Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Med Chem ; 59(14): 6838-47, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27390919

ABSTRACT

Protein arginine methyltransferases (PRMTs) represent an emerging target class in oncology and other disease areas. So far, the most successful strategy to identify PRMT inhibitors has been to screen large to medium-size chemical libraries. Attempts to develop PRMT inhibitors using receptor-based computational methods have met limited success. Here, using virtual screening approaches, we identify 11 CARM1 (PRMT4) inhibitors with ligand efficiencies ranging from 0.28 to 0.84. CARM1 selective hits were further validated by orthogonal methods. Two structure-based rounds of optimization produced 27 (SGC2085), a CARM1 inhibitor with an IC50 of 50 nM and more than hundred-fold selectivity over other PRMTs. These results indicate that virtual screening strategies can be successfully applied to Rossmann-fold protein methyltransferases.


Subject(s)
CARD Signaling Adaptor Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , CARD Signaling Adaptor Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
2.
Am J Physiol Lung Cell Mol Physiol ; 309(6): L537-42, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26254425

ABSTRACT

Exposure to moderate hyperoxia in prematurity contributes to subsequent airway dysfunction and increases the risk of developing recurrent wheeze and asthma. The nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic GMP (cGMP) axis modulates airway tone by regulating airway smooth muscle (ASM) intracellular Ca(2+) ([Ca(2+)]i) and contractility. However, the effects of hyperoxia on this axis in the context of Ca(2+)/contractility are not known. In developing human ASM, we explored the effects of novel drugs that activate sGC independent of NO on alleviating hyperoxia (50% oxygen)-induced enhancement of Ca(2+) responses to bronchoconstrictor agonists. Treatment with BAY 41-2272 (sGC stimulator) and BAY 60-2770 (sGC activator) increased cGMP levels during exposure to 50% O2. Although 50% O2 did not alter sGCα1 or sGCß1 expression, BAY 60-2770 did increase sGCß1 expression. BAY 41-2272 and BAY 60-2770 blunted Ca(2+) responses to histamine in cells exposed to 50% O2. The effects of BAY 41-2272 and BAY 60-2770 were reversed by protein kinase G inhibition. These novel data demonstrate that BAY 41-2272 and BAY 60-2770 stimulate production of cGMP and blunt hyperoxia-induced increases in Ca(2+) responses in developing ASM. Accordingly, sGC stimulators/activators may be a useful therapeutic strategy in improving bronchodilation in preterm infants.


Subject(s)
Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Hydrocarbons, Fluorinated/pharmacology , Hyperoxia/drug therapy , Myocytes, Smooth Muscle/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Bronchi/pathology , Calcium Signaling , Cells, Cultured , Cyclic GMP/metabolism , Drug Evaluation, Preclinical , Guanylate Cyclase/metabolism , Humans , Hyperoxia/enzymology , Muscle, Smooth/drug effects , Muscle, Smooth/embryology , Muscle, Smooth/pathology , Myocytes, Smooth Muscle/drug effects , Oxygen/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Trachea/pathology
3.
World Neurosurg ; 84(4): 954-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26093361

ABSTRACT

OBJECTIVE: Decreased 3'-5'-cyclic adenosine monophosphate (cAMP), protein kinase A (PKA), and increased N-methyl-d-aspartate (NMDA) related apoptosis were observed in traumatic brain injury (TBI). It is of interest to examine the effect of magnesium lithospermate B (MLB) on cAMP/PKA pathway and NMDAR in TBI. METHODS: A rodent weight-drop TBI model was used. Administration of MLB was initiated 1 week before (precondition) and 24 hours later (reversal). Cortical homogenates were harvested to measure cAMP (enzyme-linked immunosorbent assay), soluble guanylyl cyclases, PKA and NMDA receptor-2ß (Western blot). In addition, cAMP kinase antagonist and H-89 dihydrochloride hydrate were used to test MLB's effect on the cytoplasm cAMP/PKA pathway after TBI. RESULTS: Morphologically, vacuolated neuron and activated microglia were observed in the TBI groups but absent in the MLB preconditioning and healthy controls. Induced cAMP, soluble guanylyl cyclase α1, and PKA were observed in the MLB groups, when compared with the TBI group (P < 0.01) Administration of H-89 dihydrochloride hydrate reversed the effect of MLB on cortical PKA and NMDA-2ß expression after TBI. CONCLUSIONS: This study showed that MLB exerted an antioxidant effect on the enhancement of cytoplasm cAMP and PKA. This compound also decreased NMDA-2ß levels, which may correspond to its neuroprotective effects. This finding lends credence to the presumption that MLB modulates the NMDA-2ß neurotoxicity through a cAMP-dependent mechanism in the pathogenesis of TBI.


Subject(s)
Brain Injuries/pathology , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP , Drugs, Chinese Herbal/pharmacology , Free Radical Scavengers/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Signal Transduction/drug effects , Animals , Behavior, Animal , Brain Edema/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Isoquinolines/pharmacology , Male , Nervous System Diseases/etiology , Nervous System Diseases/psychology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Soluble Guanylyl Cyclase , Sulfonamides/pharmacology
4.
Ann Emerg Med ; 65(4): 410-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25441767

ABSTRACT

STUDY OBJECTIVE: Calcium channel blocker poisonings account for a substantial number of reported deaths from cardiovascular drugs. Although supportive care is the mainstay of treatment, experimental therapies such as high-dose insulin-euglycemia and lipid emulsion have been studied in animal models and used in humans. In the most severe cases, even aggressive care is inadequate and deaths occur. In both experimental models and clinical cases of vasodilatory shock, methylene blue improves hemodynamic measures. It acts as a nitric oxide scavenger and inhibits guanylate cyclase that is responsible for the production of cyclic guanosine monophosphate (cGMP). Excessive cGMP production is associated with refractory vasodilatory shock in sepsis and anaphylaxis. The aim of this study is to determine the efficacy of methylene blue in an animal model of amlodipine-induced shock. METHODS: Sprague-Dawley rats were anesthetized, ventilated, and instrumented for continuous blood pressure and pulse rate monitoring. The dose of amlodipine that produced death within 60 minutes was 17 mg/kg per hour (LD50). Rats were divided into 2 groups: amlodipine followed by methylene blue or amlodipine followed by normal saline solution, with 15 rats in each group. Rats received methylene blue at 2 mg/kg during 5 minutes or an equivalent amount of normal saline solution in 3 intervals from the start of the protocol: minutes 5, 30, and 60. The animals were observed for a total of 2 hours after the start of the protocol. Mortality risk and survival time were analyzed with Fisher's exact test and Kaplan-Meier survival analysis with the log rank test. RESULTS: Overall, 1 of 15 rats (7%) in the saline solution-treated group survived to 120 minutes compared with 5 of 15 (33%) in the methylene blue-treated group (difference -26%; 95% confidence interval [CI] -54% to 0.3%). The median survival time for the normal saline solution group was 42 minutes (95% CI 28.1 to 55.9 minutes); for the methylene blue group, 109 minutes (95% CI 93.9 to 124.1 minutes). Pulse rate and mean arterial pressure (MAP) differences between groups were analyzed until 60 minutes. Pulse rate was significantly higher in the methylene blue-treated group beginning 25 minutes after the start of the amlodipine infusion (95% CI 30 to 113 minutes) that was analyzed until 60 minutes. MAP was significantly higher in the methylene blue-treated group starting 25 minutes after the amlodipine infusion (95% CI 2 to 30 minutes) that was analyzed until 60 minutes. CONCLUSION: Methylene blue did not result in a significant difference in mortality risk. There was an increased pulse rate, MAP, and median survival time in the methylene blue group.


Subject(s)
Calcium Channel Blockers/poisoning , Free Radical Scavengers/therapeutic use , Methylene Blue/therapeutic use , Shock/chemically induced , Amlodipine/poisoning , Animals , Disease Models, Animal , Guanylate Cyclase/antagonists & inhibitors , Rats, Sprague-Dawley
5.
Am J Respir Cell Mol Biol ; 52(6): 762-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25353067

ABSTRACT

Inspiratory resistive breathing (RB), encountered in obstructive lung diseases, induces lung injury. The soluble guanylyl cyclase (sGC)/cyclic guanosine monophosphate (cGMP) pathway is down-regulated in chronic and acute animal models of RB, such as asthma, chronic obstructive pulmonary disease, and in endotoxin-induced acute lung injury. Our objectives were to: (1) characterize the effects of increased concurrent inspiratory and expiratory resistance in mice via tracheal banding; and (2) investigate the contribution of the sGC/cGMP pathway in RB-induced lung injury. Anesthetized C57BL/6 mice underwent RB achieved by restricting tracheal surface area to 50% (tracheal banding). RB for 24 hours resulted in increased bronchoalveolar lavage fluid cellularity and protein content, marked leukocyte infiltration in the lungs, and perturbed respiratory mechanics (increased tissue resistance and elasticity, shifted static pressure-volume curve right and downwards, decreased static compliance), consistent with the presence of acute lung injury. RB down-regulated sGC expression in the lung. All manifestations of lung injury caused by RB were exacerbated by the administration of the sGC inhibitor, 1H-[1,2,4]oxodiazolo[4,3-]quinoxalin-l-one, or when RB was performed using sGCα1 knockout mice. Conversely, restoration of sGC signaling by prior administration of the sGC activator BAY 58-2667 (Bayer, Leverkusen, Germany) prevented RB-induced lung injury. Strikingly, direct pharmacological activation of sGC with BAY 58-2667 24 hours after RB reversed, within 6 hours, the established lung injury. These findings raise the possibility that pharmacological targeting of the sGC-cGMP axis could be used to ameliorate lung dysfunction in obstructive lung diseases.


Subject(s)
Guanylate Cyclase/metabolism , Lung Diseases, Obstructive/enzymology , Lung Injury/enzymology , Airway Resistance , Animals , Benzoates/pharmacology , Benzoates/therapeutic use , Cyclic GMP/metabolism , Drug Evaluation, Preclinical , Enzyme Activation , Guanylate Cyclase/antagonists & inhibitors , Lung Diseases, Obstructive/drug therapy , Lung Injury/drug therapy , Male , Mice, Inbred C57BL
6.
J Med Food ; 17(7): 742-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24971771

ABSTRACT

The aim of this study was to clarify the efficacy of procyanidin C1 (Pro C1) for modulating vascular tone. Pro C1 induced a potent vasorelaxant effect on phenylephrine-constricted endothelium-intact thoracic aortic rings, but had no effect on denuded thoracic aortic rings. Moreover, Pro C1 caused a significant increase in nitric oxide (NO) production in endothelial cells. Pro C1-induced vasorelaxation and Pro C1-induced NO production were significantly decreased in the presence of a nonspecific potassium channel blocker (tetraethylammonium chloride [TEA]), an endothelial NO synthase inhibitor (N(G)-monomethyl-L-arginine [L-NMMA]), and a store-operated calcium entry inhibitor (2-aminoethyl diphenylborinate [2-APB]). Pro C1-induced vasorelaxation was also completely abolished by an inhibitor of soluble guanyl cyclase, which suggests that the Pro C1 effects observed involved cyclic guanosine monophosphate (cGMP) production. Interestingly, Pro C1 significantly enhanced basal cGMP levels. Taken together, these results indicate that Pro C1-induced vasorelaxation is associated with the activation of the calcium-dependent NO/cGMP pathway, involving potassium channel activation. Thus, Pro C1 may represent a novel and potentially therapeutically relevant compound for the treatment of cardiovascular diseases.


Subject(s)
Biflavonoids/pharmacology , Catechin/pharmacology , Cyclic GMP/metabolism , Nitric Oxide/metabolism , Proanthocyanidins/pharmacology , Signal Transduction/drug effects , Vasodilation/drug effects , Animals , Aorta, Thoracic/drug effects , Boron Compounds/pharmacology , Cells, Cultured , Endothelial Cells/drug effects , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , In Vitro Techniques , Male , Nitric Oxide Synthase Type III/metabolism , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Tetraethylammonium/pharmacology , Vasodilator Agents/pharmacology , omega-N-Methylarginine/pharmacology
7.
Biomed Res Int ; 2014: 728019, 2014.
Article in English | MEDLINE | ID: mdl-24868545

ABSTRACT

Amarogentin, an active principle of Gentiana lutea, possess antitumorigenic, antidiabetic, and antioxidative properties. Activation of platelets is associated with intravascular thrombosis and cardiovascular diseases. The present study examined the effects of amarogentin on platelet activation. Amarogentin treatment (15~60 µM) inhibited platelet aggregation induced by collagen, but not thrombin, arachidonic acid, and U46619. Amarogentin inhibited collagen-induced phosphorylation of phospholipase C (PLC) γ2, protein kinase C (PKC), and mitogen-activated protein kinases (MAPKs). It also inhibits in vivo thrombus formation in mice. In addition, neither the guanylate cyclase inhibitor ODQ nor the adenylate cyclase inhibitor SQ22536 affected the amarogentin-mediated inhibition of platelet aggregation, which suggests that amarogentin does not regulate the levels of cyclic AMP and cyclic GMP. In conclusion, amarogentin prevents platelet activation through the inhibition of PLC γ2-PKC cascade and MAPK pathway. Our findings suggest that amarogentin may offer therapeutic potential for preventing or treating thromboembolic disorders.


Subject(s)
Iridoids/chemistry , MAP Kinase Signaling System , Phospholipase C gamma/metabolism , Platelet Activation/drug effects , Protein Kinase C/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/chemistry , Adenine/analogs & derivatives , Adenine/chemistry , Adenosine Triphosphate/chemistry , Animals , Arachidonic Acid/chemistry , Collagen/chemistry , Cyclic GMP/metabolism , Guanylate Cyclase/antagonists & inhibitors , Humans , Mice , Oxadiazoles/chemistry , Plant Extracts/chemistry , Quinoxalines/chemistry , Thrombin/chemistry , Thromboembolism/drug therapy , Thromboembolism/prevention & control , Thrombosis/drug therapy
8.
Phytomedicine ; 21(6): 787-92, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24680617

ABSTRACT

Tetra-acetylajugasterone C (TAAC) was found to be one of the naturally occurring compounds of the Cameroonian medicinal plant Vitex cienkowskii which is responsible for a vasorelaxant activity of an extract of this plant. The evaluation of the underlying mechanisms for the relaxing effect of TAAC was determined using aortic rings of rats and mice. TAAC produced a concentration-dependent relaxation in rat artery rings pre-contracted with 1µM noradrenaline (IC50: 8.40µM) or 60mM KCl (IC50: 36.30µM). The nitric oxide synthase inhibitor l-NAME (100µM) and the soluble guanylate cyclase inhibitor ODQ (10µM) significantly attenuated the vasodilatory effect of TAAC. TAAC also exerted a relaxing effect in aorta of wild-type mice (cGKI(+/+); IC50=13.04µM) but a weaker effect in aorta of mice lacking cGMP-dependent protein kinase I (cGKI(-/-); IC50=36.12µM). The involvement of calcium channels was studied in rings pre-incubated in calcium-free buffer and primed with 1µM noradrenaline prior to addition of calcium to elicit contraction. TAAC (100µM) completely inhibited the resulting calcium-induced vasoconstriction. The same concentration of TAAC showed a stronger effect on the tonic than on the phasic component of noradrenaline-induced contraction. This study shows that TAAC, a newly detected constituent of Vitex cienkowskii contributes to the relaxing effect of an extract of the plant. The effect is partially mediated by the involvement of the NO/cGMP pathway of the smooth muscle but additionally inhibition of calcium influx into the cell may play a role.


Subject(s)
Ecdysterone/analogs & derivatives , Endothelium, Vascular/drug effects , Plant Extracts/pharmacology , Vasoconstriction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Vitex/chemistry , Animals , Aorta/drug effects , Calcium/metabolism , Calcium Channels/metabolism , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Ecdysterone/isolation & purification , Ecdysterone/pharmacology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Norepinephrine/pharmacology , Plant Bark , Plant Extracts/chemistry , Plant Stems , Rats , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Soluble Guanylyl Cyclase , Vasodilator Agents/isolation & purification
9.
Psychopharmacology (Berl) ; 231(4): 663-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24101156

ABSTRACT

RATIONALE: Nitric oxide (NO) modulates the dopamine uptake and release processes and appears to be implicated in dopamine-related pathologies, such as schizophrenia. However, it is unclear whether there is excess or deficient NO synthesis in schizophrenia pathophysiology. Analyses of the intracellular pathways downstream of NO system activation have identified the cyclic nucleotide cyclic guanosine monophosphate (cGMP) as a possible target for drug development. Defects in the sensorimotor gating of the neural mechanism underlying the integration and processing of sensory information have been detected across species through prepulse inhibition (PPI). OBJECTIVES: The aim of this study was to investigate the effects of NO/cGMP increase on sensorimotor gating modulation during dopamine hyperfunction. METHODS: Mice were treated with NO donors and subjected to the PPI test. Treatment with the NO donor sodium nitroprusside was preceded by pretreatment with a soluble guanylate cyclase (sGC) inhibitor. Additionally, the mice were treated with NO donors and phosphodiesterases inhibitors prior to amphetamine treatment. RESULTS: Pretreatment with the NO donors enhanced the PPI response and attenuated the amphetamine-disruptive effects on the PPI. The sGC inhibitor did not modify the sodium nitroprusside effects. Additionally, the cGMP increase induced by a specific phosphodiesterase inhibitor did not modify the amphetamine-disruptive effect. CONCLUSIONS: This study provides the first demonstration that an increase in NO can improve the PPI response and block the amphetamine-disruptive effects on the PPI response. Our data are consistent with recent clinical results. However, these effects do not appear to be related to an increase in cGMP levels, and further investigation is thus required.


Subject(s)
Cyclic GMP/metabolism , Inhibition, Psychological , Nitric Oxide/metabolism , Reflex, Startle/physiology , Sensory Gating/physiology , Acoustic Stimulation , Amphetamine/pharmacology , Animals , Cyclic AMP/metabolism , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Male , Mice , Neuropsychological Tests , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Purinones/pharmacology , Reflex, Startle/drug effects , Sensory Gating/drug effects
10.
Phytomedicine ; 20(14): 1241-6, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23880329

ABSTRACT

Current work was conducted to evaluate the vasorelaxant effect of dihydrospinochalcone-A (1) and isocordoin (2), compounds type chalcone isolated from Lonchocarpus xuul, an endemic tree of the Yucatan Peninsula, Mexico. Compounds 1 and 2 were found to induce significant relaxant effect in a concentration-dependent manner on aortic rat rings pre-contracted with noradrenaline (NA, 0.1 µM). Compound 1 was the most active and its effect was endothelium-dependent (Emax=79.67% and EC50=21.46 µM with endothelium and Emax=23.58% and EC50=91.8 µM without endothelium, respectively). The functional mechanism of action for 1 was elucidated. Pre-incubation with L-NAME (unspecific nitric oxide synthase inhibitor), indomethacin (unspecific COX inhibitor), ODQ (soluble guanylyl cyclase inhibitor), atropine (cholinergic receptor antagonist), TEA (unspecific potassium channel blocker) reduced relaxations induced by 1. Oral administration of 50 mg/kg of compound 1 exhibited significant decrease in diastolic and systolic blood pressure in SHR rats. The heart rate was not modified. Compound 1 was docked with a crystal structure of eNOS. Dihydrospinochalcone-A showed calculated affinity with eNOS in the C1 binding pockets, near the catalytic site; Trp449, Trp447 and His373 through aromatic and π-π interactions, also His463 and Arg367 are the residues that make hydrogen bonds with the carbonyl and hydroxyl groups. In conclusion, dihydrospinochalcone-A induces a significant antihypertensive effect due to its direct vasorelaxant action on rat aorta rings, through NO/sCG/PKG pathway and potassium channel opening.


Subject(s)
Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Catechols/pharmacology , Chalcones/pharmacology , Fabaceae/chemistry , Nitric Oxide/biosynthesis , Vasodilator Agents/pharmacology , Animals , Antihypertensive Agents/isolation & purification , Aorta, Thoracic/drug effects , Atropine/pharmacology , Catechols/isolation & purification , Chalcones/isolation & purification , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Guanylate Cyclase/antagonists & inhibitors , Heart Rate/drug effects , Indomethacin/pharmacology , Mexico , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/metabolism , Norepinephrine , Plant Extracts/chemistry , Plant Extracts/pharmacology , Potassium Channel Blockers/pharmacology , Protein Binding , Rats , Rats, Inbred SHR , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Soluble Guanylyl Cyclase , Vasoconstriction/drug effects , Vasodilator Agents/isolation & purification
11.
Phytother Res ; 27(12): 1784-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23401173

ABSTRACT

Cubebin, the most abundant lignan in Piper cubeba, has been described as having several effects as trypanocidal, antimycobacterial, antispasmodic, antimicrobial, anti-inflammatory, and analgesic. This study investigated the vasorelaxant effect produced by (-)-cubebin in isolated rat aortic rings pre-contracted with phenylephrine (Phe), and the possible mechanism involved in this event was evaluated. Endothelium-dependent relaxation was evoked by acetylcholine and (-)-cubebin in intact aortic rings, while endothelium-independent vasorelaxation was elicited by sodium nitroprusside and (-)-cubebin in denuded rings. Cumulative concentration-response curves for Phe (10(-10) -10(-5) M) were determined for endothelium-intact and endothelium-denuded aortic rings in either the presence or absence of (-)-cubebin. Dose-response curves were also constructed for pre-incubation of vascular rings with Nω-nitro-L-arginine methyl ester (L-NAME) (a non-specific nitric oxide synthase inhibitor), indomethacin (an unspecific cyclooxygenase inhibitor), and 1H-[1,2,4] oxadiazolo [4,3-a]quinoxalin-1-one (ODQ) (a guanylyl cyclase inhibitor). (-)-Cubebin was found to exert a vasorelaxant effect irrespective of the presence of endothelium, which was abolished by pretreatment with L-NAME and ODQ, but not with indomethacin. In addition, (-)-cubebin was able to reduce Phe contraction in the case of intact rings. These results suggest that (-)-cubebin promotes vasorelaxation via NO/cGMP pathway in rat aorta, without prostacyclin involvement.


Subject(s)
Aorta/drug effects , Endothelium, Vascular/drug effects , Lignans/pharmacology , Nitric Oxide/physiology , Vasodilation/drug effects , Acetylcholine/pharmacology , Animals , Aorta/physiology , Cyclic GMP/physiology , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , In Vitro Techniques , Indomethacin/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Piper/chemistry , Quinoxalines/pharmacology , Rats , Rats, Wistar
12.
Phytomedicine ; 20(5): 387-93, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23369344

ABSTRACT

Fructus Alpiniae Zerumbet (FAZ) is an herb widely used to treat vascular disorders in Guizhou province, China, the essential oil has been identified as one of it vasodilation effect active components, and especial, the composition was significantly difference from the leaves. Vasodilation effects and mechanism of essential oil from FAZ (EOFAZ) were investigated. The EOFAZ showed significant vasodilation effect on endothelium-with rat thoracic aortic rings preincubated with norepinephrine (NE, 1.0µM) or KCl (60mM) in a concentration-dependent manner (1.14-72.96µg/ml). The non-selective nitric oxide synthase inhibitor l-NAME, as well as the soluble guanylate cyclase inhibitor MB, attenuated the relaxation of EOFAZ in endothelium-intact rat thoracic aortic rings. However, there were not significantly affected the vasodilation effects pretreated with cyclooxygenase inhibition by indomethacin (Indo) or ß-noradrenergic inhibition by propranolol (Prop). The present results first demonstrated that vasodilation effect of EOFAZ depending upon the endothelium and concentration, and the mechanism involvement of NOS-cGMP system. In contrast, prostacyclin and ß-adrenoceptor may not be associated with EOFAZ-induced vasorelaxation.


Subject(s)
Aorta, Thoracic/drug effects , Endothelium-Dependent Relaxing Factors/pharmacology , Oils, Volatile/pharmacology , Zingiberaceae/chemistry , Animals , Drug Evaluation, Preclinical , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Endothelium, Vascular/drug effects , Endothelium-Dependent Relaxing Factors/chemistry , Endothelium-Dependent Relaxing Factors/isolation & purification , Guanylate Cyclase/antagonists & inhibitors , In Vitro Techniques , Indomethacin/pharmacology , Male , Muscle Relaxation , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Oils, Volatile/chemistry , Oils, Volatile/isolation & purification , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley
13.
Eur J Pharmacol ; 697(1-3): 81-7, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23051676

ABSTRACT

Salviae miltiorrhizae, a traditional Chinese medicine, is widely used in the treatment of cardiovascular and cerebrovascular diseases. Salvianolic acid B is identified as one of the most important water-soluble active ingredients in Salviae miltiorrhizae and associated with the activation of Ca(2+) channel of cytomembrane. But the further mechanism of action was not very clearly. In our study, we investigated the vasodilation activity of salvianolic acid B using the isolated thoracic aortic rings from Japanese white rabbit. Salvianolic acid B significantly released the contraction of the isolated thoracic aortic rings induced by phenylephrine and CaCl(2) while had no effects on the aortic rings with KCl stimulated. Different with Di-ao-xin-xue-kang capsule, salvianolic acid B caused an increase of Ca(2+) in cytoplasm from not only activation of Ca(2+) channel in cytomembrane but also release of endogenous Ca(2+). Then, a series of endogenous Ca(2+) inhibitors were pretreated to explore the mechanism of salvianolic acid B, and the results provided further evidences that salvianolic acid B causes intracellular calcium release in ryanodine receptors-dependent manners. Moreover, combining l-arginine (l-Arg) with salvianolic acid B promoted the vasodilation activity suggesting a relationship with nitric oxide (NO). To further investigated its mechanism, both guanylate cyclase (GC) inhibitor and NO Synthase inhibitor were used and demonstrated to block vasodilation activity of the aortic rings. Our findings reveal a NO-sGC-cGMP signals dependence mechanism of salvianolic acid B on its vasodilation activity which provide an evidence for its subsequent application in clinic.


Subject(s)
Aorta, Thoracic/drug effects , Benzofurans/pharmacology , Nitric Oxide/metabolism , Signal Transduction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/metabolism , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rabbits , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Soluble Guanylyl Cyclase , Vasoconstrictor Agents/pharmacology
14.
Eur J Heart Fail ; 14(8): 946-53, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22719060

ABSTRACT

AIMS: Pulmonary hypertension (PH) due to systolic left ventricular dysfunction (PH-sLVD) frequently complicates heart failure (HF), and greatly worsens the prognosis of patients with sLVD, but as yet has no approved treatment. The LEPHT study aims to characterize the haemodynamic profile, safety, tolerability, and pharmacokinetic profile of riociguat (BAY 63-2521), an oral stimulator of soluble guanylate cyclase, in patients with PH-sLVD. METHODS AND RESULTS: This 16-week, phase IIb, randomized, placebo-controlled, double-blind study enrols patients with PH-sLVD, defined as left ventricular ejection fraction (LVEF) ≤40% and mean pulmonary arterial pressure (PAP(mean)) ≥25 mmHg at rest. Patients using optimized HF medication will receive placebo or riociguat 0.5 mg, 1 mg, or up to 2 mg three times daily. The dose will be titrated for 8 weeks, based on systolic blood pressure and well-being, followed by 8 weeks of treatment at a stable dose. The primary efficacy variable is PAP(mean), while secondary efficacy endpoints include LVEF, exercise capacity, quality of life, and other haemodynamic and echocardiographic measurements. Safety and pharmacokinetics will also be assessed. After the 16-week study, patients will have the opportunity to be treated with riociguat in a long-term extension phase. CONCLUSION: The LEPHT study will provide valuable information on the haemodynamic, echocardiographic, and preliminary clinical effects of riociguat in patients with PH-sLVD. Trial registration NCT01065454.


Subject(s)
Heart Failure, Systolic/drug therapy , Hypertension, Pulmonary/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Research Design , Ventricular Dysfunction, Left/complications , Dose-Response Relationship, Drug , Double-Blind Method , Exercise Tolerance/drug effects , Exercise Tolerance/physiology , Guanylate Cyclase/antagonists & inhibitors , Heart Failure, Systolic/etiology , Hemodynamics/drug effects , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Prognosis , Quality of Life/psychology , Stroke Volume , Treatment Outcome , Ventricular Function, Left
15.
J Nat Med ; 66(1): 17-24, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21607822

ABSTRACT

The aqueous extract of Lespedeza cuneata G. Don. (ALC) induced vasorelaxation of phenylephrine precontracted aorta in a dose-dependent manner. This effect disappeared in the absence of functional endothelium. Pretreatment of the aortic tissues with N(G)-nitro-L-arginine methyl ester (L-NAME), or 1H-[1,2,4]-oxadiazole-[4,3-α]-quinoxalin-1-one (ODQ) blocked ALC-induced vascular relaxation. Incubation of endothelium-intact thoracic aortic rings with ALC increased cGMP production. ALC-induced cGMP production was blocked by pretreatment with L-NAME or ODQ. ALC-induced vascular relaxation was also markedly attenuated by addition of verapamil or diltiazem, but was not blocked by pretreatment with indomethacine, glibenclamide, tetraethylammonium, atropine, or propranolol. The results suggest that ALC dilates vascular smooth muscle via endothelium-dependent NO-cGMP signaling.


Subject(s)
Aorta, Thoracic/drug effects , Cyclic GMP/metabolism , Lespedeza , Nitric Oxide/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Aorta, Thoracic/metabolism , Calcium Channel Blockers/pharmacology , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Lespedeza/chemistry , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Plant Extracts/isolation & purification , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Vasodilator Agents/isolation & purification
16.
Clin Exp Pharmacol Physiol ; 39(1): 63-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22032308

ABSTRACT

1. Tanshinone IIA, one of the active components of the Radix of Salvia miltiorrhiza, is used in traditional Chinese medicine to treat cardiovascular diseases. However, the intracellular mechanism of action of tanshinone IIA remain to be determined. The aims of the present study were to test the hypothesis that tanshinone IIA alters strain-induced endothelin (ET)-1 expression and nitric oxide (NO) production, as well as to identify the putative signalling pathways involved, in human umbilical vein endothelial cells (HUVEC). 2. Cultured HUVEC were exposed to cyclic strain in the presence of 1-10 µmol/L tanshinone IIA. Expression of ET-1 was examined by reverse transcription-polymerase chain reaction and ELISA. Phosphorylation of endothelial NO synthase (eNOS) and activating transcription factor (ATF) 3 was assessed by western blot analysis. 3. Tanshinone IIA (3 and 10 µmol/L) inhibited strain-induced ET-1 expression. In contrast, NO production, eNOS phosphorylation and ATF3 expression were enhanced by tanshinone IIA. The eNOS inhibitor N(G) -nitro-L-arginine methyl ester (l-NAME; 100 µmol/L), the phosphatidylinositol 3-kinase inhibitor LY294002 (5 µmol/L) and the soluble guanylyl cyclase inhibitor 1H-[1,2,4] oxadiazolo [4,3-a] quinoxalin-1-one (ODQ; 10 µmol/L) inhibited tanshinone IIA-induced increases in ATF3 expression. Moreover, treatment of HUVEC with either an NO donor (3,3-bis [aminoethyl]-1-hydroxy-2-oxo-1-triazene; 500 µmol/L) or an ATF3 activator (carbobenzoxy-L-leucyl-L-leucyl-L-leucinal; 5 µmol/L) resulted in the repression of strain-induced ET-1 expression. The inhibitory effect of tanshinone IIA on strain-induced ET-1 expression was significantly attenuated by l-NAME, ODQ and the transfection of small interfering RNA for ATF3. 4. In conclusion, tanshinone IIA inhibits strain-induced ET-1 expression by increasing NO and upregulating ATF3 in HUVEC. The present study provides important new insights into the molecular pathways that may contribute to the beneficial effects of tanshinone IIA in the cardiovascular system.


Subject(s)
Abietanes/pharmacology , Cardiovascular Diseases/prevention & control , Cellular Microenvironment , Down-Regulation/drug effects , Endothelin-1/metabolism , Endothelium, Vascular/drug effects , Activating Transcription Factor 3/agonists , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cells, Cultured , Endothelin-1/genetics , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Signal Transduction/drug effects , Soluble Guanylyl Cyclase
17.
Pharmacology ; 88(1-2): 65-71, 2011.
Article in English | MEDLINE | ID: mdl-21846997

ABSTRACT

Peroxisome proliferator-activated receptor α (PPAR-α) is a ligand-activated transcription factor that exerts strong effects on metabolic pathways. Our aim was to elucidate the effect of clofibrate, a PPAR-α agonist, on the longitudinal muscle of the mouse distal colon. We initially found that clofibrate induced a relaxation response in this muscle. Notably, the PPAR-α antagonists GW9662 and T0070907 did not attenuate this clofibrate-induced relaxation. The structurally related PPAR-α agonists fenofibrate and bezafibrate induced relaxation in the distal colon as effectively as clofibrate. In contrast, wy-14643, which activates PPAR-α more selectively than clofibrate, had no effect. Furthermore, clofibrate-induced relaxation was not affected by N-nitro-L-arginine, an NO synthase inhibitor, 1H-[1,2,4]-oxadiazolo-[4,3- a]quinoxaline-1-one, a soluble guanylate cyclase inhibitor, or H89, a protein kinase A inhibitor. Tetrodotoxin, an Na⁺ channel blocker, and glibenclamide, apamin, charybdotoxin and XE991, various K⁺ channel blockers, had no effect on clofibrate-induced relaxation. Importantly, clofibrate induced a relaxation response that was not accompanied by any alteration in the cytoplasmic Ca²âº concentration in the longitudinal muscle of the mouse distal colon. Moreover, calyculin A, a myosin light-chain phosphatase (MLCP) inhibitor, attenuated clofibrate-induced relaxation. Our findings indicate that clofibrate relaxes the longitudinal smooth muscle of the mouse distal colon by regulating MLCP activity.


Subject(s)
Anticholesteremic Agents/pharmacology , Calcium/physiology , Clofibrate/pharmacology , Colon/physiology , Muscle Relaxation/physiology , Muscle, Smooth/physiology , Anilides/pharmacology , Animals , Anticholesteremic Agents/metabolism , Benzamides/pharmacology , Clofibrate/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Disease Models, Animal , Drug Evaluation, Preclinical , Guanylate Cyclase/antagonists & inhibitors , Male , Marine Toxins , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Oxazoles/pharmacology , PPAR alpha/agonists , PPAR alpha/metabolism , Potassium Channel Blockers/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Sodium Channel Blockers/pharmacology , Soluble Guanylyl Cyclase
18.
Biochemistry ; 50(36): 7787-99, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21823650

ABSTRACT

Nitric oxide (NO) regulates cardiovascular hemostasis by binding to soluble guanylyl cyclase (sGC), leading to cGMP production, reduced cytosolic calcium concentration ([Ca(2+)](i)), and vasorelaxation. Thrombospondin-1 (TSP-1), a secreted matricellular protein, was recently discovered to inhibit NO signaling and sGC activity. Inhibition of sGC requires binding to cell-surface receptor CD47. Here, we show that a TSP-1 C-terminal fragment (E3CaG1) readily inhibits sGC in Jurkat T cells and that inhibition requires an increase in [Ca(2+)](i). Using flow cytometry, we show that E3CaG1 binds directly to CD47 on the surface of Jurkat T cells. Using digital imaging microscopy on live cells, we further show that E3CaG1 binding results in a substantial increase in [Ca(2+)](i), up to 300 nM. Addition of angiotensin II, a potent vasoconstrictor known to increase [Ca(2+)](i), also strongly inhibits sGC activity. sGC isolated from calcium-treated cells or from cell-free lysates supplemented with Ca(2+) remains inhibited, while addition of kinase inhibitor staurosporine prevents inhibition, indicating inhibition is likely due to phosphorylation. Inhibition is through an increase in K(m) for GTP, which rises to 834 µM for the NO-stimulated protein, a 13-fold increase over the uninhibited protein. Compounds YC-1 and BAY 41-2272, allosteric stimulators of sGC that are of interest for treating hypertension, overcome E3CaG1-mediated inhibition of NO-ligated sGC. Taken together, these data suggest that sGC not only lowers [Ca(2+)](i) in response to NO, inducing vasodilation, but also is inhibited by high [Ca(2+)](i), providing a fine balance between signals for vasodilation and vasoconstriction.


Subject(s)
Angiotensin II/pharmacology , Calcium/metabolism , Guanylate Cyclase/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Thrombospondin 1/pharmacology , Vasoconstrictor Agents/pharmacology , CD47 Antigen , Cells, Cultured , Flow Cytometry , Guanylate Cyclase/metabolism , Humans , Jurkat Cells , Kinetics , Nitric Oxide/metabolism , Phosphorylation , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Vasoconstriction/drug effects
19.
J Ethnopharmacol ; 137(1): 547-52, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21704693

ABSTRACT

AIM OF THE STUDY: Sophora flavescens (SF) is a known medicinal herb for the treatment of cardiovascular symptoms associated with arrhythmia in China. However, the pharmacological action mechanisms involved have not been well studied. The aim of the present study was to define effects of roots of SF on the vascular tension and responsible mechanisms in rat thoracic aorta. MATERIALS AND METHODS: Ethanol extract of the roots of SF (ESF) was examined for their vascular relaxant effect in isolated phenylephrine-precontracted rat thoracic aorta. RESULTS: ESF (0.1-100 µg/ml) induced relaxation of the phenylephrine-precontracted aortic rings in a concentration-dependent manner. Endothelium-denudation abolished the ESF-induced vasorelaxation. Pretreatment of the endothelium-intact aortic rings with l-NAME, an inhibitor of nitric oxide synthase, and ODQ, an inhibitor of soluble guanylyl cyclase (sGC), inhibited ESF-induced vasorelaxation. ESF increased cGMP levels of the aortic rings in a concentration-dependent manner and the effect was blocked by l-NAME and ODQ. Inhibition of K(+) channels with glibenclamide and tetraethylammonium, cyclooxygenase inhibition with indomethacin, and ß-adrenergic and muscarinic receptors blockade had no effect on the ESF-induced vasorelaxation. CONCLUSION: These findings suggest that ESF relaxes vascular smooth muscle via endothelium-dependent NO-sGC-cGMP signaling pathway.


Subject(s)
Aorta, Thoracic/drug effects , Drugs, Chinese Herbal/pharmacology , Endothelium, Vascular/drug effects , Ethanol/chemistry , Solvents/chemistry , Sophora , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Aorta, Thoracic/metabolism , Cyclic GMP/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Male , Muscarinic Antagonists/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Plant Roots , Plants, Medicinal , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Sophora/chemistry , Vasodilator Agents/chemistry , Vasodilator Agents/isolation & purification
20.
J Ethnopharmacol ; 137(1): 184-91, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21600275

ABSTRACT

AIMS OF THE STUDY: The ethanolic extract of Kaempferia parviflora (KPE) has been reported to contain a range of flavonoids and to enhance endothelial synthesis of NO. We investigated the vascular relaxant, antioxidant and cardioprotective activities of KPE. MATERIALS AND METHODS: Vascular function was assessed in rat aortic rings and superoxide generation determined using lucigenin enhanced chemiluminescence. Ischaemia and reperfusion were induced in rat isolated, perfused hearts. RESULTS: KPE caused vasorelaxation (R(max) 102 ± 2%), which was partly inhibited by removal of the endothelium (R(max) 91 ± 1%) or by N(G)-nitro-l-arginine (L-NNA, R(max) 83 ± 3%) or 1H-[1,2,4] oxadiazolo[4,3-a]quinoxaline-1-one (ODQ, R(max) 80 ± 2%). In addition KPE caused concentration-dependent inhibition of the contractile response to exogenous Ca(2+). KPE (10(-3)M) also significantly inhibited superoxide radical generation induced by of xanthine/xanthine oxidase (2.3 ± 0.4% of control) to a similar extent to the xanthine oxidase inhibitor allopurinol (10(-4)M, 1.6 ± 0.5%) or by rat isolated aorta in the presence of NADPH (30.0 ± 6.3% of control) similarly to the NADPH oxidase inhibitor diphenyliodonium (5 × 10(-6)M, 23.1 ± 5.6%). In the presence of oxidant stress generated by pyrogallol endothelium-dependent relaxation of rat aortic rings was impaired (ACh R(max) control 99 ± 1%; pyrogallol 44 ± 5%), an effect that was significantly reduced by KPE (10(-4)M, ACh R(max) 82 ± 4%). In addition, KPE was found to attenuate the ventricular dysfunction caused by 20 min global ischaemia and 30 min reperfusion (I/R) in rat isolated hearts (dP/dt IR 1016 ± 242, IR+KPE 2238±233 mm Hg/s). CONCLUSION: KPE is an effective vasodilator and antioxidant that is able to prevent myocardial ischaemia-reperfusion injury. We suggest that KPE may be useful as an adjunct to thrombolytic therapy in the management of reperfusion injury.


Subject(s)
Antioxidants/pharmacology , Ethanol/chemistry , Myocardial Reperfusion Injury/prevention & control , Plant Exudates/pharmacology , Solvents/chemistry , Vasodilator Agents/pharmacology , Zingiberaceae , Animals , Antioxidants/chemistry , Antioxidants/isolation & purification , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , In Vitro Techniques , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Perfusion , Plant Exudates/chemistry , Plant Exudates/isolation & purification , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Superoxides/metabolism , Vasodilation/drug effects , Vasodilator Agents/chemistry , Vasodilator Agents/isolation & purification , Ventricular Function, Left/drug effects , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism , Zingiberaceae/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL