Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 11(21)2022 10 29.
Article in English | MEDLINE | ID: mdl-36359815

ABSTRACT

Primary graft dysfunction (PGD) is characterized by alveolar epithelial and vascular endothelial damage and inflammation, lung edema and hypoxemia. Up to one-third of recipients develop the most severe form of PGD (Grade 3; PGD3). Animal studies suggest that neutrophils contribute to the inflammatory process through neutrophil extracellular traps (NETs) release (NETosis). NETs are composed of DNA filaments decorated with granular proteins contributing to vascular occlusion associated with PGD. The main objective was to correlate NETosis in PGD3 (n = 9) versus non-PGD3 (n = 27) recipients in an exploratory study. Clinical data and blood samples were collected from donors and recipients pre-, intra- and postoperatively (up to 72 h). Inflammatory inducers of NETs' release (IL-8, IL-6 and C-reactive protein [CRP]) and components (myeloperoxidase [MPO], MPO-DNA complexes and cell-free DNA [cfDNA]) were quantified by ELISA. When available, histology, immunohistochemistry and immunofluorescence techniques were performed on lung biopsies from donor grafts collected during the surgery to evaluate the presence of activated neutrophils and NETs. Lung biopsies from donor grafts collected during transplantation presented various degrees of vascular occlusion including neutrophils undergoing NETosis. Additionally, in recipients intra- and postoperatively, circulating inflammatory (IL-6, IL-8) and NETosis biomarkers (MPO-DNA, MPO, cfDNA) were up to 4-fold higher in PGD3 recipients compared to non-PGD3 (p = 0.041 to 0.001). In summary, perioperative elevation of NETosis biomarkers is associated with PGD3 following human lung transplantation and these biomarkers might serve to identify recipients at risk of PGD3 and initiate preventive therapies.


Subject(s)
Cell-Free Nucleic Acids , Extracellular Traps , Lung Transplantation , Primary Graft Dysfunction , Humans , Biomarkers/metabolism , DNA/metabolism , Extracellular Traps/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Lung Transplantation/adverse effects , Primary Graft Dysfunction/metabolism
2.
Ann Thorac Surg ; 110(6): 1861-1868, 2020 12.
Article in English | MEDLINE | ID: mdl-32652069

ABSTRACT

BACKGROUND: Ischemia-reperfusion associated with prolonged warm ischemia during donation after circulatory death (DCD) induces acute lung injury. The objective of this study was to combine ex vivo lung perfusion (EVLP) and a heat shock protein-90 inhibitor (HSP90i) to recondition DCD organs and prevent primary graft dysfunction. METHODS: Pigs (55 to 65 kg) were anesthetized, ventilated, and hemodynamically monitored. Cardiac arrest was induced with potassium chloride, and animals were left nonventilated for 2 hours. Lungs were procured and perfused in an EVLP platform for 4 hours by using a cellular perfusate. In the study group, the perfusate contained HSP90i and its transport vehicle (n = 4). In the control group, the perfusate contained only the transport vehicle (n = 4). Gas exchange, airway pressures, and compliance were measured. Pulmonary edema was assessed by bronchoscopy and weight measurement. Lung biopsy samples were obtained for histologic analyses and protein expression measurements. RESULTS: The use of HSP90i reduced lung weight gain to 8.4 ± 3.4% vs 26.6 ± 6.2% in the control group (P < .05). There was reduced edema formation. The ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen at the end of EVLP was 423 ± 65 in the study group vs 339 ± 25 mm Hg in the control group, but this difference was not statistically significant. Lactate metabolism, pulmonary vascular resistance, and pulmonary arterial pressure improved during EVLP with the use of the HSP90i. CONCLUSIONS: The use of HSP90i with EVLP improves the lung reconditioning process. Further research is required to confirm whether these findings translate to benefit once transplanted and observed in vivo. Successful pharmacologic inhibitors may expand the donor pool in the context of DCD donors.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heart Arrest , Lung Transplantation/adverse effects , Lung/drug effects , Organ Preservation/methods , Animals , Disease Models, Animal , Extracorporeal Circulation , Lung/pathology , Lung/physiopathology , Male , Perfusion , Swine , Warm Ischemia/adverse effects
3.
PLoS One ; 14(7): e0219205, 2019.
Article in English | MEDLINE | ID: mdl-31269062

ABSTRACT

In the rat, oxytocin (OT) produces dose-dependent diuretic and natriuretic responses. Post-translational enzymatic conversion of the OT biosynthetic precursor forms both mature and C-terminally extended peptides. The plasma concentrations of these C-terminally extended peptides (OT-G; OT-GK and OT-GKR) are elevated in newborns and pregnant rats. Intravenous injection of OT-GKR to rats inhibits diuresis, whereas injection of amidated OT stimulates diuresis. Since OT and OT-GKR show different effects on the urine flow, we investigated whether OT-GKR modulates renal action by inhibition of the arginine-vasopressin (AVP) receptor V2 (V2R), the receptor involved in renal water reabsorption. Experiments were carried out in the 8-week-old Wistar rats receiving intravenous (iv) injections of vehicle, OT, OT-GKR or OT+OT-GKR combination. OT (10 µmol/kg) increased urine outflow by 40% (P<0.01) and sodium excretion by 47% (P<0.01). Treatment with OT-GKR (10 µmol/kg) decreased diuresis by 50% (P<0.001), decreased sodium excretion by 50% (P<0.05) and lowered potassium by 42% (P<0.05). OT antagonist (OTA) reduced diuresis and natriuresis exerted by OT, whereas the anti-diuretic effect of OT-GKR was unaffected by OTA. The treatment with V2R antagonist (V2A) in the presence and absence of OT induced diuresis, sodium and potassium outflow. V2A in the presence of OT-GKR only partially increased diuresis and natriuresis. Autoradiography and molecular docking analysis showed potent binding of OT-GKR to V2R. Finally, the release of cAMP from CHO cells overexpressing V2 receptor was induced by low concentration of AVP (EC50:4.2e-011), at higher concentrations of OT (EC50:3.2e-010) and by the highest concentrations of OT-GKR (EC50:1.1e-006). OT-GKR potentiated cAMP release when combined with AVP, but blocked cAMP release when combined with OT. These results suggest that OT-GKR by competing for the OT renal receptor (OTR) and binding to V2R in the kidney, induces anti-diuretic, anti-natriuretic, and anti-kaliuretic effects.


Subject(s)
Diuresis , Natriuresis , Oxytocin/metabolism , Animals , Autoradiography , Binding, Competitive , CHO Cells , Cell Line , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Electrolytes/metabolism , Humans , Kidney/metabolism , Molecular Docking Simulation , Peptides/metabolism , Rats , Rats, Wistar , Receptors, Vasopressin/metabolism , Urination , Vasopressins/metabolism
4.
Mol Cell Endocrinol ; 412: 170-81, 2015 Sep 05.
Article in English | MEDLINE | ID: mdl-25963797

ABSTRACT

Oxytocin (OT) stimulates cardioprotection. Here we investigated heart-derived H9c2 cells in simulated ischemia-reperfusion (I-R) experiments in order to examine the mechanism of OT protection. I-R was induced in an anoxic chamber for 2 hours and followed by 2 h of reperfusion. In comparison to normoxia, I-R resulted in decrease of formazan production by H9c2 cells to 63.5 ± 1.7% (MTT assay) and in enhanced apoptosis from 1.7 ± 0.3% to 2.8 ± 0.4% (Tunel test). Using these assays it was observed that treatment with OT (1-500 nM) exerted significant protection during I-R, especially when OT was added at the time of ischemia or reperfusion. Using the CM-H2DCFDA probe we found that OT triggers a short-lived burst in reactive oxygen species (ROS) production in cells but reduces ROS production evoked by I-R. In cells treated with OT, Western-blot revealed the phosphorylation of Akt (Thr 308, p-Akt), eNOS and ERK 1/2. Microscopy showed translocation of p-Akt and eNOS into the nuclear and perinuclear area and NO production in cells treated with OT. The OT-induced protection against I-R was abrogated by an OT antagonist, the Pi3K inhibitor Wortmannin, the cGMP-dependent protein kinase (PKG) inhibitor, KT5823, as well as soluble guanylate cyclase (GC) inhibitor, ODQ, and particulate GC antagonist, A71915. In conditions of I-R, the cells with siRNA-mediated reduction in OT receptor (OTR) expression responded to OT treatment by enhanced apoptosis. In conclusion, the OTR protected H9c2 cells against I-R, especially if activated at the onset of ischemia or reperfusion. The OTR-transduced signals include pro-survival kinases, such as Akt and PKG.


Subject(s)
Cardiotonic Agents/pharmacology , Myocytes, Cardiac/physiology , Oxytocin/pharmacology , Cell Hypoxia , Cell Line , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/metabolism , Drug Evaluation, Preclinical , Myoblasts/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Oxytocin/metabolism
5.
Endocrinology ; 156(4): 1416-28, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25562615

ABSTRACT

Oxytocin (OT) is involved in the regulation of energy metabolism and in the activation of cardioprotective mechanisms. We evaluated whether chronic treatment with OT could prevent the metabolic and cardiac abnormalities associated with diabetes and obesity using the db/db mice model. Four-week-old male db/db mice and their lean nondiabetic littermates (db/+) serving as controls were treated with OT (125 ng/kg · h) or saline vehicle for a period of 12 weeks. Compared with db/+ mice, the saline-treated db/db mice developed obesity, hyperglycemia, and hyperinsulinemia. These mice also exhibited a deficient cardiac OT/natriuretic system and developed systolic and diastolic dysfunction resulting from cardiomyocyte hypertrophy, fibrosis, and apoptosis. These abnormalities were associated with increased reactive oxygen species (ROS) production, inflammation, and suppressed 5'-adenosine monophosphate kinase signaling pathway. The db/db mice displayed reduced serum levels of adiponectin and adipsin and elevated resistin. OT treatment increased circulating OT levels, significantly reduced serum resistin, body fat accumulation (19%; P<.001), fasting blood glucose levels by (23%; P<.001), and improved glucose tolerance and insulin sensitivity. OT also normalized cardiac OT receptors, atrial natriuretic peptide, and brain natriuretic peptide, expressions and prevented systolic and diastolic dysfunction as well as cardiomyocyte hypertrophy, fibrosis, and apoptosis. Furthermore, OT reduced cardiac oxidative stress and inflammation and normalized the 5'-adenosine monophosphate-activated protein kinase signaling pathway. The complete normalization of cardiac structure and function by OT treatment in db/db mice contrasted with only partial improvement of hyperglycemia and hyperinsulinemia. These results indicate that chronic treatment with OT partially improves glucose and fat metabolism and reverses abnormal cardiac structural remodeling, preventing cardiac dysfunction in db/db mice.


Subject(s)
Cardiomyopathies/prevention & control , Diabetes Mellitus, Type 2/complications , Hyperinsulinism/complications , Obesity/complications , Oxytocin/therapeutic use , Adiponectin/blood , Animals , Blood Glucose/metabolism , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/drug effects , Hyperinsulinism/metabolism , Insulin Resistance/physiology , Male , Mice , Obesity/metabolism , Oxytocin/pharmacology , Resistin/blood
6.
Int J Cardiol ; 175(1): 38-49, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24852833

ABSTRACT

BACKGROUND: Oxytocin (OT) and functional OT receptor (OTR) are expressed in the heart and are involved in blood pressure regulation and cardioprotection. Cardiac OTR signaling is associated with atrial natriuretic peptide (ANP) and nitric oxide (NO) release. During the synthesis of OT, its precursor, termed OT-Gly-Lys-Arg (OT-GKR), is accumulated in the developing rat heart. Consequently, we hypothesized that an OT-related mechanism of ANP controls cardiomyocyte (CM) hypertrophy. METHODS: The experiments were carried out in newborn and adult rat CM cultures. The enhanced protein synthesis and increased CM volume were mediated by a 24-h treatment with endothelin-1 or angiotensin II. RESULTS: The treatment of CM with OT or its abundant cardiac precursor, OT-GKR, revealed ANP accumulation in the cell peri-nuclear region and increased intracellular cGMP. Consequently, the CM hypertrophy was abolished by the treatment of 10nM OT or 10nM OT-GKR. The ANP receptor antagonist (anantin) and NO synthases inhibitor (l-NAME) inhibited cGMP production in CMs exposed to OT. STO-609 and compound C inhibition of anti-hypertrophic OT effects in CMs indicated the contribution of calcium-calmodulin kinase kinase and AMP-activated protein kinase pathways. Moreover, in ET-1 stimulated cells, OT treatment normalized the reduced Akt phosphorylation, prevented abundant accumulation of ANP and blocked ET-1-mediated translocation of nuclear factor of activated T-cells (NFAT) into the cell nuclei. CONCLUSION: cGMP/protein kinase G mediates OT-induced anti-hypertrophic response with the contribution of ANP and NO. OT treatment represents a novel approach in attenuation of cardiac hypertrophy during development and cardiac pathology.


Subject(s)
Cell Enlargement/drug effects , Myocytes, Cardiac/drug effects , Oxytocin/pharmacology , Animals , Animals, Newborn , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxytocin/therapeutic use , Rats , Rats, Sprague-Dawley , Treatment Outcome
7.
Diabetologia ; 57(6): 1257-67, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24595856

ABSTRACT

AIMS/HYPOTHESIS: Obesity and diabetes increase the risk of developing cardiovascular diseases and heart failure. These metabolic disorders are generally reflected by natriuretic peptide system deficiency. Since brain natriuretic peptide (BNP) is known to influence metabolism and cardioprotection, we investigated the effect of chronic exogenous BNP treatment on adverse myocardial consequences related to obesity and diabetes. METHODS: Ten-week-old C57BL/KsJ-db/db obese diabetic mice (db/db) and their lean control littermates (db/+) were treated with BNP (0.6 µg kg(-1) h(-1)) or saline for 12 weeks (n = 10/group). Serial blood and tomography analysis were performed. Cardiac function was determined by echocardiography, and biochemical and histological heart and fat analyses were also performed. RESULTS: BNP treatment resulted in an average increase in plasma BNP levels of 70 pg/ml. An improvement in the metabolic profile of db/db mice was observed, including a reduction in fat content, increased insulin sensitivity, improved glucose tolerance and lower blood glucose, despite increased food intake. db/db mice receiving saline displayed both early systolic and diastolic dysfunction, whereas these functional changes were prevented by BNP treatment. The cardioprotective effects of BNP were attributed to the inhibition of cardiomyocyte apoptosis, myocardial fibrosis, cardiac hypertrophy and the AGE-receptor for AGE (RAGE) system as well as normalisation of cardiac AMP-activated protein kinase and endothelial nitric oxide synthase activities. CONCLUSIONS/INTERPRETATION: Our results indicate that chronic BNP treatment at low dose improves the metabolic profile and prevents the development of myocardial dysfunction in db/db mice.


Subject(s)
Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Natriuretic Peptide, Brain/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese
8.
Endocrinology ; 153(11): 5361-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23024264

ABSTRACT

Principal limitation of cell therapy is cell loss after transplantation because of the interplay between ischemia, inflammation, and apoptosis. We investigated the mechanism of preconditioning of mesenchymal stem cells (MSCs) with oxytocin (OT), which has been proposed as a novel strategy for enhancing therapeutic potential of these cells in ischemic heart. In this study, we demonstrate that rat MSCs express binding sites for OT receptor and OT receptor transcript and protein as detected by RT-PCR and immunofluorescence, respectively. In response to OT (10(-10) to 10(-6) M) treatment, MSCs respond with rapid calcium mobilization and up-regulation of the protective protein kinase B (PKB or Akt) and phospho-ERK1/2 proteins. In OT-stimulated cells, phospho-Akt accumulates intracellularly close to the mitochondrial marker cytochrome c oxidase subunit 4. Functional analyses reveal the involvement of Akt/ERK1/2 pathways in cell proliferation, migration, and protection against the cytotoxic and apoptotic effects of hypoxia and serum deprivation. In addition, OT preconditioning increases MSC glucose uptake. Genes with angiogenic, antiapoptotic, and cardiac antiremodeling properties, such as heat shock proteins (hsps) HSP27, HSP32, HSP70, vascular endothelial growth factor, thrombospondin, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, TIMP-3, and matrix metalloproteinase-2, were also up-regulated upon OT exposure. Moreover, coculture with OT-preconditioned MSC reduces apoptosis, as measured using terminal transferase dUTP nick end labeling assay in newborn rat cardiomyocytes exposed to hypoxia and reoxygenation. In conclusion, these results indicate that OT treatment evokes MSC protection through both intrinsic pathways and secretion of cytoprotective factors. Ex vivo cellular treatment with OT represents an attractive strategy aimed to maximize the biological and functional properties of effector cells.


Subject(s)
Cell Survival/drug effects , Mesenchymal Stem Cells/drug effects , Oxytocin/pharmacology , Receptors, Oxytocin/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Calcium/metabolism , Cell Hypoxia/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Electron Transport Complex IV/metabolism , Mesenchymal Stem Cells/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptors, Oxytocin/genetics , Up-Regulation/drug effects
9.
Hypertension ; 53(5): 775-81, 2009 May.
Article in English | MEDLINE | ID: mdl-19273740

ABSTRACT

The renin-angiotensin and sympathetic nervous systems play critical interlinked roles in the development of left ventricular hypertrophy, fibrosis, and dysfunction. These studies investigated the hemodynamic and cardiac effects of monoblockade and coblockade of renin-angiotensin and sympathetic nervous systems. Stroke-prone spontaneously hypertensive rats (16 weeks old; male; n=12 per group) received the sympatholytic imidazoline compound, moxonidine (2.4 mg/kg per day); the angiotensin-receptor blocker eprosartan (30 mg/kg per day), separately or in combination; or saline vehicle for 8 weeks, SC, via osmotic minipumps. Blood pressure and heart rate were continuously measured by radiotelemetry. After 8 weeks, in vivo cardiac function and structure were measured by transthoracic echocardiography and a Millar conductance catheter, and the rats were then euthanized and blood and heart ventricles collected for various determinations. Compared with vehicle, the subhypotensive dose of moxonidine resulted in lower (P<0.01) heart rate, left ventricular hypertrophy, cardiomyocyte cross-sectional area, interleukin 1 beta, tumor necrosis factor-alpha, and mRNA for natriuretic peptides. Eprosartan reduced pressure (P<0.01), as well as extracellular signal-regulated kinase (ERK) 44 phosphorylation, Bax/Bcl-2, and collagen I/III, and improved left ventricular diastolic function (P<0.03). Combined treatment resulted in greater reductions in blood pressure, heart rate, left ventricular hypertrophy, collagen I/III, and inhibited inducible NO synthase and increased endothelial NO synthase phosphorylation, as well as reduced left ventricular anterior wall thickness, without altering the other parameters. Thus, in advanced hypertension complicated with cardiac fibrosis, sympathetic inhibition and angiotensin II blockade resulted in greater reduction in blood pressure and heart rate, inhibition of inflammation, and improved left ventricular pathology but did not add to the benefits of angiotensin II blockade on cardiac function.


Subject(s)
Acrylates/pharmacology , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Heart Rate/drug effects , Hypertension/drug therapy , Imidazoles/pharmacology , Thiophenes/pharmacology , Ventricular Function, Left/drug effects , Acrylates/administration & dosage , Animals , Atrial Natriuretic Factor/blood , Hypertension/complications , Hypertension/physiopathology , Imidazoles/administration & dosage , Male , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Rats , Rats, Inbred SHR , Stroke/etiology , Thiophenes/administration & dosage
10.
Eur J Pharmacol ; 541(1-2): 73-9, 2006 Jul 10.
Article in English | MEDLINE | ID: mdl-16774751

ABSTRACT

Imidazoline I1-receptors are present in the heart and may be involved in atrial natriuretic peptide (ANP) release. The following studies investigated whether moxonidine (an antihypertensive imidazoline I1-receptor and alpha2-adrenoceptor agonist) acts directly on the heart to stimulate ANP release, and to characterize the receptor type involved in this action. Perfusion of rat (200-225 g) isolated hearts with moxonidine (10(-6) and 10(-5) M), for 30 min, resulted in ANP release (83+/-29 and 277+/-70 ng/30 min, above basal, respectively), significantly (P<0.01) different from perfusion with buffer (-6+/-31 ng/30 min). ANP release stimulated by moxonidine (10(-6) M) was inhibited by co-perfusion with the antagonists, AGN192403 (imidazoline I1-receptor), phenoxybenzamine (alpha2>alpha1-adrenoceptors), and prazosin (alpha1>alpha2-adrenoceptors), but increased by rauwolscine (alpha2-adrenoceptors). Perfusion with 10(-5) M brimonidine (full alpha2-adrenoceptor agonist) inhibited moxonidine-stimulated ANP release. Similarly, moxonidine (10(-6) M) tended to reduce coronary flow, but significantly increased coronary flow in the presence of brimonidine, which was vasoconstrictive when perfused alone. Coronary flow was reduced by 10(-5) M each, brimonidine>clonidine>moxonidine; while similar bradycardia was observed with clonidine and moxonidine, but not with brimonidine. In conclusion, these results argue in favor of moxonidine acting primarily on imidazoline I1-receptors to release ANP, with both alpha2-adrenoceptor and imidazoline I1-receptors exerting inhibitory inter-relation. In contrast, the coronary vasodilatory effect of moxonidine requires full activation of alpha2-adrenoceptor. The sympatholytic and ANP-releasing effects of moxonidine appear to be mediated by cardiac imidazoline receptors that may be differentially localized. Most importantly, moxonidine can stimulate ANP release from the heart without contribution of the central nervous system.


Subject(s)
Atrial Natriuretic Factor/metabolism , Heart/drug effects , Imidazoles/pharmacology , Myocardium/metabolism , Receptors, Drug/physiology , Adrenergic alpha-2 Receptor Agonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Antihypertensive Agents/pharmacology , Bridged Bicyclo Compounds/pharmacology , Brimonidine Tartrate , Clonidine/pharmacology , Coronary Circulation/drug effects , Drug Synergism , Female , Heart Rate/drug effects , Heptanes/pharmacology , Imidazoline Receptors , In Vitro Techniques , Perfusion/methods , Phenoxybenzamine/pharmacology , Prazosin/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/physiology , Receptors, Drug/agonists , Receptors, Drug/antagonists & inhibitors , Yohimbine/pharmacology
11.
Br J Pharmacol ; 145(1): 50-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15700025

ABSTRACT

We have previously shown that acute intravenous injections of moxonidine and clonidine increase plasma atrial natriuretic peptide (ANP), a vasodilator, diuretic and natriuretic hormone. We hypothesized that moxonidine stimulates the release of ANP, which would act on its renal receptors to cause diuresis and natriuresis, and these effects may be altered in hypertension. Moxonidine (0, 10, 50, 100 or 150 microg in 300 microl saline) and clonidine (0, 1, 5 or 10 microg in 300 microl saline) injected intravenously in conscious normally hydrated normotensive Sprague-Dawley rats (SD, approximately 200 g) and 12-14-week-old Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHR) dose-dependently stimulated diuresis, natriuresis, kaliuresis and cGMP excretion, with these effects being more pronounced during the first hour post-injection. The actions of 5 microg clonidine and 50 microg moxonidine were inhibited by yohimbine, an alpha2-adrenoceptor antagonist, and efaroxan, an imidazoline I1-receptor antagonist. Moxonidine (100 microg) stimulated (P<0.01) diuresis in SHR (0.21+/-0.04 vs 1.16+/-0.06 ml h(-1) 100 g(-1)), SD (0.42+/-0.06 vs 1.56+/-0.19 ml h(-1) 100 g(-1)) and WKY (0.12+/-0.04 vs 1.44+/-0.21 ml h(-1) 100 g(-1)). Moxonidine-stimulated urine output was lower in SHR than in SD and WKY. Moxonidine-stimulated sodium and potassium excretions were lower in SHR than in SD, but not WKY, demonstrating an influence of strain but not of pressure. Pretreatment with the natriuretic peptide antagonist anantin (5 or 10 microg) resulted in dose-dependent inhibition of moxonidine-stimulated urinary actions. Anantin (10 microg) inhibited (P<0.01) urine output to 0.38+/-0.06, 0.12+/-0.01, and 0.16+/-0.04 ml h(-1) 100 g(-1) in SD, WKY, and SHR, respectively. Moxonidine increased (P<0.01) plasma ANP in SD (417+/-58 vs 1021+/-112 pg ml(-1)) and WKY (309+/-59 vs 1433+/-187 pg ml(-1)), and in SHR (853+/-96 vs 1879+/-229 pg ml(-1)). These results demonstrate that natriuretic peptides mediate the urinary actions of moxonidine through natriuretic peptide receptors.


Subject(s)
Antihypertensive Agents/pharmacology , Clonidine/pharmacology , Diuresis/drug effects , Imidazoles/pharmacology , Natriuretic Peptides/physiology , Animals , Antihypertensive Agents/antagonists & inhibitors , Benzofurans/pharmacology , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Female , Imidazoles/antagonists & inhibitors , Imidazoline Receptors , Natriuresis/drug effects , Peptides, Cyclic/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Drug/drug effects , Yohimbine/pharmacology
12.
J Pharmacol Exp Ther ; 310(2): 446-51, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15075383

ABSTRACT

We have recently identified imidazoline I(1)-receptors in the heart. In the present study, we tested regulation of cardiac I(1)-receptors versus alpha(2) -adrenoceptors in response to hypertension and to chronic exposure to agonist. Spontaneously hypertensive rats (SHR, 12-14 weeks old) received moxonidine (10, 60, and 120 microg/kg/h s.c.) for 1 and 4 weeks. Autoradiographic binding of (125)I-paraiodoclonidine (0.5 nM, 1 h, 22 degrees C) and inhibition of binding with epinephrine (10(-10)-10(-5) M) demonstrated the presence of alpha(2)-adrenoceptors in heart atria and ventricles. Immunoblotting and reverse transcription-polymerase chain reaction identified alpha(2A)-alpha(2B)-, and alpha(2C), and -adrenoceptor proteins and mRNA, respectively. However, compared with normotensive controls, cardiac alpha(2) -adrenoceptor kinetic parameters, receptor proteins, and mRNAs were not altered in SHR with or without moxonidine treatment. In contrast, autoradiography showed that up-regulated atrial I(1)-receptors in SHR are dose-dependently normalized by 1 week, with no additional effect after 4 weeks of treatment. Moxonidine (120 microg/kg/h) decreased B(max) in right (40.0 +/- 2.9-7.0 +/- 0.6 fmol/unit area; p < 0.01) and left (27.7 +/- 2.8-7.1 +/- 0.4 fmol/unit area; p < 0.01) atria, and decreased the 85- and 29-kDa imidazoline receptor protein bands, in right atria, to 51.8 +/- 3.0% (p < 0.01) and 82.7 +/- 5.2% (p < 0.03) of vehicle-treated SHR, respectively. Moxonidine-associated percentage of decrease in B(max) only correlated with the 85-kDa protein (R(2) = 0.57; p < 0.006), suggesting that this protein may represent I(2)-receptors. The weak but significant correlation between the two imidazoline receptor proteins (R(2) = 0.28; p < 0.03) implies that they arise from the same gene. In conclusion, the heart possesses I(1)-receptors and alpha(2)-adrenoceptors, but only I(1)-receptors are responsive to hypertension and to chronic in vivo treatment with a selective I(1)-receptor agonist.


Subject(s)
Hypertension/metabolism , Imidazoles/administration & dosage , Myocardium/metabolism , Receptors, Adrenergic, alpha-2/biosynthesis , Receptors, Drug/biosynthesis , Animals , Dose-Response Relationship, Drug , Female , Heart Atria/drug effects , Heart Atria/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Imidazoline Receptors , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-2/genetics , Receptors, Drug/genetics , Up-Regulation/drug effects , Up-Regulation/physiology
13.
Am J Hypertens ; 15(9): 803-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12219876

ABSTRACT

BACKGROUND: Acute intravenous administration of moxonidine, an imidazoline I1-receptor agonist, reduces blood pressure (BP) in normotensive and hypertensive rats, induces diuresis and natriuresis, and stimulates plasma atrial natriuretic peptide (ANP). In these studies we investigated the involvement of natriuretic peptides (ANP and brain natriuretic peptide) in the effects of chronic activation of imidazoline receptors. METHODS: Spontaneously hypertensive rats (SHR; 12 to 14 weeks old) received 7-day moxonidine treatment at various doses (10, 20, 60, and 120 microg/kg/h) via subcutaneously implanted osmotic minipumps. RESULTS: Hemodynamic parameters (continuously monitored by telemetry) revealed that, compared with saline-treated rats, moxonidine dose-dependently decreased blood pressures (BPs). Maximal blood pressure lowering effect was achieved by day 4 of treatment, at which point 60 microg/kg/h reduced mean arterial pressure (MAP) by 14.5 +/- 6.8 mm Hg as compared with basal levels. The decrease in MAP was influenced by a drop in both diastolic and systolic pressures. Moxonidine treatment did not alter daily urinary sodium and potassium excretions, but 120 microg/kg/h moxonidine decreased urine volume after 2 days and increased cyclic guanosine 3'5'monophosphate excretion on days 4 to 7 of treatment. Chronic moxonidine treatment dose-dependently increased plasma ANP to reach, at 120 microg/kg/h, a 40% increase (P < .01) above that of corresponding saline-treated SHR, with a concomitant increase in left and right atrial ANP mRNA (more than twofold). Plasma BNP increased by 120 microg/kg/h moxonidine (11.0 +/- 1.1 v 16.5 +/- 1.9 pg/mL, P < .002) without significant increases in atrial and ventricular BNP mRNA. CONCLUSIONS: ANP and BNP may be involved in the antihypertensive effect of chronic moxonidine treatment. Accordingly, natriuretic peptides may contribute to the sympatholytic and cardioprotective effects of chronic activation of imidazoline I1-receptors.


Subject(s)
Antihypertensive Agents/pharmacology , Atrial Natriuretic Factor/drug effects , Blood Pressure/drug effects , Imidazoles/pharmacology , Natriuretic Peptide, Brain/drug effects , Animals , Atrial Natriuretic Factor/genetics , Body Weight , Female , Gene Expression , Imidazoline Receptors , Natriuretic Peptide, Brain/genetics , Rats , Rats, Inbred SHR , Receptors, Drug/drug effects , Receptors, Drug/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...