Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Int J Mol Sci ; 21(7)2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32268492

ABSTRACT

We have previously reported that argan oil and argan press-cake from the kernels of Argania spinosa have an anti-melanogenesis effect. Here, the effect of argan fruit shell ethanol extract (AFSEE) on melanogenesis in B16F10 cells was determined, and the mechanism underlying its effect was elucidated. The proliferation of AFSEE-treated B16F10 cells was evaluated using the 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) assay, while the melanin content was quantified using a spectrophotometric method. The expression of melanogenesis-related proteins was determined by Western blot and real-time PCR, while global gene expression was determined using a DNA microarray. In vitro analysis results showed that the melanin content of B16F10 cells was significantly increased by AFSEE, without cytotoxicity, by increasing the melanogenic enzyme tyrosinase (TRY), tyrosinase related-protein 1 (TRP1), and dopachrome tautomerase (DCT) protein and mRNA expression, as well as upregulating microphthalmia-associated transcription factor (MITF) expression through mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase (ERK) and p38, and the cyclic adenosine monophosphate (cAMP) signaling pathway, as indicated by the microarray analysis results. AFSEE's melanogenesis promotion effect is primarily attributed to its polyphenolic components. In conclusion, AFSEE promotes melanogenesis in B16F10 cells by upregulating the expression of the melanogenic enzymes through the cAMP-MITF signaling pathway.AFSEE may be used as a cosmetics product component to promote melanogenesis, or as a therapeutic against hypopigmentation disorders.


Subject(s)
Cyclic AMP/metabolism , Fruit/chemistry , Melanins/biosynthesis , Plant Extracts/chemistry , Plant Extracts/pharmacology , Protein Biosynthesis/drug effects , Sapotaceae/chemistry , Second Messenger Systems/drug effects , Animals , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System , Melanoma, Experimental , Mice , Phosphorylation , Phytochemicals/chemistry , Phytochemicals/pharmacology
2.
ACS Chem Neurosci ; 10(12): 4787-4799, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31697472

ABSTRACT

Previously, we designed, synthesized, and evaluated a series of quinolone-benzofuran derivatives as multitargeted anti-Alzheimer's disease (anti-AD) compounds, and we discovered that WBQ5187 possesses superior anti-AD bioactivity. In this work, we investigated the pharmacokinetics of this new molecule, as well as its therapeutic efficacy in restoring cognition and neuropathology, in the APP/PS1 mouse model of AD. Pharmacokinetic analyses demonstrated that WBQ5187 possessed rational oral bioavailability, metabolic stability, and excellent blood-brain barrier (BBB) permeability. Pharmacodynamics studies indicated that a 12-week treatment with the lead compound at doses of 40 mg/kg or higher significantly enhanced the learning and memory performance of the APP/PS1 transgenic mice, and the effect was more potent than that of clioquinol (CQ). Furthermore, WBQ5187 notably reduced cerebral ß-amyloid pathology, gliosis, and neuronal cell loss and increased the levels of cAMP in the hippocampus of these mice. The surrogate measures of emesis indicated that WBQ5187 had no effect at its cognitive effective doses. Overall, our results demonstrated that this compound markedly improves cognitive and spatial memory functions in AD mice and represents a promising pharmaceutical agent with potential for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Benzofurans/therapeutic use , Brain Chemistry/drug effects , Clioquinol/analogs & derivatives , Neuroprotective Agents/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use , Resorcinols/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Anesthetics, General/toxicity , Animals , Benzofurans/chemistry , Benzofurans/pharmacokinetics , Biological Availability , Blood-Brain Barrier , Clioquinol/chemistry , Clioquinol/pharmacokinetics , Clioquinol/therapeutic use , Cyclic AMP/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Gliosis/drug therapy , Gliosis/prevention & control , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nausea/chemically induced , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacokinetics , Phosphodiesterase 4 Inhibitors/toxicity , Resorcinols/chemistry , Resorcinols/pharmacokinetics , Second Messenger Systems/drug effects , Vomiting/chemically induced
3.
Biomed Pharmacother ; 106: 1091-1097, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119175

ABSTRACT

Ginsenoside Re (GS-Re) is one of the main ingredients of ginseng, a widely known Chinese traditional medicine, and has a variety of beneficial effects, including vasorelaxation, antioxidative, anti-inflammatory, and anticancer properties. The aims of the present study were to observe the effect of GS-Re on balloon injury-induced neointimal hyperplasia in the arteries and to investigate the mechanisms underlying this effect. A rat vascular neointimal hyperplasia model was generated by rubbing the endothelium of the common carotid artery (CCA) with a balloon, and GS-Re (12.5, 25 or 50 mg/kg/d) were subsequently continuously administered to the rats by gavage for 14 days. After GS-Re treatment, the vessel lumen of injured vessels showed significant increases in the GS-Re 25.0 and 50.0 mg/kg/d (intermediate- and high-dose) groups according to H.E. staining. Additionally, a reduced percentage of proliferating cell nuclear antigen (PCNA)-positive cells and an increased number of SM α-actin-positive cells were detected, and the levels of NO, cyclic guanosine monophosphate (cGMP), and eNOS mRNA as well as the phos-eNOSser1177/eNOS protein ratio were obviously upregulated in the intermediate- and high-dose groups. Moreover, the promotive effects of GS-Re on NO and eNOS expression were blocked by L-NAME treatment to different degrees. These results suggested that GS-Re can suppress balloon injury-induced vascular neointimal hyperplasia by inhibiting VSMC proliferation, which is closely related to the activation of the eNOS/NO/cGMP pathway.


Subject(s)
Angioplasty, Balloon/instrumentation , Carotid Artery Injuries/prevention & control , Carotid Artery, Common/drug effects , Cyclic GMP/metabolism , Ginsenosides/pharmacology , Neointima , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Actins/metabolism , Animals , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/pathology , Carotid Artery, Common/enzymology , Carotid Artery, Common/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Hyperplasia , Male , Nitric Oxide Synthase Type III/genetics , Proliferating Cell Nuclear Antigen/metabolism , Rats, Sprague-Dawley , Second Messenger Systems/drug effects
4.
J Ethnopharmacol ; 193: 423-432, 2016 Dec 04.
Article in English | MEDLINE | ID: mdl-27660009

ABSTRACT

BACKGROUND: Kai-xin-san (KXS), composed of Ginseng Radix et Rhizoma, Polygalae Radix, Acori Tatarinowii Rhizoma and Poria, is a famous Chinese medicinal formula applied for treating stress-related psychiatric disease with the symptoms such as depression, forgetfulness and dizziness. Dependent on the symptom differentiation of patients, the composition ratio of KXS was varied and one ratio of 3:2:2:3 was widely applied. However, its molecular mechanism has seldom been investigated. PURPOSE: We aimed to reveal the action mechanism of KXS on anti-depression on synaptic protein regulation in both in vivo and in vitro models. STUDY DESIGN/METHODS: Firstly, the anti-depression effect of KXS was evaluated on a chronic mild stress induced depressive animal model and the mRNA expressions of various synaptic proteins in hippocampus of the depressive rat brains were determined. Then, KXS with different ratios as well as single herb were further evaluated on rat primary cultured hippocampus neurons and the possible signaling pathway was explored. RESULTS: Intra-gastric administration of a chemically standardized KXS for only 6h significantly alleviated the CUMS-induced depressive symptoms displayed by enhanced sucrose consumption and this effect was maintained after daily treatment for seven days. Simultaneously, the mRNA expressions of various synaptic proteins in hippocampus were regulated. Among these synaptic proteins, synaptotagmin (pre-synaptic marker) and post synaptic density protein (post-synaptic marker), with the higher altered magnitude on animal model, were further evaluated on rat primary cultured hippocampus neurons. After neuronal cultures treated with three ratios of KXS at the early and late stages of its life episode, the expression levels of synaptotagmin and PSD95 were both enhanced dramatically via stimulating cAMP dependent pathway. However, different ratio exerted different efficacy. The ratio with higher amounts of Ginseng Radix et Rhizoma and Polygalae Radix showed better effect in early life episode while higher amounts of Acori Tatarinowii Rhizoma and Poria behaved better in late life episode. The contribution of single herb on expressions of synaptic proteins was also evaluated. CONCLUSIONS: KXS was beneficial for synaptogenesis by inducing synaptic protein expressions, which might account for its anti-depression effect.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Drugs, Chinese Herbal/pharmacology , Hippocampus/drug effects , Nerve Tissue Proteins/metabolism , Stress, Psychological/complications , Synapses/drug effects , Animals , Cells, Cultured , Chronic Disease , Cyclic AMP/metabolism , Depression/metabolism , Depression/physiopathology , Depression/psychology , Dietary Sucrose/administration & dosage , Disease Models, Animal , Feeding Behavior/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Nerve Tissue Proteins/genetics , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Second Messenger Systems/drug effects , Synapses/metabolism , Time Factors , Up-Regulation
5.
Mol Nutr Food Res ; 59(8): 1443-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25943029

ABSTRACT

SCOPE: Resveratrol (RSV), a natural polyphenol, has been reported to attenuate nonalcoholic fatty liver disease (NAFLD); however, its underlying mechanism is unclear. Autophagy was recently identified as a critical protective mechanism during NAFLD development. Therefore, we investigated the role of autophagy in the beneficial effects of RSV on hepatic steatosis. METHODS AND RESULTS: Via Oil red O staining, triglyceride, and ß-hydroxybutyrate detection, we found that RSV decreased palmitate-induced lipid accumulation and stimulated fatty acid ß-oxidation in hepatocytes. Based on Western blot assay, confocal microscopy and transmission electron microscopy, we found that RSV induced autophagy in hepatocytes, whereas autophagy inhibition markedly abolished RSV-mediated hepatic steatosis improvement. Moreover, RSV increased cAMP levels and the levels of SIRT1 (sirtuin 1), pPRKA (phosphorylated protein kinase A), and pAMPK (phosphorylated AMP-activated protein kinase), as well as SIRT1 activity in HepG2 cells. Incubation with inhibitors of AC (adenylyl cyclase), PRKA, AMPK, SIRT1, or with AC, PRKA, AMPK, or SIRT1 siRNA abolished RSV-mediated autophagy. Similar results were obtained in mice with hepatic steatosis. CONCLUSION: RSV improved hepatic steatosis partially by inducing autophagy via the cAMP-PRKA-AMPK-SIRT1 signaling pathway, which provides new evidence regarding RSV's effects on NAFLD treatment.


Subject(s)
Antioxidants/therapeutic use , Autophagy , Cyclic AMP/agonists , Dietary Supplements , Liver/metabolism , Non-alcoholic Fatty Liver Disease/diet therapy , Second Messenger Systems , Stilbenes/therapeutic use , Adenylyl Cyclases/chemistry , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Antioxidants/metabolism , Autophagy/drug effects , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acids, Nonesterified/adverse effects , Fatty Acids, Nonesterified/antagonists & inhibitors , Fatty Acids, Nonesterified/metabolism , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Mice, 129 Strain , Microscopy, Electron, Transmission , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , RNA Interference , Resveratrol , Second Messenger Systems/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/chemistry , Sirtuin 1/genetics , Sirtuin 1/metabolism , Stilbenes/metabolism
6.
J Pediatr Surg ; 49(1): 39-45; discussion 45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24439578

ABSTRACT

PURPOSE: Pulmonary hypertension (pHTN), a main determinant of survival in congenital diaphragmatic hernia (CDH), results from in utero vascular remodeling. Phosphodiesterase type 5 (PDE5) inhibitors have never been used antenatally to treat pHTN. The purpose of this study is to determine if antenatal PDE5 inhibitors can prevent pHTN in the fetal lamb model of CDH. METHODS: CDH was created in pregnant ewes. Postoperatively, pregnant ewes received oral placebo or tadalafil, a PDE5 inhibitor, until delivery. Near term gestation, lambs underwent resuscitations, and lung tissue was snap frozen for protein analysis. RESULTS: Mean cGMP levels were 0.53±0.11 in placebo-treated fetal lambs and 1.73±0.21 in tadalafil-treated fetal lambs (p=0.002). Normalized expression of eNOS was 82%±12% in Normal-Placebo, 61%±5% in CDH-Placebo, 116%±6% in Normal-Tadalafil, and 86%±8% in CDH-Tadalafil lambs. Normalized expression of ß-sGC was 105%±15% in Normal-Placebo, 82%±3% in CDH-Placebo, 158%±16% in Normal-Tadalafil, and 86%±8% in CDH-Tadalafil lambs. Endothelial NOS and ß-sGC were significantly decreased in CDH (p=0.0007 and 0.01 for eNOS and ß-sGC, respectively), and tadalafil significantly increased eNOS expression (p=0.0002). CONCLUSIONS: PDE5 inhibitors can cross the placental barrier. ß-sGC and eNOS are downregulated in fetal lambs with CDH. Antenatal PDE5 inhibitors normalize eNOS and may prevent in utero vascular remodeling in CDH.


Subject(s)
Carbolines/therapeutic use , Fetal Diseases/drug therapy , Fetal Therapies , Hernias, Diaphragmatic, Congenital , Nitric Oxide Synthase Type III/biosynthesis , Phosphodiesterase 5 Inhibitors/therapeutic use , Animals , Carbolines/administration & dosage , Carbolines/pharmacology , Cyclic GMP/analysis , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Female , Hernia, Diaphragmatic/complications , Hernia, Diaphragmatic/embryology , Hernia, Diaphragmatic/enzymology , Hernia, Diaphragmatic/prevention & control , Hypertension, Pulmonary/embryology , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/embryology , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/etiology , Lung/chemistry , Lung/drug effects , Lung/embryology , Lung/pathology , Maternal-Fetal Exchange , Nitric Oxide Synthase Type III/genetics , Organ Size/drug effects , Phosphodiesterase 5 Inhibitors/administration & dosage , Phosphodiesterase 5 Inhibitors/pharmacology , Pregnancy , Random Allocation , Second Messenger Systems/drug effects , Sheep , Tadalafil
7.
J Assoc Res Otolaryngol ; 15(1): 13-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24263968

ABSTRACT

Phalloidin, a toxin isolated from the death cap mushroom, Amanita phalloides, binds to filamentous actin with high affinity, and this has made fluorophore-conjugated phalloidin a useful tool in cellular imaging. Hepatocytes take up phalloidin via the liver-specific organic anion transporting polypeptide 1b2, but phalloidin does not permeate most living cells. Rapid entry of styryl dyes into live hair cells has been used to evaluate function, but the usefulness of those fluorescence dyes is limited by broad and fixed absorption spectra. Since phalloidin can be conjugated to fluorophores with various spectra, we investigated whether it would permeate living hair cells. When we incubated mouse utricles in 66 nM phalloidin-CF488A and followed that by washes in phalloidin-free medium, we observed that it entered a subset of hair cells and labeled entire hair bundles fluorescently after 20 min. Incubations of 90 min labeled nearly all the hair bundles. When phalloidin-treated utricles were cultured for 24 h after washout, the label disappeared from the hair cells and progressively but heterogeneously labeled filamentous actin in the supporting cells. We investigated how phalloidin may enter hair cells and found that P2 receptor antagonists, pyridoxalphosphate-6-azophenyl-2', 4'-disulfonic acid and suramin, blocked phalloidin entry, while the P2Y receptor ligands, uridine-5'-diphosphate and uridine-5'-triphosphaste, stimulated uptake. Consistent with that, the P2Y6 receptor antagonist, MRS 2578, decreased phalloidin uptake. The results show that phalloidin permeates live hair cells through a pathway that requires metabotropic P2Y receptor signaling and suggest that phalloidin can be transferred from hair cells to supporting cells in culture.


Subject(s)
Amanita , Cell Membrane Permeability/physiology , Chromophore-Assisted Light Inactivation , Hair Cells, Auditory, Inner/metabolism , Phalloidine/pharmacokinetics , Plant Extracts/pharmacokinetics , Receptors, Purinergic P2Y/metabolism , Actins/metabolism , Animals , Cell Membrane Permeability/drug effects , Cells, Cultured , Fluorescent Dyes , Hair Cells, Auditory, Inner/cytology , Isothiocyanates/pharmacology , Mice , Models, Animal , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Thiourea/analogs & derivatives , Thiourea/pharmacology
8.
J Nutr ; 142(4): 724-30, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22399524

ABSTRACT

Hyperglycemia-induced vascular inflammation resulting in the enhanced monocyte-endothelial cell (EC) interaction is the key event in the pathogenesis of atherosclerosis in diabetes. Here, we investigated the effect of isoflavone genistein on hyperglycemia-stimulated vascular inflammation. Human aortic EC (HAEC) were pretreated with genistein before the addition of high glucose (HG; 25 mmol/L) for 48 h. Genistein at a physiological concentration (0.1 µmol/L) significantly inhibited HG-induced adhesion of monocytes to HAEC and suppressed endothelial production of monocyte chemotactic protein-1 (MCP-1) and IL-8. Inhibition of adenylate cyclase or protein kinase A (PKA) significantly attenuated the antiadhesion effect of genistein. Consistently, genistein improved HG-impaired intracellular cAMP production and PKA activity in HAEC. Six-week-old diabetic db/db mice were untreated (db/db) or treated with a diet containing 1 g genistein/kg diet (db/db+G) for 8 wk. Their nondiabetic db/+ mice were used as normal controls. Circulating concentrations of MCP-1/JE and KC were significantly greater, whereas IL-10 concentrations were lower in db/db mice than those in normal mice. Dietary supplementation of genistein did not normalize but significantly suppressed the elevated serum concentrations of MCP-1/JE from 286 ± 30 ng/L to 181 ± 35 ng/L and KC from 321 ± 21 ng/L to 232 ± 20 ng/L while increasing that of IL-10 from 35 ± 4 ng/L to 346 ± 35 ng/L in db/db+G mice. Further, genistein treatment suppressed diabetes-induced adhesion of monocytes to EC by 87% and endothelial secretion of adhesion molecules. We conclude that genistein improves diabetes-caused vascular inflammation, which may be mediated through promoting the cAMP/PKA pathway.


Subject(s)
Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/diet therapy , Endothelium, Vascular/immunology , Genistein/therapeutic use , Obesity/diet therapy , Second Messenger Systems , Vasculitis/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aorta/cytology , Cell Adhesion/drug effects , Cells, Cultured , Cyclic AMP/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cytokines/blood , Cytokines/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/physiopathology , Dietary Supplements , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Hyperglycemia/etiology , Male , Mice , Mice, Obese , Monocytes/drug effects , Obesity/complications , Obesity/immunology , Obesity/physiopathology , Second Messenger Systems/drug effects , Vasculitis/etiology
9.
Clin Sci (Lond) ; 122(9): 429-37, 2012 May.
Article in English | MEDLINE | ID: mdl-22087608

ABSTRACT

AM5 (adrenomedullin 5), a newly described member of the CGRP (calcitonin gene-related peptide) family, is reported to play a role in normal cardiovascular physiology. The effects of AM5 in HF (heart failure), however, have not been investigated. In the present study, we intravenously infused two incremental doses of AM5 (10 and 100 ng/min per kg of body weight each for 90 min) into eight sheep with pacing-induced HF. Compared with time-matched vehicle control infusions, AM5 produced progressive and dose-dependent increases in left ventricular dP/dt(max) [LD (low dose), +56 mmHg/s and HD (high dose), +152 mmHg/s] and cardiac output (+0.83 l/min and +1.81 l/min), together with decrements in calculated total peripheral resistance (-9.4 mmHg/min per litre and -14.7 mmHg/min per litre), mean arterial pressure (-2.8 mmHg and -8.4 mmHg) and LAP (left atrial pressure; -2.6 mmHg and -5.6 mmHg) (all P<0.001). HD AM5 significantly raised PRA (plasma renin activity) (3.5-fold increment, P<0.001), whereas plasma aldosterone levels were unchanged over the intra-infusion period and actually fell in the post-infusion period (70% decrement, P<0.01), resulting in a marked decrease in the aldosterone/PRA ratio (P<0.01). Despite falls in LAP, plasma atrial natriuretic peptide and B-type natriuretic peptide concentrations were maintained relative to controls. AM5 infusion also induced significant increases in urine volume (HD 2-fold increment, P<0.05) and urine sodium (2.7-fold increment, P<0.01), potassium (1.7-fold increment, P<0.05) and creatinine (1.4-fold increment, P<0.05) excretion and creatinine clearance (60% increment, P<0.05). In conclusion, AM5 has significant haemodynamic, endocrine and renal actions in experimental HF likely to be protective and compensatory in this setting. These results suggest that AM5 may have potential as a therapeutic agent in human HF.


Subject(s)
Adrenomedullin/pharmacology , Heart Failure/drug therapy , Adrenomedullin/administration & dosage , Adrenomedullin/classification , Adrenomedullin/physiology , Aldosterone/blood , Animals , Atrial Natriuretic Factor/blood , Cyclic AMP/blood , Disease Models, Animal , Female , Heart Failure/physiopathology , Hemodynamics/drug effects , Hemodynamics/physiology , Humans , Infusions, Intravenous , Kidney/drug effects , Kidney/physiopathology , Natriuretic Peptide, Brain/blood , Renin/blood , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Second Messenger Systems/drug effects , Sheep, Domestic
10.
Indian J Exp Biol ; 50(12): 883-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23986972

ABSTRACT

Tridax procumbens leaf extract induced aortic relaxation in a concentration-dependent manner, for both phenylephrine (PE) and KCl- induced contractions in isolated rat aortic rings. The relaxation effect of the extract on PE-induced contraction was 57% greater than that on KCl- induced contraction. The extract caused dose-dependent relaxations in precontracted isolated rat aorta with phenylephrine; the relaxation was attenuated by the removal of endothelium. However, the relaxation responses to sodium nitroprusside were not significantly abolished by the removal of endothelium. The vasorelaxatory effect of the extract was completely abolished in presence of L-NAME. The results indicate that the vasorelaxant effect of T. procumbens extract is probably mediated by both endothelium-dependent and-independent mechanisms.


Subject(s)
Aorta, Thoracic/drug effects , Asteraceae/chemistry , Endothelium, Vascular/drug effects , Plant Extracts/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Acetylcholine/pharmacology , Animals , Calcium Signaling/drug effects , Cyclic GMP/physiology , Dose-Response Relationship, Drug , Female , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/physiology , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Potassium/pharmacology , Rats , Rats, Wistar , Second Messenger Systems/drug effects , Solvents , Vasodilator Agents/isolation & purification , Water
11.
Biochem J ; 432(3): 575-84, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20923411

ABSTRACT

hESCs (human embryonic stem cells) have enormous potential for use in pharmaceutical development and therapeutics; however, to realize this potential, there is a requirement for simple and reproducible cell culture methods that provide adequate numbers of cells of suitable quality. We have discovered a novel way of blocking the spontaneous differentiation of hESCs in the absence of exogenous cytokines by supplementing feeder-free conditions with EHNA [erythro-9-(2-hydroxy-3-nonyl)adenine], an established inhibitor of ADA (adenosine deaminase) and cyclic nucleotide PDE2 (phosphodiesterase 2). hESCs maintained in feeder-free conditions with EHNA for more than ten passages showed no reduction in hESC-associated markers including NANOG, POU5F1 (POU domain class 5 transcription factor 1, also known as Oct-4) and SSEA4 (stage-specific embryonic antigen 4) compared with cells maintained in feeder-free conditions containing bFGF (basic fibroblast growth factor). Spontaneous differentiation was reversibly suppressed by the addition of EHNA, but, upon removing EHNA, hESC populations underwent efficient spontaneous, multi-lineage and directed differentiation. EHNA also acts as a strong blocker of directed neuronal differentiation. Chemically distinct inhibitors of ADA and PDE2 lacked the capacity of EHNA to suppress hESC differentiation, suggesting that the effect is not driven by inhibition of either ADA or PDE2. Preliminary structure-activity relationship analysis found the differentiation-blocking properties of EHNA to reside in a pharmacophore comprising a close adenine mimetic with an extended hydrophobic substituent in the 8- or 9-position. We conclude that EHNA and simple 9-alkyladenines can block directed neuronal and spontaneous differentiation in the absence of exogenous cytokine addition, and may provide a useful replacement for bFGF in large-scale or cGMP-compliant processes.


Subject(s)
Adenine/analogs & derivatives , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Adenine/pharmacology , Adenosine Deaminase Inhibitors/pharmacology , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Culture Techniques/methods , Cell Line , Embryonic Stem Cells/cytology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Nanog Homeobox Protein , Neurons/drug effects , Neurons/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phosphodiesterase Inhibitors/pharmacology , Pluripotent Stem Cells/cytology , Second Messenger Systems/drug effects , Stage-Specific Embryonic Antigens/metabolism , Structure-Activity Relationship , Time Factors
12.
J Neurochem ; 108(1): 126-38, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19094061

ABSTRACT

The lipid diacylglycerol (DAG) analogue 1-oleoyl-2-acetyl-sn-glycerol (OAG) was used to verify the existence of DAG-sensitive channels in cortical neurons dissociated from E13 mouse embryos. Calcium imaging experiments showed that OAG increased the cytosolic concentration of Ca(2+) ([Ca(2+)]i) in nearly 35% of the KCl-responsive cells. These Ca(2+) responses disappeared in a Ca(2+)-free medium supplemented with EGTA. Mn(2+) quench experiments showed that OAG activated Ca(2+)-conducting channels that were also permeant to Ba(2+). The OAG-induced Ca(2+) responses were unaffected by nifedipine or omega-conotoxin GVIA (Sigma-Aldrich, Saint-Quentin Fallavier, France) but blocked by 1-[beta-(3-(4-Methoxyphenyl)propoxy)-4-methoxyphenethyl]-1H-imidazole hydrochloride (SKF)-96365 and Gd(3+). Replacing Na(+) ions with N-methyl-D-glucamine diminished the amplitude of the OAG-induced Ca(2+) responses showing that the Ca(2+) entry was mediated via Na(+)-dependent and Na(+)-independent mechanisms. Experiments carried out with the fluorescent Na(+) indicator CoroNa Green showed that OAG elevated [Na(+)]i. Like OAG, the DAG lipase inhibitor RHC80267 increased [Ca(2+)]i but not the protein kinase C activator phorbol 12-myristate 13-acetate. Moreover, the OAG-induced Ca(2+) responses were not regulated by protein kinase C activation or inhibition but they were augmented by flufenamic acid which increases currents through C-type transient receptor potential protein family (TRPC) 6 channels. In addition, application of hyperforin, a specific activator of TRPC6 channels, elevated [Ca(2+)]i. Whole-cell patch-clamp recordings showed that hyperforin activated non-selective cation channels. They were blocked by SKF-96365 but potentiated by flufenamic acid. Altogether, our data show the presence of hyperforin- and OAG-sensitive Ca(2+)-permeable channels displaying TRPC6-like properties. This is the first report revealing the existence of second messenger-operated channels in cortical neurons.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Cerebral Cortex/cytology , Diglycerides/pharmacology , Neurons/drug effects , Second Messenger Systems/drug effects , TRPC Cation Channels/physiology , Aniline Compounds/metabolism , Animals , Bridged Bicyclo Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Cells, Cultured , Cyclohexanones/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Fura-2/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Potassium Chloride/pharmacology , Sodium/metabolism , Terpenes/pharmacology , Xanthenes/metabolism
13.
J Oral Pathol Med ; 38(5): 434-40, 2009 May.
Article in English | MEDLINE | ID: mdl-18811671

ABSTRACT

BACKGROUND: Tongue cancer metastasis is mainly through blood stream and possibly associated with tumor cell-induced platelet aggregation (TCIPA). METHODS: Platelet aggregation was induced by different amounts of SAS tongue cancer cells with/without inhibitors and the latent period for induction of platelet aggregation was recorded. Gene expression was analyzed by reverse transcriptase-polymerase chain reaction. RESULTS: SAS cells (4 x 10(4) to 1 x 10(6) cells/ml) induced platelet aggregation in a cell density-dependent manner. The latent period for induction of platelet aggregation reduced from 11.3 min (2 x 10(5) cells/ml) to 0.9 min (5 x 10(5) cells/ml). The extent of platelet aggregation increased from 39% to 76% by 2 x 10(5) and 5 x 10(5) SAS cells. Pre-treatment of SAS cells with aspirin showed little effect on its induction of platelet aggregation. SAS cells expressed tissue factor (TF) mRNA and the SAS cells-induced TCIPA was inhibited by TF neutralization antibody (5-20 microg/ml), heparin (5-10 U/ml), Hirudin fragment 54-65 (50 microg/ml) and D-Phenylalanyl-L-prolyl-L-arginine chloromethyl ketone. But areca nut (AN, a betel quid component known to generate reactive oxygen species (ROS)) extract showed little effect on TF expression in SAS cells. Pre-treatment with U73122 and 2-aminoethoxydiphenylborate inhibited SAS-induced TCIPA. Interestingly, catalase suppressed SAS cells-induced TCIPA, whereas AN extract enhanced this event. CONCLUSIONS: These results suggest that tongue cancer cells may induce TCIPA and enhance tumor metastasis. SAS-induced TCIPA is related to TF secretion, thrombin generation and associated with Phospholipase C-Inositol triphosphate signaling and ROS production. Betel quid chewing may potentially promote tongue cancer metastasis.


Subject(s)
Areca , Plant Extracts/pharmacology , Platelet Aggregation/physiology , Thromboplastin/metabolism , Tongue Neoplasms/metabolism , Coculture Techniques , Epithelial Cells/metabolism , Gene Expression Profiling , Gingiva/cytology , Gingiva/metabolism , Humans , Mouth Mucosa/cytology , Mouth Mucosa/metabolism , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Platelet Aggregation/drug effects , RNA, Messenger/analysis , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Statistics, Nonparametric , Thromboplastin/genetics , Time Factors , Tumor Cells, Cultured
14.
Prog Brain Res ; 170: 379-88, 2008.
Article in English | MEDLINE | ID: mdl-18655897

ABSTRACT

Glucocorticoids secreted in response to stress activation of the hypothalamic-pituitary-adrenal axis feed back onto the hypothalamus to rapidly suppress neuroendocrine activation, including oxytocin and vasopressin secretion. Here we provide a brief review focused on our recent findings of a rapid glucocorticoid-induced opposing regulation of glutamate and gamma-aminobutyric acid (GABA) inputs to magnocellular neurons via the release of distinct retrograde messengers. The stress hormone corticosterone and its synthetic analogue dexamethasone elicit the rapid retrograde release of endocannabinoids by activating a novel membrane-associated, G protein-coupled receptor in parvocellular and magnocellular neuroendocrine cells of the hypothalamic paraventricular and supraoptic nuclei. Glucocorticoids also cause the rapid retrograde release of an unknown messenger that facilitates presynaptic GABA release onto magnocellular neuroendocrine cells. These finding suggest that there is a strict synapse-specific segregation of the opposing actions of the two retrogradely released messengers. Thus, the combined actions of glucocorticoids cause a rapid synaptic inhibition of the magnocellular neurons and would be expected, therefore, to mediate a rapid feedback inhibition of the secretion of oxytocin and vasopressin during stress activation of the hypothalamic-pituitary-adrenal axis.


Subject(s)
Basal Nucleus of Meynert/physiology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Neurons/physiology , Synapses/physiology , Animals , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Glutamic Acid/physiology , Homeostasis/drug effects , Homeostasis/physiology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/drug effects , Hypothalamus/physiology , Mammals , Neurons/drug effects , Norepinephrine/pharmacology , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Synapses/drug effects , TRPV Cation Channels/drug effects , TRPV Cation Channels/physiology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology
15.
Behav Brain Res ; 187(2): 254-61, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-17964671

ABSTRACT

The effects of a single intraperitoneal administration of lithium, a drug used to prevent the recurrence of mania in bipolar disorders, were determined in the rat by studying changes in: (i) the wake-sleep cycle; (ii) autonomic parameters (hypothalamic and tail temperature, heart rate); (iii) the capacity to accumulate cAMP and IP(3) in the preoptic-anterior hypothalamic region (PO-AH) and in the cerebral cortex (CC) under an hypoxic stimulation at normal laboratory and at low ambient temperature (T(a)). In the immediate hours following the injection, lithium induced: (i) a significant reduction in REM sleep; (ii) a non-significant reduction in the delta power density of the EEG in NREM sleep; (iii) a significant decrease in the concentration of cAMP in PO-AH at normal laboratory T(a); (iv) a significant increase of IP(3) concentration in CC following exposure to low T(a). The earliest and most sensitive effects of lithium appear to be those concerning sleep. These changes are concomitant with biochemical effects that, in spite of a systemic administration of the substance, may be differentiated according to the second messenger involved, the brain region and the ambient condition.


Subject(s)
Antimanic Agents/pharmacology , Body Temperature Regulation/drug effects , Brain/drug effects , Lithium Chloride/pharmacology , Second Messenger Systems/drug effects , Sleep, REM/drug effects , Analysis of Variance , Animals , Autonomic Nervous System/drug effects , Brain/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cyclic AMP/metabolism , Electroencephalography/drug effects , Heart Rate/drug effects , Hypothalamus/drug effects , Hypoxia , Inositol 1,4,5-Trisphosphate/metabolism , Male , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric
16.
Circulation ; 114(1 Suppl): I566-72, 2006 Jul 04.
Article in English | MEDLINE | ID: mdl-16820639

ABSTRACT

BACKGROUND: Calcific aortic valve disease is a common condition and is associated with inflammatory changes and expression of osteoblast-like cell phenotypes, but the cellular mechanisms are unclear. Recent studies identified extracellular ATP and P2Y receptor cascade as important regulators of bone remodeling, whereas its breakdown product, adenosine, is known to have anti-inflammatory properties. We hypothesize that extracellular ATP and adenosine have important roles in regulating osteoblast differentiation in human valve interstitial cells, and that this can be a potential target for therapy. Method and Results- Primary cultures of human valve interstitial cells (ICs) treated for 21 days with osteogenic media, ATP, and ATP-gamma-S (a stable agonist of the P2Y receptor) revealed a significant increase in alkaline phosphatase (ALP) (an osteoblast marker) activity and expression as measured using spectrophotometric assay and immunocytochemistry staining. Valve ICs treated with adenosine alone did not cause an increase in ALP activity; however, adenosine treatment decreased the ALP activity and expression induced by osteogenic media after 21 days of incubation. In addition, atorvastatin inhibited the activity of ALP induced by ATP in human valve ICs, and enzyme studies revealed that atorvastatin upregulated the breakdown of extracellular ATP into adenosine in human valve ICs after 24-hour treatment. CONCLUSIONS: These findings identify a novel role for extracellular nucleotides in inducing osteoblast differentiation in human valve ICs in vitro and provide a potential therapeutic target for preventing the disease progression.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/physiology , Adenosine/pharmacology , Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Heptanoic Acids/pharmacology , Osteoblasts/drug effects , Pyrroles/pharmacology , Receptors, Purinergic P2/physiology , 5'-Nucleotidase/analysis , Adenosine Triphosphatases/analysis , Adenosine Triphosphate/pharmacology , Aged , Alkaline Phosphatase/analysis , Aortic Valve/cytology , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/pathology , Apyrase/analysis , Atorvastatin , Biomarkers , Calcinosis/drug therapy , Calcinosis/pathology , Cell Differentiation , Cells, Cultured/cytology , Cells, Cultured/drug effects , Drug Evaluation, Preclinical , Heptanoic Acids/therapeutic use , Humans , Middle Aged , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoblasts/physiology , Pyrroles/therapeutic use , Receptors, Purinergic P2/drug effects , Second Messenger Systems/drug effects
17.
Neuroendocrinology ; 81(6): 381-90, 2005.
Article in English | MEDLINE | ID: mdl-16276116

ABSTRACT

Gamma-aminobutyric acid (GABA) is an emerging signalling molecule in endocrine organs, since it is produced by endocrine cells and acts via GABA(A) receptors in a paracrine/autocrine fashion. Testicular Leydig cells are producers and targets for GABA. These cells express GABA(A) receptor subunits and in the murine Leydig cell line TM3 pharmacological activation leads to increased proliferation. The signalling pathway of GABA in these cells is not known in this study. We therefore attempted to elucidate details of GABA(A) signalling in TM3 and adult mouse Leydig cells using several experimental approaches. TM3 cells not only express GABA(A )receptor subunits, but also bind the GABA agonist [(3)H]muscimol with a binding affinity in the range reported for other endocrine cells (K(d) = 2.740 +/- 0.721 nM). However, they exhibit a low B(max) value of 28.08 fmol/mg protein. Typical GABA(A) receptor-associated events, including Cl(-) currents, changes in resting membrane potential, intracellular Ca(2+) or cAMP, were not measurable with the methods employed in TM3 cells, or, as studied in part, in primary mouse Leydig cells. GABA or GABA(A) agonist isoguvacine treatment resulted in increased or decreased levels of several mRNAs, including transcription factors (c-fos, hsf-1, egr-1) and cell cycle-associated genes (Cdk2, cyclin D1). In an attempt to verify the cDNA array results and because egr-1 was recently implied in Leydig cell development, we further studied this factor. RT-PCR and Western blotting confirmed a time-dependent regulation of egr-1 in TM3. In the postnatal testis egr-1 was seen in cytoplasmic and nuclear locations of developing Leydig cells, which bear GABA(A) receptors and correspond well to TM3 cells. Thus, GABA acts via an atypical novel signalling pathway in TM3 cells. Further details of this pathway remain to be elucidated.


Subject(s)
Leydig Cells/physiology , Receptors, GABA/physiology , Signal Transduction/physiology , Animals , Blotting, Western , Calcium Signaling/physiology , Cell Line , Chloride Channels/physiology , Cyclic AMP/physiology , DNA, Complementary/biosynthesis , Early Growth Response Protein 1/biosynthesis , GABA Agonists/metabolism , Gene Expression Regulation/physiology , Immunohistochemistry , Isonicotinic Acids/pharmacology , Male , Membrane Potentials/physiology , Mice , Mice, Inbred BALB C , Muscimol/metabolism , Oligonucleotide Array Sequence Analysis , Patch-Clamp Techniques , RNA/biosynthesis , RNA/genetics , Receptors, GABA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Second Messenger Systems/drug effects , Signal Transduction/genetics , Spectrometry, Fluorescence , Testis/cytology , Testis/drug effects , Testis/metabolism
18.
Front Neuroendocrinol ; 26(2): 65-84, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16009409

ABSTRACT

It is well known that many of the actions of gonadal steroids in hypothalamic neurons are mediated via intracellular receptor/transcription factors that interact with steroid response elements on target genes. Since the cloning of the intracellular steroid receptors/transcription factors, it has been assumed that most if not all of the actions of the gonadal steroids are mediated via these intracellular receptors. However, there now exist compelling evidence for membrane (G-protein-coupled) steroid receptors for estrogen and progesterone in hypothalamic and other brain neurons. But, it is not well understood how steroids signal via membrane receptors, and how these signals impact not only membrane excitability but also gene transcription in hypothalamic neurons. Indeed, it has been known for sometime that gonadal steroids can rapidly alter hypothalamic neuronal activity within seconds, indicating that some cellular effects can occur via membrane delimited events. In addition, gonadal steroids can affect second messenger systems, including calcium and various kinases to prompt and/or alter cell signaling. Therefore, this chapter will consider our current knowledge of rapid (i.e., seconds to minutes) membrane-initiated and intracellular signaling as well as classical nuclear receptor signaling by gonadal steroids in hypothalamic neurons, the nature of these receptors and how they contribute to homeostatic functions.


Subject(s)
Estrogens/physiology , Hypothalamus/physiology , Ovary/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Animals , Estradiol/physiology , Female , Gonadotropin-Releasing Hormone , Humans , MAP Kinase Signaling System/physiology , Neurons/drug effects , Neurons/physiology , Pro-Opiomelanocortin/physiology , Progesterone/physiology , Receptors, Estrogen/physiology , Receptors, Progesterone/physiology , Second Messenger Systems/drug effects , gamma-Aminobutyric Acid/physiology
19.
FASEB J ; 18(11): 1318-20, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15180965

ABSTRACT

Interleukin-23, a recently described cytokine produced by activated antigen-presenting cells, including dendritic cells, is a p19/p40 heterodimer. The p40 subunit is shared with IL-12, the major Th1-driving cytokine, while p19 is distantly related to IL-12 p35. IL-23 has pro-inflammatory actions, inducing IL-17 secretion from activated CD4+ T cells, and stimulating the proliferation of memory CD4+ T cells. Here, we examined the effects of PGE2, a well-known immunomodulator, on the production of IL-23 by bone marrow- derived dendritic cells (BM-DCs). Our results indicate that PGE2 increases the production of functional IL-23 from immature BM-DCs in a time- and dose-dependent manner. PGE2 induces both the expression of p19 and p40, without affecting p35 expression. The effect of PGE2 is mediated through the specific receptors EP2/4 and is mimicked by cAMP-inducing agents, such as forskolin and dbcAMP. Although PGE2 also induces IL-1beta and IL-6 expression in non-stimulated DCs, the stimulatory effect of PGE2 on IL-23 production is not mediated through IL-1beta or IL-6. GM-CSF, the pro-inflammatory cytokine required for the generation of BM-DCs, amplifies the IL-23 inducing activity of PGE2 in a synergistic manner. Recent studies described both pro- and anti-inflammatory effects of PGE2, and our results suggest an additional mechanism for its pro-inflammatory role, particularly significant for autoimmune diseases, such as rheumatoid arthritis.


Subject(s)
Alprostadil/analogs & derivatives , Dendritic Cells/drug effects , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Gene Expression Regulation/drug effects , Interleukins/biosynthesis , Alprostadil/pharmacology , Animals , Arthritis/metabolism , Bone Marrow Cells/metabolism , Bucladesine/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Cell Line, Tumor/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Colforsin/pharmacology , Culture Media, Conditioned/pharmacology , Cyclic AMP/physiology , Dendritic Cells/metabolism , Dinoprostone/antagonists & inhibitors , Inflammation , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-12 Subunit p35 , Interleukin-12 Subunit p40 , Interleukin-17/biosynthesis , Interleukin-17/genetics , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/genetics , Lipopolysaccharides/pharmacology , Male , Mice , Misoprostol/pharmacology , Plasmacytoma/pathology , Prostaglandin Antagonists/pharmacology , Protein Subunits/biosynthesis , Protein Subunits/genetics , Receptors, Cell Surface/physiology , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Specific Pathogen-Free Organisms , Toll-Like Receptor 2 , Toll-Like Receptor 4
20.
J Ocul Pharmacol Ther ; 19(6): 501-15, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14733708

ABSTRACT

Natural prostaglandins (PGs) such as PGD2, PGE2, PGF2(2alpha), and PGI2 exhibited the highest affinity for their respective cognate receptors, but were the least selective agents when tested in receptor binding assays. Travoprost acid ([+]-fluprostenol) was the most FP-receptor-selective compound, exhibiting a high affinity (Ki = 35 +/- 5 nM) for the FP receptor, and minimal affinity for DP (Ki = 52,000 nM), EP1 (Ki = 9540 nM), EP3 (Ki = 3501 nM), EP4 (Ki = 41,000 nM), IP (Ki > 90,000 nM), and TP (Ki = 121,000 nM) receptors. Travoprost acid was the most potent PG analog tested in FP receptor functional phosphoinositide turnover assays in the following cell types: human ciliary muscle (EC50 = 1.4 nM), human trabecular meshwork (EC50 = 3.6 nM), and mouse fibroblasts and rat aortic smooth muscle cells (EC50 = 2.6 nM). Although latanoprost acid exhibited a relatively high affinity for the FP receptor (Ki = 98 nM), it had significant functional activity at FP (EC50 = 32-124 nM) and EP1 (EC50 = 119 nM) receptors. Bimatoprost acid was less selective, exhibiting a relatively high affinity for the FP (Ki = 83 nM), EP1 (Ki = 95 nM), and EP3 (Ki = 387 nM) receptors. Bimatoprost acid exhibited functional activity at the EP1 (EC50 = 2.7 nM) and FP (EC50 = 2.8-3.8 nM in most cells) receptors. Bimatoprost (nonhydrolyzed amide) also behaved as an FP agonist at the cloned human FP receptor (EC50 = 681 nM), in h-TM (EC50 = 3245 nM) and other cell types. Unoprostone and S-1033 bound with low affinity (Ki = 5.9 microM to > 22 microM) to the FP receptor, were not selective, but activated the FP receptor. In conclusion, travoprost acid has the highest affinity, the highest FP-receptor-selectivity, and the highest potency at the FP receptor as compared to the other ocular hypotensive PG analogs known so far, including free acids of latanoprost, bimatoprost, and unoprostone isopropyl ester.


Subject(s)
Binding, Competitive/drug effects , Cloprostenol/analogs & derivatives , Dinoprost/analogs & derivatives , Intraocular Pressure/drug effects , Prostaglandins F, Synthetic/pharmacology , Receptors, Prostaglandin/drug effects , Receptors, Prostaglandin/physiology , Amides , Animals , Aorta/cytology , Aorta/drug effects , Bimatoprost , Binding, Competitive/physiology , Cattle , Cell Line , Ciliary Body/cytology , Ciliary Body/drug effects , Clinical Trials as Topic , Cloprostenol/chemistry , Cloprostenol/metabolism , Cloprostenol/pharmacology , Dinoprost/pharmacology , Drug Evaluation, Preclinical , Fibroblasts/drug effects , Humans , Intraocular Pressure/physiology , Kidney/cytology , Latanoprost , Lipid Metabolism , Lipids/pharmacology , Mice , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacology , Prostaglandins/pharmacology , Prostaglandins F, Synthetic/chemistry , Prostaglandins, Synthetic/chemistry , Prostaglandins, Synthetic/metabolism , Prostaglandins, Synthetic/pharmacology , Radioligand Assay , Rats , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/classification , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Stereoisomerism , Trabecular Meshwork/cytology , Trabecular Meshwork/drug effects , Travoprost
SELECTION OF CITATIONS
SEARCH DETAIL