Calcium Channels/chemistry , Calcium Channels, L-Type/chemistry , Cells, Cultured , Environmental Pollutants/chemistry , Gold/chemistry , Male , Membrane Potentials , Metal Nanoparticles/chemistry , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Particle Size , Patch-Clamp Techniques , Povidone/chemistry , Povidone/toxicity , Rats, Wistar , Single-Cell Analysis , Surface Properties , Tail , Toxicity Tests, Acute
7.
Biochem Pharmacol ; 140: 53-63, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28583845

ABSTRACT

To characterize the role of cAMP-dependent protein kinase (PKA) in regulating vascular Ca2+ current through Cav1.2 channels [ICa1.2], we have documented a marked capacity of the isoquinoline H-89, widely used as a PKA inhibitor, to reduce current amplitude. We hypothesized that the ICa1.2 inhibitory activity of H-89 was mediated by mechanisms unrelated to PKA inhibition. To support this, an in-depth analysis of H-89 vascular effects on both ICa1.2 and contractility was undertaken by performing whole-cell patch-clamp recordings and functional experiments in rat tail main artery single myocytes and rings, respectively. H-89 inhibited ICa1.2 with a pIC50 (M) value of about 5.5, even under conditions where PKA activity was either abolished by both the PKA antagonists KT5720 and protein kinase inhibitor fragment 6-22 amide or enhanced by the PKA stimulators 6-Bnz-cAMP and 8-Br-cAMP. Inhibition of ICa1.2 by H-89 appeared almost irreversible upon washout, was charge carrier- and voltage-dependent, and antagonised by the Cav1.2 channel agonist (S)-(-)-Bay K 8644. H-89 did not alter both potency and efficacy of verapamil, did not affect current kinetics or voltage-dependent activation, while shifting to the left the 50% voltage of inactivation in a concentration-dependent manner. H-89 docked at the α1C subunit in a pocket region close to that of (S)-(-)-Bay K 8644 docking, forming a hydrogen bond with the same, key amino acid residue Tyr-1489. Finally, both high K+- and (S)-(-)-Bay K 8644-induced contractions of rings were fully reverted by H-89. In conclusion, these results indicate that H-89 inhibited vascular ICa1.2 and, consequently, the contractile function through a PKA-independent mechanism. Therefore, caution is recommended when interpreting experiments where H-89 is used to inhibit vascular smooth muscle PKA.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Isoquinolines/metabolism , Models, Molecular , Muscle, Smooth, Vascular/physiology , Protein Kinase Inhibitors/metabolism , Sulfonamides/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/chemistry , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Binding Sites , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/chemistry , Carbazoles/chemistry , Carbazoles/metabolism , Carbazoles/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/chemistry , Electrophysiological Phenomena/drug effects , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Isoquinolines/chemistry , Isoquinolines/pharmacology , Male , Molecular Docking Simulation , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/physiology , Patch-Clamp Techniques , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrroles/chemistry , Pyrroles/metabolism , Pyrroles/pharmacology , Rats, Wistar , Sulfonamides/chemistry , Sulfonamides/pharmacology , Tail , Vasodilation/drug effects
8.
Eur J Pharmacol ; 796: 158-174, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-28012974

ABSTRACT

Ion channels underlie a wide variety of physiological processes that involve rapid changes in cell dynamics, such as cardiac and vascular smooth muscle contraction. Overexpression or dysfunction of these membrane proteins are the basis of many cardiovascular diseases that represent the leading cause of morbidity and mortality for human beings. In the last few years, flavonoids, widely distributed in the plant kingdom, have attracted the interest of many laboratories as an emerging class of fine ion, in particular Cav, channels modulators. Pieces of in vitro evidence for direct as well as indirect effects exerted by various flavonoids on ion channel currents are now accumulating in the scientific literature. This activity may be responsible, at least in part, for the beneficial and protective effects of dietary flavonoids toward cardiovascular diseases highlighted in several epidemiological studies. Here we examine numerous studies aimed at analysing this feature of flavonoids, focusing on the mechanisms that promote their sometimes controversial activities at cardiovascular Cav channels. New methodological approaches, such as molecular modelling and docking to Cav1.2 channel α1c subunit, used to elucidate flavonoids intrinsic mechanism of action, are introduced. Moreover, flavonoid-membrane interaction, bioavailability, and antioxidant activity are taken into account and discussed.


Subject(s)
Calcium Channels/metabolism , Flavonoids/pharmacology , Heart/drug effects , Myocardium/metabolism , Animals , Biological Availability , Calcium Channels/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Flavonoids/metabolism , Flavonoids/pharmacokinetics , Humans , Myocardium/cytology
9.
Curr Med Chem ; 23(25): 2836-2873, 2016.
Article in English | MEDLINE | ID: mdl-27281297

ABSTRACT

Mycobacterium tuberculosis (M. tuberculosis), an almost genetically monomorphic pathogen is a human parasite, transmitted mostly by humans and causes tuberculosis (TB). TB is firmly associated to poverty, although lack of proper nutrition and lowered immune status are contributing factors for disease development. TB remains second only to HIV/AIDS as the leading cause of mortality worldwide due to a single infectious agent and is responsible for nearly 1.5 million deaths annually. Some steps of the progress of our knowledge of M. tuberculosis physiology and its interactions with human beings, are reviewed here. This progress has provided fertile ground for improving diagnosis and cure of TB infection. For TB diagnostics laboratories in high-burden countries, primary isolation is the first step before performing drug susceptibility testing (DST) of M. tuberculosis. IGRA (interferon-γ release assay)-based tests for diagnosis of active TB are sufficiently fast, specific and sensitive to allow to contain infection and distinguish among latent TB infection and BCG vaccination individuals from those who have clinically resolved M. tuberculosis infection after anti-TB treatment.


Subject(s)
Tuberculosis/diagnosis , Antigens, Bacterial/metabolism , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Bacterial Proteins/metabolism , Carboxylic Ester Hydrolases/metabolism , Cell Wall/chemistry , Cell Wall/metabolism , Drug Resistance, Multiple, Bacterial , Humans , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/isolation & purification , Mycobacterium tuberculosis/metabolism , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Phagosomes/metabolism , Tuberculosis/drug therapy , Tuberculosis/epidemiology , Tuberculosis/microbiology , Virulence Factors/metabolism
10.
Curr Med Chem ; 23(21): 2245-74, 2016.
Article in English | MEDLINE | ID: mdl-27142291

ABSTRACT

Ineffectively treated tuberculosis (TB) is associated with substantial morbidity and mortality. Cure of TB patients is hampered by the development of multidrug resistance in M. tuberculosis and the need of long-term treatment. The diarylquinoline derivative bedaquiline was approved in December 2012 under the accelerated-approval regulations of FDA as part of a combination therapy for treating adults with pulmonary MDR-TB for whom effective cures are not otherwise available. The bicyclic nitroimidazoles delamanid and its companion pretomanid inhibit mycolic acid synthesis via an unknown mechanism. In November 2013, delamanid received conditional approval by the European Medicines Agency for MDR-TB treatment. Use of both drugs, however, is limited owing to toxicity issues. If the aim to reduce treatment duration is pursued in order to limit costs and improve patient adherence, it is mandatory to demonstrate their noninferiority with fewer months of therapy. In three phase III clinical trials the efficacy of the most recent fluoroquinolones, gatifloxacin and moxifloxacin, has been investigated in a four-month treatment regimen of drug-susceptible TB. In all three studies, after two months the culture conversion rates of observed sputum indicated that fluoroquinolone-based therapies were likely to be superior. However, this feature did not reliably predict sterilizing activity or a risk of relapse. In other words, the shortened treatments were not noninferior to standard treatments. To counteract mycobacterial survival strategies and reduce the timelength of treatment with anti-TB drugs, other novel and powerful agents, as well as tuberculosis vaccines, are under intense clinical investigation for safety and efficacy assessment.


Subject(s)
Antitubercular Agents/therapeutic use , Tuberculosis/drug therapy , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Clinical Trials as Topic , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacology , Fluoroquinolones/therapeutic use , Gatifloxacin , Humans , Moxifloxacin , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Prospective Studies , Tuberculosis, Multidrug-Resistant/drug therapy
11.
Curr Med Chem ; 23(12): 1186-216, 2016.
Article in English | MEDLINE | ID: mdl-27048337

ABSTRACT

The great progress of knowledge of both M. tuberculosis physiology and how human host and bacilli interact has provided fertile ground for improving diagnosis and cure of TB infection. Once M. tuberculosis has infected humans, it elaborates strategies for evading the risk to killing by the cells of the host immune system and by the anti-tuberculosis (anti-TB) agents employed to cure infection. These strategies give rise to a bacterial multidrug resistance (MDR) status. This stems firstly from genetic mutations targeting a constellation of drug-processing mechanisms that still need full identification, as drug efflux pumps and drug activating/ inactivating enzymes (genetic resistance). Secondly, from the bacterial adaptation to stressful environmental conditions by adopting a temporary dormancy state lasting for decades and characterized by indifference to anti-TB drugs (phenotypic resistance or tolerance). The clarification of the strategies elaborated for surviving by M. tuberculosis has brought to the identification in the last few years of a number of mycobacterial molecular targets worth to exploitation for the development of novel and powerful anti-TB drugs. These targets include drug-efflux pump systems, considered partly responsible for genetic multi-drug resistance, and several enzymes and pump systems, as well, that sustain the metabolic adaptations of M. tuberculosis in the host and give rise to its phenotypic drug resistance.


Subject(s)
Antitubercular Agents/pharmacology , Tuberculosis/genetics , Drug Design , Drug Resistance, Multiple, Bacterial/drug effects , Humans , Mycobacterium tuberculosis/drug effects , Phenotype , Tuberculosis/drug therapy
12.
Biochem Pharmacol ; 104: 42-51, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26807479

ABSTRACT

Rhodamine 123 (R123) transport substrate sensitizes P-glycoprotein (P-gp) to inhibition by compound 2c (cis-cis) N,N-bis(cyclohexanolamine)aryl ester isomer in a concentration-dependent manner in human MDR1-gene transfected mouse T-lymphoma L5178 cells as shown previously. By contrast, epirubicin (EPI) concentration changes left unaltered 2c IC50 values of EPI efflux. To clarify this discrepancy, defined molecular docking (DMD) analyses of 12 N,N-bis(cyclohexanolamine)aryl esters, the highly flexible aryl ester analog 4, and several P-gp substrate/non-substrate inhibitors were performed on human P-gp drug- or nucleotide-binding domains (DBD or NBD). DMD measurements yielded lowest binding energy (LBE, kcal/mol) values (mean ± SD) ranging from -11.8 ± 0.54 (valspodar) to -3.98 ± 0.01 (4). Lys234, Ser952 and Tyr953 residues formed H-bonds with most of the compounds. Only 2c docked also at ATP binding site (LBE value of -6.9 ± 0.30 kcal/mol). Inhibition of P-gp-mediated R123 efflux by 12 N,N-bis(cyclohexanolamine)aryl esters and 4 significantly correlated with LBE values. DMD analysis of EPI, (3)H-1EPI, (3)H-2EPI, (14)C-1EPI, (14)C-2EPI, R123 and 2c before and after previous docking of each of them indicated that pre-docking of either 2c or EPI significantly reduced LBE of both EPI and R123, and that of both (3)H-2EPI and (14)C-2EPI, respectively. Since the clusters of DBD amino acid residues interacting with EPI were different, if EPI docked alone or after pre-docking of EPI or 2c, the existence of alternative secondary binding site for EPI on P-gp is credible. In conclusion, 2c may allocate the drug-binding pocket and reduce strong binding of EPI and R123 in agreement with P-gp inhibition experiments, where 2c reduced efflux of EPI and R123.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cyclohexanols/metabolism , Epirubicin/metabolism , Molecular Docking Simulation , Rhodamine 123/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Binding Sites , Cell Culture Techniques , Cell Line, Tumor , Cyclohexanols/chemistry , Epirubicin/chemistry , Esters , Mice , Molecular Structure , Protein Binding , Protein Transport , Rhodamine 123/chemistry , Substrate Specificity , Transfection
13.
Vascul Pharmacol ; 76: 53-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586313

ABSTRACT

Papaverine is an opium alkaloid, primarily used as an antispasmodic drug and as a cerebral and coronary vasodilator. Its phosphodiesterase inhibitory activity promotes increase of cAMP levels mainly in the cytosol. As cAMP is known to modulate L-type Ca(2+) channel activity, here we tested the proposition that papaverine could affect vascular channel function. An in-depth analysis of the effect of papaverine on Ba(2+) or Ca(2+) current through L-type Ca(2+) channel [IBa(L) or ICa(L)], performed in rat tail artery myocytes using either the whole-cell or the perforated patch-clamp method, was accompanied by a functional study on rat aorta rings. Papaverine increased current amplitude under both the perforated or whole-cell configuration. Stimulation of the current by papaverine was concentration-, Vh-, frequency-, and charge carrier-dependent, and fully reverted by drug washout. The PKA inhibitor H89, but not the PKG inhibitor Rp-8-Br-cGMPS, antagonised papaverine- as well as IBMX- (another phosphodiesterase inhibitor) induced IBa(L) stimulation. In cells pre-treated with IBMX, application of papaverine failed to increase current amplitude. Papaverine sped up the inactivation kinetics of IBa(L), though only at concentrations ≥ 30 µM, and shifted the voltage dependence of the inactivation curve to more negative potentials. In rings, the vasorelaxing activity of papaverine was enhanced by previous treatment with nifedipine. In conclusion, papaverine stimulates vascular L-type Ca(2+) channel via a PKA-dependent mechanism, thus antagonising its main vasodilating activity.


Subject(s)
Calcium Channels, L-Type/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Papaverine/pharmacology , Vasodilator Agents/pharmacology , Animals , Arteries/drug effects , Arteries/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nifedipine/pharmacology , Rats
14.
Planta Med ; 81(4): 298-304, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25714723

ABSTRACT

The sesquiterpene zerumbone, isolated from the rhizome of Zingiber zerumbet Sm., besides its widespread use as a food flavouring and appetiser, is also recommended in traditional medicine for the treatment of several ailments. It has attracted great attention recently for its effective chemopreventive and therapeutic effects observed in various models of cancer. To assess the zerumbone safety profile, a pharmacology study designed to flag any potential adverse effect on vasculature was performed. Zerumbone was tested for vasorelaxing activity on rat aorta rings and for L-type Ba(2+) current blocking activity on single myocytes isolated from the rat-tail artery. The spasmolytic effect of zerumbone was more marked on rings stimulated with 60 mM than with 30 mM K(+) (IC50 values of 16 µM and 102 µM, respectively). In the presence of 60 mM K(+), zerumbone concentration-dependently inhibited the contraction induced by the cumulative additions of Ca(2+), this inhibition being inversely related to the Ca(2+) concentration. Phenylephrine-induced contraction was inhibited by the drug, though less efficiently and independently of the presence of an intact endothelium, without affecting Ca(2+) release from the intracellular stores. Zerumbone inhibited the L-type Ba(2+) current (estimated IC50 value of 458.7 µM) and accelerated the kinetics of current decay. In conclusion, zerumbone showed an overall weak in vitro vasodilating activity, partly attributable to the blocking of the L-type Ca(2+) channel, which does not seem to represent, however, a serious threat to its widespread use.


Subject(s)
Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Plant Extracts/pharmacology , Sesquiterpenes/pharmacology , Vasoconstriction/drug effects , Vasodilation/drug effects , Zingiberaceae/chemistry , Animals , Calcium/metabolism , Calcium Channels/metabolism , In Vitro Techniques , Male , Muscle, Smooth, Vascular/physiology , Parasympatholytics/adverse effects , Parasympatholytics/pharmacology , Phenylephrine , Plant Extracts/adverse effects , Rats, Wistar , Rhizome , Sesquiterpenes/adverse effects
15.
Phytother Res ; 29(4): 628-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25581420

ABSTRACT

Chronically administered organic nitrates induce nitrate tolerance and endothelial dysfunction, which limit their therapeutic use. eNOS uncoupling, ROS over-production, aldehyde dehydrogenase-2 as well as superoxide dismutase (SOD) oxidative inhibition, and cGMP desensitization are thought to play an important role. Natural polyphenols are effective antioxidants, which might counteract the mechanisms leading to nitrate tolerance. The aim of this work was to verify whether freeze-dried (dealcoholized) red wine (FDRW) was able to revert glyceryl trinitrate (GTN) tolerance and endothelial dysfunction induced in rat aorta rings with either GTN or diethyldithiocarbamate (DETCA), an irreversible inhibitor of Cu/Zn SOD. GTN induced a concentration-dependent relaxation of rings pre-contracted with phenylephrine. GTN spasmolysis was significantly reduced in rings pre-incubated with either GTN or DETCA. FDRW, at 2.8 µg of gallic acid equivalents (GAE)/mL concentration, was able to revert partially, though significantly, GTN-induced tolerance but not tolerance and endothelial dysfunction induced by DETCA. This work provides the first evidence in vitro that red wine components, at concentrations comparable to those achieved in human blood after moderate consumption of red wine, revert tolerance to nitrates with a mechanism possibly mediated by SOD.


Subject(s)
Drug Tolerance , Endothelium, Vascular/drug effects , Nitroglycerin/pharmacology , Polyphenols/pharmacology , Wine , Animals , Antioxidants/pharmacology , Aorta/drug effects , Endothelium, Vascular/physiopathology , In Vitro Techniques , Male , Rats , Rats, Wistar , Superoxide Dismutase/antagonists & inhibitors
16.
J Nat Prod ; 77(7): 1586-93, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24949913

ABSTRACT

In the search for novel natural compounds endowed with potential antihypertensive activity, a new sulfur-containing indole alkaloid, N-demethylglypetelotine (2), and its known analogue glypetelotine (1), were isolated from the leaves of Glycosmis petelotii. Their structures were established on the basis of spectroscopic evidence. The two alkaloids were assessed for vasorelaxing activity on rat aorta rings and for L-type Ba(2+) current [I(Ba(L))] blocking activity on single myocytes isolated from rat tail artery. Both glypetelotine and N-demethylglypetelotine inhibited phenylephrine-induced contraction with IC50 values of 20 and 50 µM, respectively. The presence of endothelium did not modify their spasmolytic effect. Neither glypetelotine nor N-demethylglypetelotine affected Ca(2+) release from the sarcoplasmic reticulum induced by phenylephrine. The spasmolytic effect of glypetelotine increased with membrane depolarization. In the presence of 60 mM K(+), both compounds inhibited, in a concentration-dependent manner, the contraction induced by cumulative addition of Ca(2+), this inhibition being inversely related to Ca(2+) concentration. Glypetelotine and, less efficiently N-demethylglypetelotine, inhibited I(Ba(L)), the former compound also affecting I(Ba(L)) kinetics. In conclusion, glypetelotine is a novel vasorelaxing agent which antagonizes L-type Ca(2+) channels.


Subject(s)
Calcium Channel Blockers/isolation & purification , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Indole Alkaloids/isolation & purification , Indole Alkaloids/pharmacology , Muscle, Smooth, Vascular/drug effects , Rutaceae/chemistry , Sulfur Compounds/isolation & purification , Sulfur Compounds/pharmacology , Vasodilator Agents/isolation & purification , Vasodilator Agents/pharmacology , Algorithms , Animals , Aorta/drug effects , Calcium Channel Blockers/chemistry , Indole Alkaloids/chemistry , Inhibitory Concentration 50 , Male , Molecular Structure , Muscle Contraction/drug effects , Parasympatholytics/chemistry , Parasympatholytics/isolation & purification , Parasympatholytics/pharmacology , Phenylephrine/pharmacology , Rats , Sulfur Compounds/chemistry , Vasodilation/drug effects , Vasodilator Agents/chemistry , Vietnam
17.
Can J Physiol Pharmacol ; 92(2): 171-4, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24502641

ABSTRACT

The effect of freeze-dried red wine (FDRW) on cardiac function and electrocardiogram (ECG) in Langendorff-isolated rat hearts was investigated. FDRW significantly decreased left ventricular pressure and coronary perfusion pressure, the latter being dependent on the activation of both phosphatidylinositol 3-kinase and eNOS. FDRW did not affect the QRS and QT interval in the ECG, although at 56 µg of gallic acid equivalents/mL, it prolonged PQ interval and induced a second-degree atrioventricular block in 3 out of 6 hearts. This is the first study demonstrating that at concentrations resembling a moderate consumption of red wine, FDRW exhibited negative inotropic and coronary vasodilating activity leaving unaltered ECG, whereas at very high concentrations, it induced arrhythmogenic effects.


Subject(s)
Cardiotonic Agents/pharmacology , Heart/physiology , Wine , Animals , Electrocardiography , Freeze Drying , Heart Rate , Male , Perfusion , Rats , Rats, Sprague-Dawley
18.
Eur J Pharmacol ; 714(1-3): 178-87, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23751511

ABSTRACT

The neuroprotective agent riluzole [2-amino-6-(trifluoromethoxy)benzothiazole] has been shown to antagonize neuronal high-voltage activated Ca(2+) currents. In the search for novel scaffolds leading to potential antihypertensive agents, a series of 2-aryl- and 2-amido-benzothiazoles (HUP) were assessed for their vasorelaxing property on rat aorta rings and for their L-type Ba(2+) currents [I(Ba(L))] blocking activity on single myocytes isolated from the rat tail artery. HUP5 and HUP30, the most potent of the series, inhibited phenylephrine-induced contraction with IC50 values in the range 3-6 µM. The presence of endothelium did not modify their spasmolytic activity. Both HUP5 and HUP30 increased tissue levels of cGMP and shifted to the left the concentration-response curve to sodium nitroprusside. In rings precontracted by phenylephrine, tetraethylammonium or 1H-[1,2,4] oxadiazolo [4,3-a] quinoxalin-1-one (ODQ) shifted to the right the concentration-relaxation curves of HUP5 and HUP30. The antispasmodic effect of HUP5 and HUP30 was more marked on rings stimulated with 25/30 mM than with 60 mM K(+). HUP5 and HUP30 antagonized both extracellular Ca(2+) influx and Ca(2+) mobilization from intracellular stores in response to phenylephrine: this effect was not modified by the presence of ODQ. I(Ba(L)) was partly inhibited by HUP5 and blocked by HUP30 in a concentration-dependent as well as ODQ-independent manner. In conclusion, HUP5 and HUP30 are vasorelaxing agents that stimulate soluble guanylyl cyclase, activate K(+) channels, and block extracellular Ca(2+) influx. The present benzothiazole derivatives form a novel class of multifunctional vasodilators which may give rise to effective antihypertensive agents.


Subject(s)
Arteries/drug effects , Arteries/physiology , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacology , Animals , Arteries/cytology , Arteries/metabolism , Calcium/metabolism , Cyclic GMP/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , In Vitro Techniques , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Muscle Cells/drug effects , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Wistar , Tetraethylammonium/pharmacology , Vasoconstriction/drug effects , Vasodilation/drug effects
19.
Planta Med ; 79(6): 465-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23479387

ABSTRACT

Chronic use of glyceryl trinitrate is limited by serious side effects, inter alia tolerance and endothelial dysfunction of coronary and resistance arteries. The natural flavonoid quercetin has been shown to counteract the development of glyceryl trinitrate tolerance in vitro. Two mitochondriotropic, 4-O-triphenylphosphoniumbutyl derivatives of quercetin (QTA-3BTPI and Q-3BTPI) were compared to quercetin for protection against glyceryl trinitrate-induced tolerance and endothelial dysfunction of isolated rat aorta rings. Both QTA-3BTPI and Q-3BTPI significantly counteracted the reduced vascular responsiveness to both glyceryl trinitrate and acetylcholine caused by prolonged exposure of the vessel to glyceryl trinitrate itself, their potency being much greater than that of quercetin. QTA-3BTPI, however, turned out to cause endothelial dysfunction per se. Since Q-3BTPI antagonized in vitro nitrate tolerance and endothelial dysfunction of vessels, this encourages assessing whether this effect is displayed also in vivo during long-term glyceryl trinitrate treatment.


Subject(s)
Endothelium, Vascular/drug effects , Nitroglycerin/toxicity , Quercetin/analogs & derivatives , Acetylcholine/pharmacology , Animals , Aorta/drug effects , Endothelium, Vascular/physiology , In Vitro Techniques , Oxidative Stress , Quercetin/pharmacology , Rats
20.
Pharmacol Res ; 66(6): 555-63, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23036353

ABSTRACT

Cannabinoid CB2 receptor activation has been shown to have many pharmacological but not psychotropic effects. The aim of this study was to investigate the potential protection of brain tissues afforded by the novel substituted 4-quinolone-3-carboxylic acid derivative COR167, a selective CB2 agonist, toward ischemia and reperfusion-induced injury, as well as the mechanism of this potential effect. Rat brain cortical slices subjected to oxygen and glucose deprivation (OGD) followed by re-oxygenation were used. Cell damage was quantified by measuring at the end of the reperfusion phase the release into the artificial cerebrospinal fluid (ACSF) of lactate dehydrogenase (LDH), glutamate, IL-6 and TNF-α and by evaluating in tissue the lipid-peroxides (thiobarbituric acid-reactive substances, TBARS), the free, reduced glutathione content (GSH) and the water gain (TWG), taken as an index of cell swelling. COR167 (10nM or 100 nM), added to ACSF during the entire reperfusion phase, markedly reduced LDH and glutamate release, as well as TWG. Lower (0.1-1 nM) or higher concentrations (1,000 nM) were ineffective, suggesting thereby an hormetic behavior. COR167 at 10nM concentration markedly reverted in tissues TBARS increase and GSH decrease, while reducing IL-6 and TNF-α release into ACSF. COR167 effects on glutamate and LDH release were abrogated by the selective CB2 inverse-agonists COR170 (1 nM) and AM630 (1µM) but not by the CB1 antagonist AM251 (1 µM). COR170 as well as AM630 per se were able to revert TWG. The CB2 receptor agonist COR167 potently protected rat brain cortical slices against OGD and reperfusion injury, partly through CB2 receptors activation.


Subject(s)
Adamantane/analogs & derivatives , Cannabinoid Receptor Agonists/pharmacology , Cerebral Cortex/drug effects , Glucose/deficiency , Oxygen/metabolism , Quinolones/pharmacology , Receptor, Cannabinoid, CB2/agonists , Reperfusion Injury/prevention & control , Adamantane/chemistry , Adamantane/pharmacology , Animals , Cannabinoid Receptor Agonists/chemistry , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Glutamic Acid/metabolism , Interleukin-6/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Molecular Structure , Quinolones/chemistry , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Tissue Culture Techniques , Tumor Necrosis Factor-alpha/metabolism , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...