Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Aust Endod J ; 49 Suppl 1: 330-338, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36723392

ABSTRACT

Iloprost's anti-inflammatory effects on human dental pulp stem cells (HDPCs) are currently unknown. We hypothesized that iloprost could downregulate the expression of inflammatory-related genes and protein in an inflamed HDPC in vitro model. To induce inflammation, the HDPCs were treated with a cocktail of interleukin-1 beta, interferon-gamma, and tumour necrosis alpha, at a ratio of 1:10:100. Iloprost (10-6  M) was then added or not to the cultures. Interleukin-6 (IL-6) and interleukin-12 (IL-12) mRNA expression were assessed by real-time polymerase chain reaction. IL-6 protein expression was assessed by enzyme-linked immunosorbent assay. The results were analysed using one-way ANOVA or the Kruskal-Wallis test. The cytokine cocktail induced more robust IL-6 expression than LPS treatment. Iloprost slightly, yet significantly, downregulated IL-6 and IL-12 mRNA expression. These findings suggest that iloprost might be used as a beneficial component in vital pulp therapy.


Subject(s)
Epoprostenol , Iloprost , Humans , Iloprost/pharmacology , Iloprost/metabolism , Epoprostenol/metabolism , Epoprostenol/pharmacology , Interleukin-6 , Dental Pulp/metabolism , Interleukin-12/metabolism , Interleukin-12/pharmacology , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Cells, Cultured , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism
2.
Int J Mol Sci ; 23(17)2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36077548

ABSTRACT

The prostacyclin analogue iloprost is used to treat vascular alterations and digital ulcers, the early derangements manifesting in systemic sclerosis (SSc), an autoimmune disease leading to skin and organ fibrosis. Bioindicator(s) of SSc onset and progress are still lacking and the therapeutic approach remains a challenge. The T helper 1 (Th1) chemokine interferon (IFN)γ-induced protein 10 (IP-10/CXCL10) associates with disease progression and worse prognosis. Endothelial cells and fibroblasts, under Th1-dominance, release CXCL10, further enhancing SSc's detrimental status. We analyzed the effect of iloprost on CXCL10 in endothelial cells, dermal fibroblasts, and in the serum of SSc patients. Human endothelial cells and dermal fibroblasts activated with IFNγ/Tumor Necrosis Factor (TNF)α, with/without iloprost, were investigated for CXCL10 secretion/expression and for intracellular signaling cascade underlying chemokine release (Signal Transducer and Activator of Transcription 1, STAT1; Nuclear Factor kappa-light-chain-enhancer of activated B cells, NF-kB; c-Jun NH2-terminal kinase, JNK: Phosphatidyl-Inositol 3-kinase (PI3K)/protein kinase B, AKT; Extracellular signal-Regulated Kinase 1/2, ERK1/2). CXCL10 was quantified in sera from 25 patients taking iloprost, satisfying the American College of Rheumatology (ACR)/European Alliance of Associations for Rheumatology (EULAR) 2013 classification criteria for SSc, and in sera from 20 SSc sex/age-matched subjects without therapy, previously collected. In human endothelial cells and fibroblasts, iloprost targeted CXCL10, almost preventing IFNγ/TNFα-dependent cascade activation in endothelial cells. In SSc subjects taking iloprost, serum CXCL10 was lower. These in vitro and in vivo data suggest a potential role of iloprost to limit CXCL10 at local vascular/dermal and systemic levels in SSc and warrant further translational research aimed to ameliorate SSc understanding/management.


Subject(s)
Iloprost , Scleroderma, Systemic , Chemokine CXCL10/metabolism , Chemokines/metabolism , Endothelial Cells/metabolism , Epoprostenol/metabolism , Humans , Iloprost/metabolism , Iloprost/pharmacology , Iloprost/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
3.
J Thromb Haemost ; 20(7): 1699-1711, 2022 07.
Article in English | MEDLINE | ID: mdl-35395698

ABSTRACT

BACKGROUND: Thrombotic antiphospholipid syndrome (t-PAPS) is characterized by arterial, venous, or microvascular occlusions, which are explained, in part, by the presence of antiphospholipid (aPL) antibodies. Although there is much evidence indicating that isolated aPL antibodies increase the activity of platelets obtained from healthy volunteers, platelet function in t-PAPS has not been as widely studied. OBJECTIVE: To evaluate platelet reactivity in t-PAPS patients. METHODS: Platelet aggregation, protein expression, and cyclic nucleotide levels were carried out in platelet rich plasma (PRP) or washed platelets (WPs) obtained from t-PAPS or healthy volunteers. RESULTS: ADP-induced aggregation was significantly higher in PRP obtained from t-PAPS than obtained from the control. The protein expression of P2Y12 receptor and Gs alpha was significantly higher and lower, respectively in WPs from t-PAPS patients. In PRP incubated with iloprost or sodium nitroprusside, the residual platelet reactivity induced by ADP was still higher in PRP from t-PAPS than from the control. Lower intracellular levels of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) were observed in unstimulated PRP from t-PAPS patients. The protein expression of soluble guanylate cyclase subunits and phosphodiesterases types 3 and 5 did not differ. The antiplatelet activity of ticagrelor was similar between the groups and cilostazol significantly potentiated this response. Isolated aPL antibodies obtained from t-PAPS patients potentiated ADP-induced aggregation in healthy platelets but did not affect the inhibitory responses induced by iloprost or sodium nitroprusside. CONCLUSIONS: The overexpression of P2Y12 receptor, accompanied by lower levels of cAMP and cGMP levels produced greater amplitude of ADP aggregation in platelets from t-PAPS patients.


Subject(s)
Antiphospholipid Syndrome , Blood Platelets , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Antiphospholipid Syndrome/metabolism , Blood Platelets/metabolism , Cyclic AMP , Cyclic GMP/metabolism , Humans , Iloprost/metabolism , Iloprost/pharmacology , Nitroprusside/metabolism , Nitroprusside/pharmacology , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction
4.
Drug Dev Res ; 82(2): 217-229, 2021 04.
Article in English | MEDLINE | ID: mdl-32984987

ABSTRACT

Coronavirus disease 2019 (COVID 19) was first identified in Wuhan, China near the end of 2019. To date, COVID-19 had spread to almost 235 countries and territories due to its highly infectious nature. Moreover, there is no vaccine or Food and Drug Administration (FDA)-approved drug. More time is needed to establish one of them. Consequently, the drug repurposing approach seems to be the most attractive and quick solution to accommodate this crisis. In this regard, we performed molecular docking-based virtual screening of antiplatelet FDA-approved drugs on the key two viral target proteins: main protease (Mpro ) and spike glycoprotein (S) as potential inhibitor candidates for COVID-19. In the present study, 15 antiplatelet FDA-approved drugs were investigated against the concerned targets using the Molecular Docking Server. Our study revealed that only cilostazol has the most favorable binding interaction on Mpro (PDB ID: 6LU7) and cilostazol, iloprost, epoprostenol, prasugrel, and icosapent ethyl have a higher binding affinity on spike glycoprotein (S) (PDB ID: 6VYB) compared with recent anti-CoVID-19. Therefore, cilostazol is a promising FDA drug against COVID-19 by inhibiting both Mpro and S protein. The insights gained in this study may be useful for quick approach against COVID-19 in the future.


Subject(s)
COVID-19 Drug Treatment , Coronavirus 3C Proteases/metabolism , Platelet Aggregation Inhibitors/metabolism , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Cilostazol/metabolism , Cilostazol/therapeutic use , Drug Approval , Drug Evaluation, Preclinical , Drug Repositioning , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/therapeutic use , Epoprostenol/metabolism , Epoprostenol/therapeutic use , Humans , Iloprost/metabolism , Iloprost/therapeutic use , Molecular Docking Simulation , Platelet Aggregation Inhibitors/therapeutic use , Prasugrel Hydrochloride/metabolism , Prasugrel Hydrochloride/therapeutic use , United States , United States Food and Drug Administration
5.
Br J Pharmacol ; 177(1): 161-174, 2020 01.
Article in English | MEDLINE | ID: mdl-31476020

ABSTRACT

BACKGROUND AND PURPOSE: In patients with pulmonary hypertension (PH) associated with lung disease and/or hypoxia (Group III), decreased pulmonary vascular tone and tissue hypoxia is therapeutically beneficial. PGE2 and PGI2 induce potent relaxation of human bronchi from non-PH (control) patients via EP4 and IP receptors, respectively. However, the effects of PGE2 /PGI2 and their mimetics on human bronchi from PH patients are unknown. Here, we have compared relaxant effects of several PGI2 -mimetics approved for treating PH Group I with several PGE2 -mimetics, in bronchial preparations derived from PH Group III and control patients. EXPERIMENTAL APPROACH: Relaxation of bronchial muscle was assessed in samples isolated from control and PH Group III patients. Expression of prostanoid receptors was analysed by western blot and real-time PCR, and endogenous PGE2 , PGI2 , and cAMP levels were determined by ELISA. KEY RESULTS: Maximal relaxations induced by different EP4 receptor agonists (PGE2 , L-902688, and ONO-AE1-329) were decreased in human bronchi from PH patients, compared with controls. However, maximal relaxations produced by PGI2 -mimetics (iloprost, treprostinil, and beraprost) were similar for both groups of patients. Both EP4 and IP receptor protein and mRNA expressions were significantly lower in human bronchi from PH patients. cAMP levels significantly correlated with PGI2 but not with PGE2 levels. CONCLUSION AND IMPLICATIONS: The PGI2 -mimetics retained maximal bronchodilation in PH Group III patients, whereas bronchodilation induced by EP4 receptor agonists was decreased. Restoration of EP4 receptor expression in airways of PH Group III patients with respiratory diseases could bring additional therapeutic benefit.


Subject(s)
Bronchi/metabolism , Bronchodilator Agents/metabolism , Bronchodilator Agents/therapeutic use , Dinoprostone/metabolism , Dinoprostone/therapeutic use , Hypertension, Pulmonary/metabolism , Adult , Aged , Aged, 80 and over , Antihypertensive Agents/metabolism , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Bronchi/drug effects , Bronchi/pathology , Bronchodilator Agents/pharmacology , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Epoprostenol/analogs & derivatives , Epoprostenol/metabolism , Epoprostenol/pharmacology , Epoprostenol/therapeutic use , Female , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Iloprost/metabolism , Iloprost/pharmacology , Iloprost/therapeutic use , Male , Middle Aged , Organ Culture Techniques , Pyrrolidinones/metabolism , Pyrrolidinones/pharmacology , Pyrrolidinones/therapeutic use , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Tetrazoles/metabolism , Tetrazoles/pharmacology , Tetrazoles/therapeutic use , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use , Young Adult
6.
Thromb Haemost ; 111(3): 539-48, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24258486

ABSTRACT

Vasodilator-stimulated phosphoprotein (VASP) is phosphorylated and dephosphorylated consequent to increases and decreases in cyclic nucleotide levels. Monitoring changes in VASP phosphorylation is an established method for indirect measurement of cyclic nucleotides. Here we describe the use of an innovative cocktail, VASPFix, which allows sensitive and reproducible measurement of phosphorylated VASP (VASP-P) in a simple, single-step procedure using cytometric bead technology. Frozen VASPFix-treated samples are stable for at least six months prior to analysis. We successfully used VASPFix to measure VASP-P in platelets in both platelet-rich plasma and blood in response to compounds that increase (dibutyryl cAMP, adenosine, iloprost, PGE1) and decrease (ADP, PGE1) cAMP, and to determine the effects of certain receptor antagonists on the results obtained. The change in VASP-P brought about by adding ADP to PGE1-stimulated platelets is a combination of the effect of ADP at the P2Y12 receptor and of PGE1 at both IP and EP3 receptors. For iloprost-stimulated platelets EP3 receptors are not involved. A procedure in which iloprost, ADP and VASPFix were used to determine effectiveness of clopidogrel and prasugrel in patients was compared with an established commercial procedure that uses PGE1 and ADP; the latter produced higher platelet reactivity values that were the result of PGE1 interacting with platelet EP3 receptors. We conclude that VASPFix can be used both as a research tool and for clinical investigations and provides better specificity for P2Y12 receptor inhibition. The latter confers a distinct advantage over existing methods used to monitor effects of P2Y12 antagonists on platelet function.


Subject(s)
Biomarkers, Pharmacological/metabolism , Blood Platelets/drug effects , Cell Adhesion Molecules/metabolism , Cyclic AMP/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Platelet Function Tests/methods , Thrombophilia/diagnosis , Thrombophilia/drug therapy , Adenosine/metabolism , Adenosine Diphosphate/metabolism , Alprostadil/metabolism , Blood Platelets/physiology , Bucladesine/metabolism , Cells, Cultured , Clopidogrel , Humans , Iloprost/metabolism , Phosphorylation/drug effects , Piperazines/administration & dosage , Platelet Aggregation/drug effects , Prasugrel Hydrochloride , Purinergic P2Y Receptor Antagonists/administration & dosage , Reproducibility of Results , Sensitivity and Specificity , Thiophenes/administration & dosage , Ticlopidine/administration & dosage , Ticlopidine/analogs & derivatives
7.
Rev. esp. anestesiol. reanim ; 59(3): 142-149, mar. 2012.
Article in Spanish | IBECS | ID: ibc-100355

ABSTRACT

Objetivos: La presencia de hipertensión pulmonar tiene elevada incidencia durante el perioperatorio del trasplante pulmonar y puede condicionar deterioro hemodinámico que obligue a instaurar circulación extracorpórea. Nuestro objetivo es estudiar los efectos hemodinámicos en la circulación pulmonar y sistémica de la asociación de óxido nítrico e iloprost inhalados y sildenafilo por vía oral en pacientes con hipertensión pulmonar severa durante la cirugía de trasplante pulmonar. Pacientes y métodos: Durante el perioperatorio del trasplante pulmonar, 17 pacientes recibieron 10 mg de iloprost nebulizado cuando su presión arterial pulmonar media superó los 50 mmHg. Todos los pacientes recibieron 50 mg de sildenafilo por vía oral 30 min antes de la inducción anestésica y 20 ppm de NO inhalado tras la intubación traqueal. Se registraron las variables hemodinámicas y respiratorias en los tiempos basal (tras la inducción anestésica), previamente a la administración de iloprost, y a los 5 y 30 min de su administración. Resultados: La administración de iloprost redujo de forma significativa la presión arterial pulmonar e incrementó significativamente el índice cardiaco y la fracción de eyección del ventrículo derecho. No se produjeron modificaciones significativas de la presión arterial sistémica. Conclusiones: La triple asociación reduce significativamente las presiones pulmonares en el perioperatorio del trasplante pulmonar y debe considerarse en presencia de hipertensión pulmonar severa durante la intervención quirúrgica o el postoperatorio inmediato del trasplante pulmonar(AU)


Objectives: There is a high incidence of pulmonary hypertension during the lung transplant peri-operative period, and could lead to a haemodynamic deterioration that may require the need of extracorporeal circulation. Our aim was to study the haemodynamic effects on the pulmonary and systemic circulation of the combination of inhaled nitric oxide and iloprost and oral sildenafil in patients with severe pulmonary hypertension during lung transplant surgery. Patients and methods: Seventeen patients received 10mg of nebulised iloprost during the peri-operative period of the lung transplant when their mean pulmonary pressure exceeded 50 mmHg. All the patients received 50 mg of oral sildenafil 30 min before anaesthetic induction, 20 ppm of inhaled nitric oxide after tracheal intubation. The haemodynamic and respiratory variables were recorded at baseline (after anaesthetic induction), prior to the administering of iloprost, and at 5 and 30 min after it was given. Results: The administering of iloprost significantly reduced the pulmonary arterial pressure and significantly increases the cardiac index and the right ventricular ejection fraction. There were no significant changes occurred in the systemic arterial pressure. Conclusions: The triple combination significantly reduces the pulmonary pressures in the lung transplant peri-operative and should be considered when there is severe pulmonary hypertension during the surgery or during the immediate post-operative period of lung transplantation(AU)


Subject(s)
Humans , Male , Female , Iloprost/metabolism , Iloprost/pharmacokinetics , Iloprost/therapeutic use , Nitric Oxide/therapeutic use , Lung Transplantation/methods , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/surgery , Anesthesia/methods , Anesthesia , Adjuvants, Anesthesia/therapeutic use , Hypertension, Pulmonary/drug therapy , Monitoring, Intraoperative/instrumentation , Treatment Outcome , Lung Transplantation , Evaluation of the Efficacy-Effectiveness of Interventions
8.
PLoS One ; 7(12): e52248, 2012.
Article in English | MEDLINE | ID: mdl-23300624

ABSTRACT

BACKGROUND: Inhaled iloprost potentially improves hemodynamics and gas exchange in patients with chronic obstructive pulmonary disease (COPD) and secondary pulmonary hypertension (PH). OBJECTIVES: To evaluate acute effects of aerosolized iloprost in patients with COPD-associated PH. METHODS: A randomized, double blind, crossover study was conducted in 16 COPD patients with invasively confirmed PH in a single tertiary care center. Each patient received a single dose of 10 µg iloprost (low dose), 20 µg iloprost (high dose) and placebo during distinct study-visits. The primary end-point of the study was exercise capacity as assessed by the six minute walking distance. RESULTS: Both iloprost doses failed to improve six-minute walking distance (p = 0.36). Low dose iloprost (estimated difference of the means -1.0%, p = 0.035) as well as high dose iloprost (-2.2%, p<0.001) significantly impaired oxygenation at rest. Peak oxygen consumption and carbon dioxide production differed significantly over the three study days (p = 0.002 and p = 0.003, accordingly). As compared to placebo, low dose iloprost was associated with reduced peak oxygen consumption (-76 ml/min, p = 0.002), elevated partial pressure of carbon dioxide (0.27 kPa, p = 0.040) and impaired ventilation during exercise (-3.0l/min, p<0.001). CONCLUSIONS: Improvement of the exercise capacity after iloprost inhalation in patients with COPD-associated mild to moderate PH is very unlikely. TRIAL REGISTRATION: Controlled-Trials.com ISRCTN61661881.


Subject(s)
Hypertension, Pulmonary/complications , Hypertension, Pulmonary/drug therapy , Iloprost/administration & dosage , Iloprost/pharmacology , Pulmonary Disease, Chronic Obstructive/complications , Administration, Inhalation , Adult , Aerosols , Aged , Cross-Over Studies , Female , Humans , Hypertension, Pulmonary/physiopathology , Iloprost/metabolism , Iloprost/therapeutic use , Male , Oxygen/metabolism , Pulmonary Gas Exchange/drug effects
9.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L645-55, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21856819

ABSTRACT

Prostaglandin E(2) (PGE(2)) is a lipid mediator that is produced via the metabolism of arachidonic acid by cyclooxygenase enzymes. In the lung, PGE(2) acts as an anti-inflammatory factor and plays an important role in tissue repair processes. Although several studies have examined the role of PGE(2) in the pathogenesis of pulmonary fibrosis in rodents, results have generally been conflicting, and few studies have examined the therapeutic effects of PGE(2) on the accompanying lung dysfunction. In this study, an established model of pulmonary fibrosis was used in which 10-12-wk-old male C57BL/6 mice were administered a single dose (1.0 mg/kg) of bleomycin via oropharyngeal aspiration. To test the role of prostaglandins in this model, mice were dosed, via surgically implanted minipumps, with either vehicle, PGE(2) (1.32 µg/h), or the prostacyclin analog iloprost (0.33 µg/h) beginning 7 days before or 14 days after bleomycin administration. Endpoints assessed at 7 days after bleomycin administration included proinflammatory cytokine levels and measurement of cellular infiltration into the lung. Endpoints assessed at 21 days after bleomycin administration included lung function assessment via invasive (FlexiVent) analysis, cellular infiltration, lung collagen content, and semiquantitative histological analysis of the degree of lung fibrosis (Ashcroft method). Seven days after bleomycin administration, lymphocyte numbers and chemokine C-C motif ligand 2 expression were significantly lower in PGE(2)- and iloprost-treated animals compared with vehicle-treated controls (P < 0.05). When administered 7 days before bleomycin challenge, PGE(2) also protected against the decline in lung static compliance, lung fibrosis, and collagen production that is associated with 3 wk of bleomycin exposure. However, PGE(2) had no therapeutic effect on these parameters when administered 14 days after bleomycin challenge. In summary, PGE(2) prevented the decline in lung static compliance and protected against lung fibrosis when it was administered before bleomycin challenge but had no therapeutic effect when administered after bleomycin challenge.


Subject(s)
Bleomycin/adverse effects , Collagen/biosynthesis , Dinoprostone/pharmacology , Iloprost/pharmacology , Lung/drug effects , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin/administration & dosage , Bronchoalveolar Lavage Fluid/cytology , Collagen/analysis , Cytokines/biosynthesis , Dinoprostone/metabolism , Disease Models, Animal , Drug Administration Schedule , Histocytochemistry , Humans , Iloprost/metabolism , Infusion Pumps, Implantable , Leukocyte Count , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Pulmonary Fibrosis/prevention & control , Real-Time Polymerase Chain Reaction , Severity of Illness Index
10.
Exp Biol Med (Maywood) ; 235(9): 1142-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20682601

ABSTRACT

Exposure of erythrocytes to reduced oxygen (O(2)) tension activates the heterotrimeric G-protein Gi, resulting in the accumulation of cyclic AMP (cAMP) and release of ATP. The mechanism by which exposure of erythrocytes to reduced O(2) tension activates Gi is not known. Here we investigate the hypothesis that, in rabbit erythrocytes, ATP release in response to exposure to reduced O(2) tension is linked to erythrocyte membrane deformability. If this hypothesis is correct, then decreasing the deformability of the erythrocyte membrane should decrease the release of ATP in response to reduced O(2) tension. We report that treating erythrocytes with diamide, a compound that decreases erythrocyte deformability, inhibits low O(2) tension-induced ATP release. Treating erythrocytes with diamide does not, however, interfere with cAMP accumulation or ATP release in response to a direct activator of Gi (mastoparan 7) or in response to receptor-mediated activation of Gs (the prostacyclin analog, iloprost). These results demonstrate that diamide (100 micromol/L) does not directly inhibit the signaling pathways for ATP release from rabbit erythrocytes and support the hypothesis that low O(2) tension-induced ATP release from these cells is linked to membrane deformability.


Subject(s)
Erythrocytes/metabolism , Oxygen/blood , Oxygen/metabolism , Adenosine Triphosphate/analogs & derivatives , Animals , Cell Membrane/metabolism , Cyclic AMP/blood , Cyclic AMP/metabolism , Diamide/metabolism , Erythrocyte Deformability/drug effects , Erythrocyte Membrane/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Iloprost/metabolism , Iloprost/pharmacology , Intercellular Signaling Peptides and Proteins , Male , Peptides , Rabbits , Signal Transduction/drug effects , Wasp Venoms
11.
Biomaterials ; 31(10): 2903-11, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20045181

ABSTRACT

Molecular conjugates comprising targeting ligands hold great promise for site-specific gene delivery to distant tumors and individual organs including the lung. Here we show that prostaglandin I2 analogues can be used to improve gene transfer efficiency of polyethylenimine (PEI) gene vectors on bronchial and alveolar epithelial cells in vitro and lungs of mice in vivo. Prostacyclin (IP1) receptor expression was confirmed in pulmonary epithelial cell lines by western blot. Iloprost (ILO) and treprostinil (TRP), two prostaglandin I2 analogues, were conjugated to fluorescein-labeled BSA (FLUO-BSA) and compared for IP1 receptor binding/uptake in different lung cell lines. Binding of FLUO-BSA-ILO was 2-4-fold higher than for FLUO-BSA-TRP and could be specifically inhibited by free ILO and IP1 receptor antagonist CAY10449. Internalization of FLUO-BSA-ILO was confirmed by confocal microscopy. Molecular conjugates of PEI and ILO (PEI-g-ILO) were synthesized with increasing coupling degree (F(ILO) (ILO:PEI) = 2, 5, 8, 16) and analyzed for DNA binding, particle formation and transfection efficiency. At optimized conditions (N/P 4, F(ILO) = 5), gene expression using PEI-g-ILO was significantly up to 46-fold higher than for PEI gene vectors and specifically inhibited by CAY10449. Gene expression in the lungs of mice after aerosol delivery was 14-fold higher with PEI-g-ILO F(ILO) = 5 than for PEI. We suggest that targeting of IP1 receptor using ILO represents a promising approach to improve pulmonary gene transfer.


Subject(s)
Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Lung/metabolism , Receptors, Epoprostenol/metabolism , Animals , Blotting, Western , Cattle , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fluorescein/metabolism , Genetic Vectors/genetics , Humans , Iloprost/metabolism , Iloprost/pharmacology , Ligands , Mice , Mice, Inbred BALB C , Organ Specificity/drug effects , Polyethyleneimine/metabolism , Reproducibility of Results , Serum Albumin, Bovine/metabolism , Transfection
12.
Arterioscler Thromb Vasc Biol ; 28(3): 534-40, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18162607

ABSTRACT

OBJECTIVE: Stimulation of protease-activated receptor-1 (PAR-1) by thrombin causes vascular smooth muscle cell (SMC) mitogenesis and has been implicated in the vascular response to injury. Vascular injury is also associated with enhanced formation of PGE2 and PGI2 (prostacyclin). This study investigates whether PGI2 and PGE2 modify the expression of PAR-1 and the cellular response to thrombin in human SMC. METHODS AND RESULTS: The PGI2-mimetic iloprost (1 to 100 nmol/L) attenuated mRNA, total protein, and cell surface expression of PAR-1. This was associated with inhibition of thrombin-induced mitogenesis and migration. Comparable inhibition of PAR-1 expression was observed with the selective IP-receptor agonist cicaprost, the adenylyl cyclase activator forskolin, the phosphodiesterase inhibitor isobutylmethylxanthine and the PKA activator dibutyryl-cAMP. Similar effects of PGE2 required micromolar concentrations. The specific PKA-inhibitor Myr-PKI prevented PAR-1 downregulation by iloprost. The potential role of Rho family GTPases in PAR-1 regulation was also investigated. Iloprost decreased Rac1 mRNA and the Rac1 inhibitor NSC23766 mimicked the inhibitory effects of iloprost on PAR-1 protein--but not mRNA. The Rho kinase inhibitor Y27632 did not influence PAR-1 expression. CONCLUSIONS: IP-receptor agonists may limit the mitogenic actions of thrombin in human SMC by downregulating PAR-1 via modulation of cAMP-/PKA- and Rac1-dependent signaling pathways.


Subject(s)
Epoprostenol/physiology , Iloprost/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, PAR-1/metabolism , Transcription, Genetic/physiology , Analysis of Variance , Blotting, Western , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Dinoprostone/pharmacology , Dinoprostone/physiology , Down-Regulation , Epoprostenol/pharmacology , Flow Cytometry , Gene Expression , Humans , Iloprost/pharmacology , Immunohistochemistry , Muscle, Smooth, Vascular/cytology , Probability , RNA, Messenger/analysis , Receptor, PAR-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saphenous Vein/cytology , Signal Transduction/genetics , Signal Transduction/physiology
13.
Biochem J ; 386(Pt 1): 177-89, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15469414

ABSTRACT

Like Ras, farnesylation of the IP (prostacyclin receptor) is required for its efficient intracellular signalling, and hence the IP represents a potential target for inhibition by FTIs [FTase (farnesyl protein transferase) inhibitors]. Herein, the effect of SCH66336 on the isoprenylation and function of the human and mouse IPs overexpressed in human embryonic kidney 293 cells, and by the IP endogenously expressed in human erythroleukaemia cells, was investigated. SCH66336 yielded concentration-dependent decreases in IP-mediated cAMP generation (IC50 0.27-0.62 nM), [Ca2+]i mobilization (IC50 26.6-48.3 nM) and IP internalization, but had no effect on signalling by the non-isoprenylated beta2 adrenergic receptor or b isoform of the TP (prostanoid thromboxane A2 receptor). Additionally, SCH66336 impaired IP-mediated crossdesensitization of TPa signalling (IC50 56.1 nM) and reduced farnesylation of the molecular chaperone protein HDJ-2 (IC50 3.1 nM). To establish whether farnesylation of the IP is inhibited and/or whether its 'CaaX motif' might undergo alternative geranylgeranylation in the presence of SCH66336, a series of chimaeric Ha (Harvey)-Ras fusions were generated by replacing its CaaX motif (-CVLS) with that of the IP (-CSLC) or, as controls, of Ki (Kirsten)-Ras 4B (-CVIM) or Rac 1 (-CVLL). Whereas SCH66336 had no effect on Ha-RasCVLL isoprenylation in vitro or in whole cells, it supported alternative geranylgeranylation of Ha-RasCVIM, but completely impaired isoprenylation of both Ha-RasCVLS and Ha-RasCSLC. These data confirm that the -CSLC motif of the IP is a direct target for inhibition by the FTI SCH66336, and in the presence of strong FTase inhibition, the IP does not undergo compensatory geranylgeranylation


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Epoprostenol/analogs & derivatives , Piperidines/pharmacology , Proline/analogs & derivatives , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Pyridines/pharmacology , Receptors, Epoprostenol/drug effects , Signal Transduction/drug effects , Adrenergic beta-Agonists/pharmacology , Amino Acid Motifs , Animals , Calcium Signaling/drug effects , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor/metabolism , Cyclic AMP/biosynthesis , Dose-Response Relationship, Drug , Endocytosis/drug effects , Epoprostenol/pharmacology , Farnesyltranstransferase , HSP40 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Humans , Iloprost/metabolism , Isoproterenol/pharmacology , Kidney , Leukemia, Erythroblastic, Acute/pathology , Mice , Mutagenesis, Site-Directed , Organophosphorus Compounds/metabolism , Proline/metabolism , Propanolamines/metabolism , Protein Prenylation/drug effects , Proto-Oncogene Proteins p21(ras)/chemistry , Receptors, Adrenergic, beta-2/drug effects , Receptors, Adrenergic, beta-2/genetics , Receptors, Epoprostenol/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/drug effects , Recombinant Fusion Proteins/metabolism , Transfection
14.
Arch Biochem Biophys ; 418(1): 25-33, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-13679079

ABSTRACT

In previous studies, we have determined the solution structure of the second extracellular loop (eLP(2)) of the human thromboxane A(2) receptor (TP) and identified the residues in the eLP(2) domain involved in ligand recognition, by using a combination of approaches including a constrained synthetic peptide, 2D NMR spectroscopy, and recombinant proteins. These findings led us to hypothesize that the specific ligand recognition sites may be localized in the eLP(2) for all the prostanoid receptors. To test this hypothesis, we have investigated the ligand recognition site for another prostanoid receptor, the prostacyclin receptor (IP), which mediates an opposite biological function compared to that of the TP receptor. The identification of the interaction between the IP receptor and its agonist, iloprost, was achieved with a constrained synthetic peptide mimicking the eLP(2) region of the receptor. The IP eLP(2) segment was designed and synthesized to form a constrained loop, using a homocysteine disulfide bond connecting the ends of the peptide, based on the distance predicted from the IP receptor model created by homology modeling using the crystal structure of bovine rhodopsin as a template. The evidence of the constrained IP eLP(2) interaction with iloprost was found by the identification of the conformational changes of the eLP(2) induced by iloprost using fluorescence spectroscopy, and was further confirmed by 1D and 2D 1H NMR experiments. In addition, the IP eLP(2)-induced structure of iloprost in solution was elucidated through a complete assignment of the 2D 1H NMR spectra for iloprost in the presence of the IP eLP(2) segment. In contrast, no ordered structure was observed in the 2D 1H NMR experiments for iloprost alone in solution. These studies not only identified that the eLP(2) segment of the IP receptor is involved in ligand recognition, but also solved the 3D solution structure of the bound-form of iloprost, which could be used to study the receptor-ligand interaction in structural terms.


Subject(s)
Iloprost/chemistry , Receptors, Prostaglandin/chemistry , Binding Sites , Humans , Iloprost/metabolism , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Receptors, Epoprostenol , Receptors, Prostaglandin/metabolism
15.
J Recept Signal Transduct Res ; 23(1): 83-97, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12680591

ABSTRACT

Mouse prostacyclin (mIP) receptors transiently expressed in Chinese hamster ovary (CHO) cells activated both adenylyl cyclase and phospholipase C, with a 33-fold preference for signaling through Gs. The prostacyclin (IP) receptor agonists cicaprost, iloprost, carbacyclin, and prostaglandin E1 showed a similar order of potency for activation of both signaling pathways in cells transiently transfected with the mIP and the chimeric prostacyclin/prostaglandin D2 (IPN-VII/DPC and IPN-V/DPVI-C) receptors. Substitution of the carboxyl-terminal tail of the prostacyclin receptor with the corresponding region of the mDP receptor (IPN-VII/DPC) produced a receptor with increased coupling to both Gs and Gq. However, this increased G-protein coupling was lost in the IPN-V/DPVI-C receptor. The observation that both these chimeric receptors can activate phospholipase C indicates that the carboxyl-terminal tail of the IP receptor is not entirely responsible for its ability to couple to Gq. Site-directed mutagenesis studies suggest that isoleucine at position 323 in the IPN-VII/DPC receptor plays an important role in mediating the increased potency of this chimeric receptor.


Subject(s)
Receptors, Epoprostenol/chemistry , Receptors, Epoprostenol/genetics , Receptors, Immunologic , Receptors, Prostaglandin/chemistry , Receptors, Prostaglandin/genetics , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , CHO Cells , Cricetinae , Enzyme Activation , Iloprost/metabolism , In Vitro Techniques , Isoleucine/chemistry , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Epoprostenol/metabolism , Receptors, Prostaglandin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transfection , Type C Phospholipases/metabolism
16.
J Biol Chem ; 278(6): 4250-7, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12446735

ABSTRACT

The human prostacyclin receptor is a seven-transmembrane alpha-helical G-protein coupled receptor, which plays important roles in both vascular smooth muscle relaxation as well as prevention of blood coagulation. The position of the native ligand-binding pocket for prostacyclin as well as other derivatives of the 20-carbon eicosanoid, arachidonic acid, has yet to be determined. Through the use of prostanoid receptor sequence alignments, site-directed mutagenesis, and the 2.8-A x-ray crystallographic structure of bovine rhodopsin, we have developed a three-dimensional model of the agonist-binding pocket within the seven-transmembrane (TM) domains of the human prostacyclin receptor. Upon mutation to alanine, 11 of 29 candidate residues within TM domains II, III, IV, V, and VII exhibited a marked decrease in agonist binding. Of this group, four amino acids, Arg-279 (TMVII), Phe-278 (TMVII), Tyr-75 (TMII), and Phe-95 (TMIII), were identified (via receptor amino acid sequence alignment, ligand structural comparison, and computer-assisted homology modeling) as having direct molecular interactions with ligand side-chain constituents. This binding pocket is distinct from that of the biogenic amine receptors and rhodopsin where the native ligands (also composed of a carbon ring and a carbon chain) are accommodated in an opposing direction. These findings should assist in the development of novel and highly specific ligands including selective antagonists for further molecular pharmacogenetic studies of the human prostacyclin receptor.


Subject(s)
Receptors, Prostaglandin/metabolism , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Humans , Iloprost/metabolism , Iloprost/pharmacology , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Receptors, Epoprostenol , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/chemistry , Receptors, Prostaglandin/genetics
17.
Hum Reprod ; 17(12): 3053-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12456602

ABSTRACT

BACKGROUND: It was recently discovered that prostacyclin constituted 40-50% of prostaglandins (PG) produced by minced human oviduct. It is well established that prostacyclin relaxes vascular smooth muscle, but whether oviductal smooth muscle synthesizes prostacyclin and whether its contraction is affected by prostacyclin remain unclear. METHODS: Smooth muscle microdissected from human oviducts was used for the study. The expression of prostacyclin synthase (PGIS) and prostacyclin receptor (IP) was confirmed by Western blot analysis. Metabolites of [(3)H]PGH(2) were analysed for prostacyclin. Functional coupling of IP to adenyl cyclase was assessed by the accumulation of intracellular cAMP upon prostacyclin challenge. The presence of saturable, specific binding sites for prostacyclin was confirmed by binding assay. The identity of IP was further confirmed by RT-PCR and nucleotide sequence analysis. Finally, the effects of prostacyclin on muscle contraction were studied. RESULTS: Human oviductal smooth muscle expresses functionally active PGIS and IP. The IP expressed is the same as that cloned from human lung tissue. The ED(50) of prostacyclin to increase intracellular cAMP was 16 nmol/l. Prostacyclin dose-dependently decreased the amplitude of muscle contraction. CONCLUSIONS: Human oviductal smooth muscle produces prostacyclin, which, in turn, decreases its contractility. Prostacyclin may regulate embryo transport.


Subject(s)
Epoprostenol/physiology , Fallopian Tubes/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Adenylyl Cyclases/metabolism , Binding Sites , Blotting, Western , Cells, Cultured , Cyclic AMP/metabolism , Cytochrome P-450 Enzyme System/analysis , Epoprostenol/metabolism , Epoprostenol/pharmacology , Female , Humans , Iloprost/metabolism , Intramolecular Oxidoreductases/analysis , Muscle Contraction/drug effects , Prostaglandin H2 , Prostaglandins H/metabolism , Receptors, Epoprostenol , Receptors, Prostaglandin/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tritium
18.
J Pharmacol Exp Ther ; 303(2): 741-5, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12388660

ABSTRACT

Iloprost is a potent prostacyclin analog, which has been shown to exert beneficial effects in several vascular disorders. Inhalation of aerosolized iloprost was found to cause selective pulmonary vasodilatation, and this approach is under current investigation for treatment of chronic pulmonary hypertension. The present study investigated pharmacokinetics and metabolism of aerosolized iloprost in isolated buffer-perfused rabbit lungs, compared with intravascular administration of the prostanoid. After buffer admixture of iloprost, a steady decline of perfusate concentrations of the intact prostanoid was noted (half-life approximately 3.5 h), mostly attributable to progressive metabolism to dinor- and tetranoriloprost. Inhaled iloprost rapidly entered the intravascular compartment, with peak buffer concentrations being noted after 30 min (bioavailability approximately 63%). Compared with infused iloprost, significantly more rapid metabolism to dinor- and tetranoriloprost was noted for iloprost administered via the inhalative route of application. However, the percentage of the nebulized agent that enters the intravascular space as intact iloprost displays the same clearance rate as directly perfusate-admixed prostanoid. We conclude that a high percentage of inhaled iloprost rapidly enters the intravascular compartment in intact rabbit lungs. The lung is capable of metabolizing iloprost via beta-oxidation, and more rapid appearance of dinor- and tetranoriloprost is noted for the inhalative as compared with the intravascular route of iloprost administration.


Subject(s)
Iloprost/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacokinetics , Administration, Inhalation , Aerosols , Animals , Bronchoalveolar Lavage Fluid , Buffers , Chromatography, High Pressure Liquid , Female , Iloprost/administration & dosage , Iloprost/metabolism , In Vitro Techniques , Infusions, Intravenous , Lung/metabolism , Male , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/metabolism , Rabbits
19.
J Biol Chem ; 277(18): 15439-44, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11854299

ABSTRACT

The human prostacyclin receptor (hIP) is a seven transmembrane-spanning G-protein-coupled receptor that plays an important role in vascular homeostasis. Recent genetic analyses (SNP database, NCBI) have revealed the first two polymorphisms within the coding sequence, V25M and R212H. Here we present structure-function characterizations of these polymorphisms at physiological pH (7.4) and at an acidic pH (6.8) that would be encountered during stress such as renal, respiratory, or heart failure. Through a series of competition binding and G-protein activation assays (measured by cAMP production), we determined that the V25M polymorph exhibited agonist binding and G-protein activation similar to wild-type receptor at normal pH (7.4). However, the R212H variant demonstrated a significant decrease in binding affinity at lower pH (R212H at pH 7.4, K(i) = 2.2 +/- 1.2 nm; pH 6.8 K(i) = 45.6 +/- 12.0 nm). The R212H polymorph also exhibited abnormal activation at both pH 7.4 and pH 6.8 (pH 7.4, R212H EC(50) = 2.8 +/- 0.5 nm versus wild-type hIP EC(50) = 0.5 +/- 0.1 nm; pH 6.8, R212H EC(50) = 3.2 +/- 1.6 nm versus wild-type hIP EC(50) = 0.5 +/- 0.2 nm). Polymorphisms of the human prostacyclin receptor potentially may be important predictors of disease progress during biological stressors such as acidosis in which urgent correction of bodily pH may be required to restore normal hemostasis and vasodilation. This study provides the mechanistic basis for further research into genetic risk factors and pharmacogenetics of cardiovascular disease associated with hIP.


Subject(s)
Polymorphism, Genetic , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding, Competitive , COS Cells , Chlorocebus aethiops , Cyclic AMP/pharmacology , GTP-Binding Proteins/metabolism , Genetic Variation , Humans , Hydrogen-Ion Concentration , Iloprost/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Receptors, Epoprostenol , Transfection
20.
Article in English | MEDLINE | ID: mdl-11993717

ABSTRACT

The objective of these studies was to characterize the effects of a broad range of prostanoid agonists upon the stimulation of cAMP production in National Cancer Bank (NCB-20; mouse neuroblastoma/hamster brain hybridoma) cells. The pharmacology of these functional responses in NCB-20 cells was compared with that of the classic endogenous IP receptor present on human platelets using [3H]-iloprost binding techniques. In both assay systems, agonists from the IP prostanoid class exhibited the highest affinities and functional potencies. Specific prostanoids exhibited the following rank order of potency (EC50 +/- SEM) in stimulating cAMP production in the NCB-20 cells: carbaprostacyclin (4.3 +/- 0.9 nM) = PGI2 (6.6 +/-1.5 nM) > iloprost (75+/-13 nM) > 11-deoxy PGE, (378+/-138 nM) > misoprostol (1,243+/-48) > PGE2 (3020+/-700 nM) > ZK-118182 (7265+/-455 nM). Iloprost wasthe most potent compound in the human platelet binding assay while prostanoidsfromthe DPand EP receptor classes showed modest affinity. These studies provide functional and binding information for a broad range of both natural and synthetic prostanoid receptor ligands at the endogenous IP receptor in two different cell types.


Subject(s)
Alprostadil/analogs & derivatives , Blood Platelets/metabolism , Dinoprost/analogs & derivatives , Receptors, Prostaglandin/metabolism , Alprostadil/metabolism , Alprostadil/pharmacology , Animals , Cyclic AMP/metabolism , Dinoprost/metabolism , Dinoprost/pharmacology , Dinoprostone/metabolism , Dinoprostone/pharmacology , Humans , Iloprost/metabolism , Iloprost/pharmacology , Mice , Misoprostol/metabolism , Misoprostol/pharmacology , Neuroblastoma/metabolism , Radioimmunoassay , Receptors, Epoprostenol , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...