Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters











Publication year range
1.
Eur J Pharmacol ; 946: 175579, 2023 May 05.
Article in English | MEDLINE | ID: mdl-36914083

ABSTRACT

Pulmonary hypertension (PH) is associated with pulmonary vasoconstriction and endothelial dysfunction leading to impaired nitric oxide (NO) and prostacyclin (PGI2) pathways. Metformin, the first line treatment for type 2 diabetes and AMP-activated protein kinase (AMPK) activator, has been recently highlighted as a potential PH treatment. AMPK activation has been reported to improve endothelial function by enhancing endothelial NO synthase (eNOS) activity and to have relaxant effects in blood vessels. In this study, we examined the effect of metformin treatment on PH as well as on NO and PGI2 pathways in monocrotaline (MCT)-injected rats with established PH. Moreover, we investigated the anti-contractile effects of AMPK activators on endothelium-denuded human pulmonary arteries (HPA) from Non-PH and Group 3 PH patients (due to lung diseases and/or hypoxia). Furthermore, we explored the interaction between treprostinil and the AMPK/eNOS pathway. Our results showed that metformin protected against PH progression in MCT rats where it reduced the mean pulmonary artery pressure, pulmonary vascular remodeling and right ventricular hypertrophy and fibrosis compared to vehicle-treated MCT rats. The protective effects on rat lungs were mediated in part by increasing eNOS activity and protein kinase G-1 expression but not through the PGI2 pathway. In addition, incubation with AMPK activators reduced the phenylephrine-induced contraction of endothelium-denuded HPA from Non-PH and PH patients. Finally, treprostinil also augmented eNOS activity in HPA smooth muscle cells. In conclusion, we found that AMPK activation can enhance the NO pathway, attenuate vasoconstriction by direct effects on smooth muscles, and reverse established MCT-induced PH in rats.


Subject(s)
Diabetes Mellitus, Type 2 , Hypertension, Pulmonary , Metformin , Rats , Humans , Animals , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/prevention & control , Pulmonary Artery , Metformin/adverse effects , AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Type 2/metabolism , Monocrotaline/adverse effects
2.
Prostaglandins Other Lipid Mediat ; 165: 106700, 2023 04.
Article in English | MEDLINE | ID: mdl-36528331

ABSTRACT

In patients with coronary artery disease (CAD), plasma levels of pro-inflammatory lipid mediators such as PGE2 and TxA2 are increased. They could increase vascular contraction while EPA and DHA could reduce it. Studies have been mostly conducted on animal vessels. Therefore, the aim of the study was to investigate if EPA, DHA, and DHA-derived metabolites: RvD1, RvD5 and MaR1 can modulate contraction of human coronary arteries (HCA) induced by PGE2 or TxA2 stable analogue (U46619). DHA and EPA relaxed HCA pre-contracted with PGE2. 18 h-incubation with DHA but not EPA reduced the PGE2-induced contractions. Pre-incubation with RvD1, RvD5 and MaR1 reduced the PGE2-induced contractions. Indomethacin did not significantly modify the PGE2 responses. L-NOARG (inhibitor of nitric oxide synthase), reduced only the PGE2-induced contractions in RvD1-treated rings. Finally, FPR2/ALX, GPR32 and LGR6 receptors are detected in HCA by immunofluorescence. Our results indicate that DHA and its metabolites could be beneficial for HCA blood flow and could be a therapeutic perspective for patients with CAD.


Subject(s)
Coronary Vessels , Dinoprostone , Animals , Humans , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Dinoprostone/pharmacology , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Thromboxane A2 , Eicosapentaenoic Acid
3.
J Cardiovasc Surg (Torino) ; 63(1): 69-77, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34472766

ABSTRACT

BACKGROUND: Vasospasm and atherosclerosis due to low endothelial capacity are the most important causes of coronary artery bypass graft failure observed in internal mammary artery (IMA) and saphenous vein (SV). Vasospasm can be mimicked in in-vitro studies by inducing vasoconstriction of graft materials. In the present study, we aimed to compare the vascular contraction induced by several spasmogens including prostaglandin E2 (PGE2), prostaglandin F2 alpha (PGF), phenylephrine (PE), leukotriene C4 (LTC4), LTD4, potassium chloride (KCl), and arachidonic acid between IMA and SV preparations. Furthermore, endothelial capacity, nitrite and asymmetric dimethylarginine (ADMA) levels were compared between two grafts. METHODS: By using organ bath, contractile responses induced by different spasmogens were compared between IMA and SV preparations derived from patients underwent coronary artery bypass surgery (N.=35). The endothelial capacity was determined by acetylcholine-induced (ACh) relaxation in PE-precontracted vessels. Nitrite and ADMA levels were measured in organ culture supernatant of IMA and SV preparations. RESULTS: Contractile responses induced by PGE2, PGF, PE, LTC4, LTD4, KCl and arachidonic acid were significantly lower in IMA preparations versus SV preparations. ACh-induced relaxation was significantly more prominent in IMA than SV preparations. Nitrite levels were greater and ADMA levels were lower in IMA versus SV preparations. CONCLUSIONS: IMA has reduced capacity to constrict to several vasoconstrictor agents. Furthermore, IMA has greater endothelial capacity associated with higher nitrite levels and lower ADMA levels. Our results support the greater patency rate observed in IMA versus SV preparations.


Subject(s)
Arginine/analogs & derivatives , Coronary Artery Bypass , Mammary Arteries/metabolism , Saphenous Vein/metabolism , Vasoconstriction , Arginine/metabolism , Biomarkers/metabolism , Humans , In Vitro Techniques , Mammary Arteries/drug effects , Saphenous Vein/drug effects , Vascular Patency , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
4.
Life Sci ; 286: 120073, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34688694

ABSTRACT

AIMS: Thromboxane (TxA2) is synthesized from arachidonic acid (AA) via thromboxane synthase (TxS) enzyme and induces vasoconstriction via TP receptor. Our aim is to compare the effects of aspirin, TxS inhibitor and TP receptor antagonist on vascular reactivity of bypass grafts (saphenous vein and internal mammary artery). MAIN METHODS: Using isolated organ bath, saphenous vein and internal mammary artery preparations were incubated with TP receptor antagonist, TxS inhibitor, aspirin, IP or EP4 receptor antagonist. Then prostaglandin (PG)E2, PGF2α, phenylephrine and AA were administered in concentration-dependent manner. The expression of prostanoid receptor and PGI2 synthase (PGIS) enzyme was determined by Western Blot. KEY FINDINGS: TP receptor antagonist inhibited the contraction induced by PGE2, PGF2α, and AA but not that induced by phenylephrine in both types of vessels. Aspirin increased phenylephrine-induced contraction only in internal mammary artery and decreased AA-induced contraction in saphenous vein. TxS inhibitor decreased both PGE2 and AA-induced contraction in both types of vessels. This decrease was reversed by co-incubation of TxS inhibitor and IP/EP4 receptor antagonists. The expressions of EP3 receptor and PGIS enzyme were greater in internal mammary artery compared to saphenous vein while IP and TP receptors expressed at similar levels. SIGNIFICANCE: TP receptor antagonist and TxS inhibitor are more effective to reduce contraction induced by different spasmogens in comparison to aspirin. Our results suggest that TP receptor antagonist and TxS inhibitor might have an advantage over aspirin due to their preventive effect on increased vascular reactivity observed in post-operative period of coronary artery bypass grafting.


Subject(s)
Mammary Arteries/drug effects , Saphenous Vein/drug effects , Arachidonic Acid/metabolism , Aspirin/pharmacology , Benzofurans/pharmacology , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Mammary Arteries/metabolism , Muscle, Smooth, Vascular/metabolism , Phenylephrine/pharmacology , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/drug effects , Receptors, Thromboxane/metabolism , Saphenous Vein/metabolism , Sulfonamides/pharmacology , Thromboxane A2/pharmacology , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxane-A Synthase/drug effects , Thromboxane-A Synthase/metabolism , Thromboxanes/antagonists & inhibitors , Thromboxanes/metabolism , Vasoconstriction/drug effects
5.
Sci Rep ; 11(1): 20052, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625635

ABSTRACT

The association between matrix metalloproteinases (MMPs), tissue inhibitor of metalloproteinases (TIMPs) and obesity as well as obesity-related disease including metabolic syndrome is not fully explored. Our aims are that: (i) to evaluate the plasma levels of MMP-1, MMP-2, MMP-3, MMP-9, TIMP-1, TIMP-2 and their ratios in non-obese people, overweight and obese people with or without metabolic syndrome, (ii) to investigate correlations between MMPs or TIMPs levels and several anthropometric parameters, blood pressure, endothelial function. Anthropometric and biochemical parameters were determined in 479 randomly selected participants, subdividing according to body mass index (BMI) and metabolic syndrome status. Plasma MMPs and TIMPs levels were measured. The assessment of endothelial function was characterized in people with obesity, overweight and non-obese, using laser Doppler Flowmetry. Obese people have elevated MMP-1, MMP-2, TIMP-1, TIMP-2 levels and decreased MMP-3/TIMP-1 and MMP-9/TIMP-1 ratios compared with non-obese people. MMP-1 levels and MMP-1/TIMP-1 ratio were positively correlated with BMI and waist circumference (WC) while MMP-2 levels were negatively correlated with BMI and WC values in obese people. MMP-3 levels and MMP-3/TIMP-1 ratio were positively correlated with systolic blood pressure (SBP) or diastolic blood pressure (DBP) in obese and metabolic syndrome people. Additionally, MMP-9 levels and MMP-9/TIMP-1 ratio were negatively correlated with endothelium-dependent response in obese and metabolic syndrome people. MMP-1, MMP-2, TIMP-1, TIMP-2 levels were increased in obese subjects. Significant correlations between anthropometric parameters and MMP-1 as well as MMP-1/TIMP-1 ratio supported these results. MMP-3 and -9 levels as well as their ratios with TIMP-1 were associated with blood pressure and endothelial-dependent response, respectively. In conclusion, our results demonstrated that MMP-1, MMP-3 and MMP-9 levels were correlated with several obesity-related parameters including BMI, WC, blood pressure and endothelial-dependent response. Our findings will hopefully provide new aspects for the use of MMPs and TIMPs as clinical biomarkers in obesity-related cardiovascular diseases such as metabolic syndrome and hypertension. The lack of measure of MMPs activity in plasma and relevant organs/tissues in obesity and metabolic syndrome is considered as a limitation in this report.


Subject(s)
Blood Pressure , Body Mass Index , Endothelium, Vascular/physiopathology , Matrix Metalloproteinase Inhibitors/metabolism , Matrix Metalloproteinases/metabolism , Obesity/pathology , Adult , Case-Control Studies , Female , Humans , Male , Obesity/metabolism
6.
Soft Matter ; 17(32): 7585-7595, 2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34341819

ABSTRACT

Mucus is a viscoelastic gel secreted by the pulmonary epithelium in the tracheobronchial region of the lungs. The coordinated beating of cilia moves mucus upwards towards the pharynx, removing inhaled pathogens and particles from the airways. The efficacy of this clearance mechanism depends primarily on the rheological properties of mucus. Here we use magnetic wire based microrheology to study the viscoelastic properties of human mucus collected from human bronchus tubes. The response of wires between 5 and 80 µm in length to a rotating magnetic field is monitored by optical time-lapse microscopy and analyzed using constitutive equations of rheology, including those of Maxwell and Kelvin-Voigt. The static shear viscosity and elastic modulus can be inferred from low frequency (3 × 10-3-30 rad s-1) measurements, leading to the evaluation of the mucin network relaxation time. This relaxation time is found to be widely distributed, from one to several hundred seconds. Mucus is identified as a viscoelastic liquid with an elastic modulus of 2.5 ± 0.5 Pa and a static viscosity of 100 ± 40 Pa s. Our work shows that beyond the established spatial variations in rheological properties due to microcavities, mucus exhibits secondary inhomogeneities associated with the relaxation time of the mucin network that may be important for its flow properties.


Subject(s)
Magnetics , Mucus , Humans , Magnetic Phenomena , Rheology , Viscosity
7.
Article in English | MEDLINE | ID: mdl-34403986

ABSTRACT

INTRODUCTION: Prostacyclin (PGI2) is synthetized by PGI2 synthase (PGIS) and induces vasorelaxation via activation of cyclic AMP (cAMP) generating IP-receptor. Several components of the PGI2 signaling pathway are reduced in patients with pulmonary hypertension (PH). AIM: To study the effect of 17ß-estradiol (E2) on the PGI2 signaling pathway in human pulmonary arteries (HPA) and in their smooth muscle cells (hPASMC) derived from Group-3 PH and non-PH patients. METHODS: Following E2-treatments of isolated HPA and cultured hPASMC, we measured: 6-keto-Prostaglandin F1α (PGI2 stable metabolite) by ELISA, PGIS and IP protein levels by Western blot and HPA vasorelaxations with an organ bath system. RESULTS: Incubation with E2 (24/48 h, doses ≥ 10 nM) significantly increased the expression of PGIS in hPASMC derived from both PH (65-98%) and non-PH (21-33%) patients, whereas incubation with E2 (2 h, 0.1 and 1 µM) increased 6-keto-PGF1α production in HPA from Group-3 PH patients only, and did not affect 6-keto-PGF1α production in hPASMC from either non-PH or Group-3 PH patients. Increases in IP receptor expression were observed following 10 mM E2-treatment of hPASMC from non-PH (33% after 48 h) and Group-3 PH (23% after 24 h) patient lungs. Finally, preincubation with 100 nM E2 significantly increased arachidonic acid-induced vasorelaxation of HPA from non-PH patient lungs but not of HPA from Group-3 PH patient lungs. CONCLUSION: E2-treatment may help to restore the PGI2-pathway in Group-3 PH.


Subject(s)
6-Ketoprostaglandin F1 alpha/metabolism , Cytochrome P-450 Enzyme System/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Hypertension, Pulmonary/metabolism , Intramolecular Oxidoreductases/drug effects , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , Vasodilation/drug effects , Antihypertensive Agents/pharmacology , Arachidonic Acid/pharmacology , Case-Control Studies , Cytochrome P-450 Enzyme System/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Female , Humans , Hypertension, Pulmonary/physiopathology , Intramolecular Oxidoreductases/metabolism , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology
8.
Biochem Genet ; 59(6): 1457-1486, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33929697

ABSTRACT

The purpose of this study was to determine the impact of six PGHS-2 genetic variants on obesity development and microvascular dysfunction. The study included 305 Tunisian subjects (186 normal weights, 35 overweights and 84 obeses). PCR analyses were used for allelic discrimination between polymorphisms. Prostaglandin (PGE2, PGI2), leptin, and matrix metalloproteinase (MMP1, 2, 3, 9) levels were evaluated by ELISA. Fatty acid composition was performed by gas chromatography-mass spectrometry. Our results revealed that subjects carrying the PGHS-2 306CC (rs5277) and 8473CC (rs5275) genotypes present higher anthropometric values compared to wild-type genotypes (306GG, BMI (Kg/m2): 27.11 ± 0.58; WC (cm): 93.09 ± 1.58; 306CC, BMI: 33.83 ± 2.46; WC: 109.93 ± 5.41; 8473TT, BMI: 27.75 ± 0.68; WC: 93.96 ± 1.75; 8473CC, BMI: 33.72 ± 2.2; WC: 117.89 ± 2.94). A reduced microvascular reactivity and a higher PGE2 level were also found in individuals with the 306CC and 8473CC genotypes in comparison to 306GG and 8473TT carriers (306GG, Peak Ach-CVC (PU/mmHg): 0.46 ± 0.03; PGE2 (pg/ml): 7933.1 ± 702; 306CC, Peak Ach-CVC: 0.24 ± 0.01; PGE2: 13,380.3 ± 966.2; 8473TT, Peak Ach-CVC: 0.48 ± 0.05; PGE2: 7086.41 ± 700.31; 8473CC, Peak Ach-CVC: 0.23 ± 0.01; PGE2: 13,175.7 ± 1165.8). Fatty acid analysis showed a significant increase of palmitic acid (PA) (34.2 ± 2.09 vs. 16.82% ± 1.76, P < 0.001), stearic acid (SA) (25.76 ± 3.29 vs. 9.05% ± 2.53, P < 0.001), and linoleic acid (LA) (5.25 ± 1.18 vs. 0.5% ± 0.09, P < 0.001) levels in individuals carrying the PGHS-2 306CC genotype when compared to GG genotype individuals. Subjects with the 8473CC genotype showed also a significant increase of PA, SA ,and LA levels when compared to TT genotype carriers (PA: 38.02 ± 1.51 vs. 12.65% ± 1.54, P < 0.001; SA: 32.96 ± 1.87 vs. 1.38% ± 0.56, P < 0.001; LA: 26.84 ± 2.09 vs. 3.7% ± 1.54, P < 0.001). Logistic regression analysis revealed that PGHS-2 306CC and 8473CC variants are significantly associated with obesity status (OR 6.25, CI (1.8-21.6), P = 0.004; OR 3.01, CI (1.13-8.52), P = 0.03, respectively). Haplotypes containing the C306:T8473 (OR 2.91; P = 0.01) and G306:C8473 (OR 5.25; P = 0.002) combinations were associated with an enhanced risk for obesity development in the studied population. In conclusion, our results highlight that PGHS-2 306G/C and 8473T/C variants could be useful indicators of obesity development, inflammation, and microvascular dysfunction among Tunisians.


Subject(s)
Dinoprostone , Obesity , Cyclooxygenase 2/genetics , Genetic Markers , Humans , Obesity/genetics , Prostaglandin H2
9.
Article in English | MEDLINE | ID: mdl-33545665

ABSTRACT

We hypothesized that polymorphisms of genes involved in the prostaglandin pathway could be associated with COPD. In this study we explored the involvement of genetic polymorphisms in PTGS2, PTGER2 and PTGER4 genes in the development and severity of COPD and their effects on plasma concentrations of inflammatory/oxidative stress markers. We identified genotypes of PTGS2, PTGER2 and PTGER4 SNPs in a Tunisian cohort including COPD patients (n = 138) and control subjects (n = 216) using PCR-RFLP and PCR TaqMan. Pulmonary function (FEV1 and FVC) were assessed by plethsmography. PGE2, PGD2 and cytokine plasma (IL-6, IL-18, TNF-α, TGF-ß) concentrations were measured using ELISA and colorimetric standard methods were used to determine oxidative stress concentrations. Genotype frequencies of rs2745557 in PTGS2 and rs2075797 in PTGER2 were different between COPD cases and controls. There was no correlation between these polymorphisms and lung function parameters. For rs2745557, the A allele frequency was higher in COPD cases than in controls. For rs2075797, carriers of the GG genotype were more frequent in the COPD group than in controls. Only rs2745557 in PTGS2 had an effect on PGD2 and cytokine plasma concentrations. PGD2 was significantly decreased in COPD patients with the GA or AA genotypes. In contrast, IL-18 and NO plasma concentrations were increased in COPD rs2745557 A allele carriers as compared to homozygous GG subjects. Our findings suggest that rs2745557 in PTGS2 and rs2075797 in PTGER2 are associated with COPD development but not with its severity.


Subject(s)
Cyclooxygenase 2/genetics , Polymorphism, Single Nucleotide , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/genetics , Receptors, Prostaglandin E, EP2 Subtype/genetics , Aged , Alleles , Case-Control Studies , Cohort Studies , Cytokines/blood , Dinoprostone/blood , Female , Gene Frequency , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Patient Acuity , Prostaglandin D2/blood , Pulmonary Disease, Chronic Obstructive/blood , Receptors, Prostaglandin E, EP4 Subtype/genetics , Risk Factors , Tunisia/epidemiology
10.
Pharmacol Rev ; 72(4): 910-968, 2020 10.
Article in English | MEDLINE | ID: mdl-32962984

ABSTRACT

Prostaglandins are derived from arachidonic acid metabolism through cyclooxygenase activities. Among prostaglandins (PGs), prostacyclin (PGI2) and PGE2 are strongly involved in the regulation of homeostasis and main physiologic functions. In addition, the synthesis of these two prostaglandins is significantly increased during inflammation. PGI2 and PGE2 exert their biologic actions by binding to their respective receptors, namely prostacyclin receptor (IP) and prostaglandin E2 receptor (EP) 1-4, which belong to the family of G-protein-coupled receptors. IP and EP1-4 receptors are widely distributed in the body and thus play various physiologic and pathophysiologic roles. In this review, we discuss the recent advances in studies using pharmacological approaches, genetically modified animals, and genome-wide association studies regarding the roles of IP and EP1-4 receptors in the immune, cardiovascular, nervous, gastrointestinal, respiratory, genitourinary, and musculoskeletal systems. In particular, we highlight similarities and differences between human and rodents in terms of the specific roles of IP and EP1-4 receptors and their downstream signaling pathways, functions, and activities for each biologic system. We also highlight the potential novel therapeutic benefit of targeting IP and EP1-4 receptors in several diseases based on the scientific advances, animal models, and human studies. SIGNIFICANCE STATEMENT: In this review, we present an update of the pathophysiologic role of the prostacyclin receptor, prostaglandin E2 receptor (EP) 1, EP2, EP3, and EP4 receptors when activated by the two main prostaglandins, namely prostacyclin and prostaglandin E2, produced during inflammatory conditions in human and rodents. In addition, this comparison of the published results in each tissue and/or pathology should facilitate the choice of the most appropriate model for the future studies.


Subject(s)
Receptors, Prostaglandin E/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Dinoprostone/immunology , Dinoprostone/metabolism , Epoprostenol/immunology , Epoprostenol/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Mice , Polymorphism, Single Nucleotide , Protein Multimerization , Rats , Receptors, Prostaglandin E/chemistry , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/immunology , Species Specificity
11.
Article in English | MEDLINE | ID: mdl-32673988

ABSTRACT

Pulmonary hypertension (PH) is a progressive and life-threating lung disorder characterized by elevated pulmonary artery pressure and vascular remodeling. PH is classified into five groups, and one of the most common and lethal forms, PH Group-III is defined as PH due to lung diseases and/or hypoxia. Due to the lack of studies in this group, PH-specific drug therapies including prostacyclin (PGI2) analogues have not been approved or recommended for use in these patients. PGI2 is synthesized by the PGI2 synthase (PGIS) enzyme, and its production is determined by measuring its stable metabolite, 6-keto-PGF1α. An impaired PGI2 pathway has been observed in PH animal models and in PH Group-I patients; however, there are contradictory results. The aim of this study is to determine whether PH Group-III is associated with altered expression of PGIS and production of PGI2 in humans. To explore this hypothesis, we measured PGIS expression (by western blot) and PGI2 production (by ELISA) in a large variety of preparations from the pulmonary circulation including human pulmonary artery, pulmonary vein, distal lung tissue, pulmonary artery smooth muscle cells (hPASMC), and bronchi in PH Group-III (n = 35) and control patients (n = 32). Our results showed decreased PGIS expression and/or 6-keto-PGF1α levels in human pulmonary artery, hPASMC, and distal lung tissue derived from PH Group-III patients. Moreover, the production of 6-keto-PGF1α from hPASMC positively correlated with PGIS expression and was inversely correlated with mean pulmonary artery pressure. On the other hand, PH Group-III pulmonary veins and bronchi did not show altered PGI2 production compared to controls. The deficit in PGIS expression and/or PGI2 production observed in pulmonary artery and distal lung tissue in PH Group-III patients may have important implications in the pathogenesis and treatment of PH Group-III.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/metabolism , Hypertension, Pulmonary/metabolism , Intramolecular Oxidoreductases/metabolism , Pulmonary Artery/metabolism , Bronchi/enzymology , Bronchi/metabolism , Cell Hypoxia/physiology , Cells, Cultured , Dinoprost/metabolism , Down-Regulation , Female , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Lung/enzymology , Lung/metabolism , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/enzymology , Pulmonary Veins/enzymology , Pulmonary Veins/metabolism
12.
Prostaglandins Other Lipid Mediat ; 151: 106476, 2020 12.
Article in English | MEDLINE | ID: mdl-32721526

ABSTRACT

Saphenous vein (SV) is one of the most widely used graft material in patients undergoing coronary artery bypass graft surgery (CABG). Thromboxane A2 (TXA2) is implicated in graft failure by inducing vasoconstriction and platelet aggregation. The aim of this study is to investigate the mechanism involved in TXA2-induced vasoconstriction in human SV. The role of different inhibitors and blockers on U46619 (TXA2-mimetic)-induced vasoconstriction is investigated by using an isolated organ bath system. Relaxation responses to several mediators are evaluated in SV pre-contracted with U46619 and compared with those pre-contracted with phenylephrine. Our results demonstrate that U46619-induced contraction is completely blocked by myosin light chain kinase inhibitor ML-9 or TP receptor antagonist BAY u3405. Furthermore, U46619-induced contraction is partially inhibited by phospholipase C inhibitor U73122, protein kinase C inhibitor calphostin C, Rho-kinase inhibitor Y-27632, L-type calcium channel blocker nifedipine, store-operated channel inhibitor SKF96365 or removal of extracellular calcium. Relaxation responses to NO donor (sodium nitroprusside), guanylate cyclase (GC) stimulator (riociguat), phosphodiesterase (PDE) inhibitors (sildenafil, IBMX), adenylate cyclase (AC) activator (forskolin) and acetylcholine (ACh) are markedly reduced when U46619 is used as a pre-contraction agent. Our results demonstrate that influx of extracellular Ca2+ (through L-type calcium channels and store-operated calcium channels) and intracellular Ca2+ release together with Ca2+ sensitization (through Rho-kinase activation) are necessary components for TXA2-induced vasoconstriction in SV. Moreover, more pronounced decrease in vasorelaxation induced by several mediators (SNP, riociguat, sildenafil, IBMX, forskolin, and ACh) in the presence of U46619 when compared with phenylephrine suggests that there is a crosstalk between the TP receptor signaling pathway and PDE, AC, GC enzymes. We believe that the investigation of mechanism of the TXA2-induced vasoconstriction in SV will provide additional information for the prevention of SV graft failure.


Subject(s)
Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Saphenous Vein/physiology , Vasoconstriction , Humans , Male , Saphenous Vein/metabolism , Thromboxane A2/metabolism , Vasodilation
14.
Prostaglandins Other Lipid Mediat ; 146: 106388, 2020 02.
Article in English | MEDLINE | ID: mdl-31672620

ABSTRACT

Pulmonary hypertension (PH) is characterized by an elevation of mean pulmonary artery pressure and it is classified into five groups. Among these groups, PH Group-III is defined as PH due to lung disease or hypoxia. Prostacyclin (PGI2) analogues (iloprost, treprostinil) and endothelin-1 (ET-1) receptor antagonists (ERA) (used alone or in combination) are therapies used for treating PH. The mechanisms underlying the positive/negative effects of combination treatment are not well documented, and in this study, we tested the hypothesis that the combination of a PGI2 analogue (iloprost, treprostinil) and an ERA may be more effective than either drug alone to treat vasculopathies observed in PH Group-III patients. Using Western blotting, ETA and ETB receptor expression were determined in human pulmonary artery (HPA) preparations derived from control and PH Group-III patients, and the physiologic impact of altered expression ratios was assessed by measuring ET-1 induced contraction of ex vivo HPA and human pulmonary veins (HPV) in an isolated organ bath system. In addition, the effects of single agent or combination treatments with a PGI2 analogue and an ERA on ET-1 release and HPA smooth muscle cells (hPASMCs) proliferation were determined by ELISA and MTT techniques, respectively. Our results indicate that the increased ETA/ETB receptor expression ratio in HPA derived from PH Group-III patients is primarily governed by a greatly depressed ETB receptor expression. However, contractions induced by ET-1 are not impacted in HPA and HPV derived from PH Group-III patients as compared to controls. Also, we found that the combination of an ETA receptor antagonist (BQ123) with iloprost provides greater inhibition of hPASMCs proliferation (-48±14% control; -32±06% PH) than either agent alone. Of note, while the ETB receptor antagonist (BQ788) increases ET-1 production from PH Group-III patients' preparations (HPA, parenchyma), even under these more proliferative conditions, iloprost and treprostinil are still effective to inhibit hPASMCs proliferation (-22/-24%). Our findings may provide new insights for the treatment of PH Group-III by combining a PGI2 analogue and a selective ETA receptor antagonist.


Subject(s)
Endothelin-1/metabolism , Epoprostenol/metabolism , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Signal Transduction , Aged , Endothelin-1/pharmacology , Epoprostenol/pharmacology , Female , Humans , Hypertension, Pulmonary/pathology , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Veins/metabolism , Pulmonary Veins/pathology , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism
15.
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
16.
Biochem Pharmacol ; 166: 242-252, 2019 08.
Article in English | MEDLINE | ID: mdl-31158340

ABSTRACT

BACKGROUND AND PURPOSE: Beraprost is a prostacyclin analogue and IP receptor agonist which is approved to treat pulmonary arterial hypertension (PAH) in Asia. The beraprost-314d isomer (esuberaprost) is one of four stereoisomers contained within the racemic mixture of beraprost. The pharmacological profile of esuberaprost is now evaluated to determine how stereoisomer separation affects its potency and mode of action in functional assays. EXPERIMENTAL APPROACH: Vascular tone was assessed using wire myography in rat and human distal pulmonary arteries (PAs) pre-contracted with U46619 (100 nM). HEK-293 cells stably expressing the human IP receptor (HEK-293-IP) and pulmonary arterial smooth muscle cells (PASMCs) derived from PAH patients were used to assess cyclic AMP (cAMP) generation and cell proliferation, respectively. KEY RESULTS: Esuberaprost relaxed rat PAs with a 5-fold greater potency compared with beraprost, and effects were strongly inhibited by RO3244794 (IP receptor antagonist) or L-NAME (NO synthase inhibitor). Esuberaprost caused EP3 receptor-dependent vasoconstriction at high concentrations ≥ 1000 nM, but contractions were 50% lower compared to beraprost. In HEK-293-IP cells, esuberaprost was 26-fold more potent (EC50 0.4 nM) at increasing cAMP than beraprost. In human PASMCs, esuberaprost was 40-fold more potent than beraprost at inhibiting cell proliferation (EC50 3 nM versus 120 nM), contrasting the 5-fold potency difference for cAMP elevation. Antiproliferative effects of esuberaprost appeared more dependent on NO than on the IP receptor. In PAs from patients with pulmonary hypertension, esuberaprost, caused some relaxation whereas beraprost instead produced a weak contraction. CONCLUSIONS AND IMPLICATIONS: Stereoisomer separation of beraprost has a significant effect on the pharmacology of the individual isomer, esuberaprost, identified in vitro as a highly potent prostanoid IP receptor agonist.


Subject(s)
Epoprostenol/analogs & derivatives , Hypertension, Pulmonary/drug therapy , Muscle, Smooth, Vascular/drug effects , Receptors, Epoprostenol/agonists , Receptors, Epoprostenol/antagonists & inhibitors , Vasodilator Agents/pharmacology , Animals , Cell Proliferation , Cells, Cultured , Dose-Response Relationship, Drug , Epoprostenol/chemistry , Epoprostenol/pharmacology , Epoprostenol/therapeutic use , Female , HEK293 Cells , Humans , Hypertension, Pulmonary/physiopathology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Rats , Rats, Sprague-Dawley , Receptors, Epoprostenol/physiology , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/chemistry , Vasodilator Agents/therapeutic use
17.
Pharmacol Rep ; 71(4): 659-668, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31195343

ABSTRACT

BACKGROUND: Sildenafil (PDE5-inhibitor) and alprostadil (PGE1) are used in combination clinically for the management of some cases of erectile dysfunction. Despite the roles of prostaglandins (PG) and nitric oxide (NO) pathways in contractility of bladder smooth muscle are frequently studied, the effect of sildenafil/alprostadil combination and the crosstalk between NO/cGMP and PG pathways on bladder activity is not documented. METHODS: Organ-bath experiments were performed using isolated rat detrusor muscle. Direct and neurogenic contractions were induced using ACh and electric stimulation (EFS, 4Hz, 80V, 1ms), respectively. The contractile responses in absence and presence of the tested drugs at different concentrations were compared. Results are expressed as mean ± SEM (n = 5-7). RESULTS: Alprostadil (0.01-10 µM) concentration-dependently potentiated ACh (100µM)- and EFS (4 Hz)- induced contraction. Maximum potentiation of ACh-contraction in presence of alprostadil was 40 ± 5%. Sildenafil potentiated ACh-induced contraction at low concentrations (0.01-1 µM), but inhibited it at higher ones (10-100 µM). IBMX (non-selective PDE-inhibitor, 0.01-100µM) and SNP (NO-donor, 1nM-1 mM) produced the same biphasic pattern. The potentiatory phase of sildenafil was inhibited by atropine (0.1µM), L-NAME (non-selective NOS-inhibitor, 100µM), N-PLA (nNOS-inhibitor, 30µM) or MB (nonselective GC-inhibitor, 10µM). In presence of sildenafil (0.1µM), the concentration-response curve of alprostadil (0.01-10µM) on both ACh and EFS-induced contraction was clearly shifted downward. CONCLUSIONS: A crosstalk between PGE1 and NO/cGMP pathways may exist. At low concentrations only, the effect of sildenafil on bladder contractility is dependent on NO/cGMP. cGMP intracellularly-elevated by sildenafil, may inhibit the activity of PLC and hence the cascade of EP1-receptors, thus masking the hyperactivity of bladder caused by alprostadil, which adds to the advantages of this combination.


Subject(s)
Alprostadil/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Sildenafil Citrate/pharmacology , Urinary Bladder/drug effects , Animals , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Electric Stimulation , In Vitro Techniques , Male , Muscle, Smooth/metabolism , Muscle, Smooth/physiopathology , Nitric Oxide/metabolism , Rats, Wistar , Receptor Cross-Talk , Signal Transduction , Urinary Bladder/metabolism , Urinary Bladder/physiopathology
18.
Biomed Pharmacother ; 111: 1458-1466, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30841461

ABSTRACT

INTRODUCTION: Prostaglandins (PGs) play an important role in corpus cavernosum relaxation, as evidenced by alprostadil being used as a drug for erectile dysfunction. Reports about the effect of cyclooxygenase (COX) inhibitors on erectile function are highly contradictory. AIM: To compare the potential effects of some COX inhibitors with varying COX-1/COX-2 selectivities (indomethacin, ketoprofen and diclofenac) with that of the selective COX-2 inhibitor (DFU) on corpus cavernosal tone in-vitro. The role played by PGE1, PGI2-analogue and PGE4 receptor (EP4)-agonist in controlling corpus cavernosum function and the modulation of their action by sildenafil is also studied. METHODS: Organ bath experiments were performed using isolated rat corpus cavernosum. Direct relaxations and changes to electric field stimulation (EFS, 2-16 Hz, 60 V, 0.8 ms, 10 s train)-induced relaxation by the effect of the selected drugs were studied. Strips were precontracted using phenylephrine (PE, 10-5 M). Results are expressed as mean ± SEM of 5-9 rats. RESULTS: Alprostadil, iloprost and L902688 (selective EP4 agonist) induced direct relaxation where L902688 showed greater relaxant effect. Sildenafil potentiated the Emax of alprostadil and iloprost but not L902688. EFS and acetylcholine (ACh)-induced relaxations were significantly potentiated in presence of indomethacin, ketoprofen and diclofenac (20, 100 µM) but not in presence of selective COX-2 inhibitor (DFU, 1 µM). GR32191B (Thromboxane A2 receptor antagonist, 10-6 M) significantly reduced the potentiatory effect of indomethacin. Only diclofenac succeeded to potentiate sodium nitroprusside (SNP)-induced relaxation. CONCLUSIONS: EP4 receptors may play an important nitric oxide (NO)/cGMP-independent role in corpus cavernosal relaxation. Nonselective COX inhibitors seem of no harm concerning cavernosal tissue relaxation, possibly because they inhibit the synthesis of the highly contracting mediator thromboxane A2.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Muscle Relaxation/drug effects , Penis/drug effects , Prostaglandins/metabolism , Alprostadil/pharmacology , Animals , Cyclic GMP/metabolism , Diclofenac/pharmacology , Erectile Dysfunction/drug therapy , Erectile Dysfunction/metabolism , Iloprost/pharmacology , Indomethacin/pharmacology , Ketoprofen/pharmacology , Male , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Penile Erection/drug effects , Penis/metabolism , Piperazines/pharmacology , Rats , Sildenafil Citrate/pharmacology , Sulfones/pharmacology
19.
Naunyn Schmiedebergs Arch Pharmacol ; 392(4): 437-450, 2019 04.
Article in English | MEDLINE | ID: mdl-30552456

ABSTRACT

Various studies have confirmed that prostaglandins (PG) alter the bladder motor activity and micturition reflex in both human and animals. However, no sufficient data is reported about the effect of cyclooxygenase (COX) inhibitors neither in normal bladder physiology nor in pathological conditions. This study aims to compare the potential effects of some COX inhibitors with varying COX-1/COX-2 selectivities (indomethacin, ketoprofen, and diclofenac) with that of the selective COX-2 inhibitor (DFU) on bladder function. The role played by some PGs and their receptors in controlling detrusor muscle function in normal condition and in cystitis is also studied. Organ bath experiments were performed using isolated rat detrusor muscle. Direct and neurogenic contractions were induced using ACh and electric stimulation (EFS), respectively. A model of hemorrhagic cystitis was induced by single injection of cyclophosphamide (300 mg/kg) in rats, and confirmed by histophathological examination. Results are expressed as mean ± SEM of 5-9 rats. Alprostadil and iloprost (1 nM- 10 µM) concentration-dependently potentiated ACh (100 µM)- and EFS (4 Hz)-induced contraction, with maximum potentiation of 40.01 ± 5.29 and 27.59 ± 6.64%, respectively, in case of ACh contractions. In contrast, ONO-AE1-259 (selective EP2 agonist, 1 nM-10 µM) inhibited muscle contraction. SC51322 (EP1-antagonist, 10 µM) and RO1138452 (IP antagonist, 10 µM) inhibited both direct and neurogenic responses. Hemorrhagic cystitis reduced both ACh and EFS responses as well as the potentiatory effect of iloprost and the inhibitory effect of RO1138452 on ACh contractions. ONO-AE3-237 (DP1 antagonist, 1 µM) significantly potentiated contractions in cystitis but showed no effect in normal bladder. A significant inhibition of contractile response was observed in presence of indomethacin, ketoprofen, and diclofenac at all tested concentrations (20, 50, and 100 µM). Highest effect was induced by diclofenac. The effect of these COX inhibitors on EFS contractions was intensified in case of cystitis, indomethacin being the most potent. Atropine (1 nM) significantly reduced indomethacin effect on ACh contraction only in normal rats. On the other hand, DFU (10-6 M) significantly potentiated the contractile effect of ACh in case of cystitis although it showed no effect in normal rats. EP1 receptors seem to play an important role in rat bladder contractility. DP1 receptors as COX-2, on the other hand, gain an important role only in case of cystitis. The use of non-selective COX inhibitors in cystitis may be associated with bladder hypoactivity; selective COX-2 inhibitors may be a safer option.


Subject(s)
Cyclooxygenase Inhibitors/adverse effects , Cystitis/pathology , Muscle, Smooth/drug effects , Receptors, Prostaglandin/antagonists & inhibitors , Urinary Bladder/drug effects , Animals , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Cyclophosphamide , Cystitis/chemically induced , Cystitis/physiopathology , Disease Models, Animal , Male , Muscle Contraction/drug effects , Muscle, Smooth/pathology , Muscle, Smooth/physiology , Rats, Wistar , Receptors, Prostaglandin/physiology , Urinary Bladder/pathology , Urinary Bladder/physiology
20.
Int J Mol Sci ; 19(8)2018 Aug 12.
Article in English | MEDLINE | ID: mdl-30103548

ABSTRACT

Prostacyclins are extensively used to treat pulmonary arterial hypertension (PAH), a life-threatening disease involving the progressive thickening of small pulmonary arteries. Although these agents are considered to act therapeutically via the prostanoid IP receptor, treprostinil is the only prostacyclin mimetic that potently binds to the prostanoid EP2 receptor, the role of which is unknown in PAH. We hypothesised that EP2 receptors contribute to the anti-proliferative effects of treprostinil in human pulmonary arterial smooth muscle cells (PASMCs), contrasting with selexipag, a non-prostanoid selective IP agonist. Human PASMCs from PAH patients were used to assess prostanoid receptor expression, cell proliferation, and cyclic adenosine monophosphate (cAMP) levels following the addition of agonists, antagonists or EP2 receptor small interfering RNAs (siRNAs). Immunohistochemical staining was performed in lung sections from control and PAH patients. We demonstrate using selective IP (RO1138452) and EP2 (PF-04418948) antagonists that the anti-proliferative actions of treprostinil depend largely on EP2 receptors rather than IP receptors, unlike MRE-269 (selexipag-active metabolite). Likewise, EP2 receptor knockdown selectively reduced the functional responses to treprostinil but not MRE-269. Furthermore, EP2 receptor levels were enhanced in human PASMCs and in lung sections from PAH patients compared to controls. Thus, EP2 receptors represent a novel therapeutic target for treprostinil, highlighting key pharmacological differences between prostacyclin mimetics used in PAH.


Subject(s)
Cell Proliferation/drug effects , Epoprostenol/analogs & derivatives , Hypertension, Pulmonary/drug therapy , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Prostaglandin E, EP2 Subtype/biosynthesis , Up-Regulation/drug effects , Adolescent , Adult , Child , Epoprostenol/pharmacology , Female , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Second Messenger Systems/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL