Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Pharmacol Toxicol Methods ; 124: 107480, 2023.
Article in English | MEDLINE | ID: mdl-37979811

ABSTRACT

Despite medical advances in the treatment of heart failure (HF), mortality remains high. It has been shown that alterations of the autonomic-nervous-system (ANS) are associated with HF progression and increased mortality. Preclinical models are required to evaluate the effectiveness of novel treatments modulating the autonomic imbalance. However, there are neither standard models nor diagnostic methods established to measure sympathetic and parasympathetic outflow continuously. Digital technologies might be a reliable tool for continuous assessment of autonomic function within experimental HF models. Telemetry devices and pacemakers were implanted in beagle dogs (n = 6). HF was induced by ventricular pacing. Cardiac hemodynamics, plasma catecholamines and parameter describing the ANS ((heart rate variability (HRV), deceleration capacity (DC), and baroreflex sensitivity (BRS)) were continuously measured at baseline, during HF conditions and during recovery phase. The pacing regime led to the expected depression in cardiac hemodynamics. Telemetric assessment of the ANS function showed a significant decrease in Total power, DC, and Heart rate recovery, whereas BRS was not significantly affected. In contrast, plasma catecholamines, revealing sympathetic activity, showed only a significant increase in the recovery phase. A precise diagnostic of the ANS in the context of HF is becoming increasingly important in experimental models. Up to now, these models have shown many limitations. Here we present the continuous assessment of the autonomic function in the progression of HF. We could demonstrate the advantage of highly resolved ANS measurement by HR and BP derived parameters due to early detection of an autonomic imbalance in the progression of HF.


Subject(s)
Autonomic Nervous System , Heart Failure , Animals , Dogs , Autonomic Nervous System/physiology , Hemodynamics/physiology , Heart Rate/physiology , Catecholamines
2.
Br J Pharmacol ; 180(19): 2500-2513, 2023 10.
Article in English | MEDLINE | ID: mdl-37170767

ABSTRACT

BACKGROUND AND PURPOSE: First-generation soluble guanylate cyclase (sGC) stimulators have shown clinical benefit in pulmonary hypertension (riociguat) and chronic heart failure (vericiguat). However, given the broad therapeutic opportunities for sGC stimulators, tailored molecules for distinct indications are required. EXPERIMENTAL APPROACH: We report the high-throughput screening (HTS)-based discovery of a second generation of sGC stimulators from a novel imidazo[1,2-a]pyridine lead series. An intense medicinal chemistry programme resulted in the discovery of the sGC stimulator BAY 1165747 (BAY-747). The pharmacokinetic profile of BAY-747 was determined in different species, and it was broadly characterized in pharmacological model systems relevant for vasodilatation and hypertension. KEY RESULTS: BAY-747 is a highly potent sGC stimulator in vitro. In addition, BAY-747 showed an excellent pharmacokinetic profile with long half-life and low peak-to-trough ratio. BAY-747 was investigated in experimental in vivo models of malignant and resistant hypertension (rHT). In spontaneously hypertensive (SH) rats, BAY-747 caused a dose-related and long-lasting decrease in mean arterial blood pressure (MAP). Oral treatment over 12 days resulted in a persistent decrease. BAY-747 provided additional benefit when dosed on top of losartan, amlodipine or spironolactone and even on top of triple combinations of frequently used antihypertensive drugs. In a new canine model of rHT, BAY-747 caused a dose-related and long-lasting (>6 h) MAP decrease. CONCLUSION AND IMPLICATIONS: BAY-747 is a potent, orally available sGC stimulator. BAY-747 shows long-acting pharmacodynamic effects with a very low peak-to-trough ratio. BAY-747 could be a treatment alternative for patients with hypertension, especially those not responding to standard-of-care therapy.


Subject(s)
Heart Failure , Hypertension, Pulmonary , Hypertension , Rats , Animals , Dogs , Soluble Guanylyl Cyclase , Hypertension/drug therapy , Hypertension, Pulmonary/drug therapy , Heart Failure/drug therapy , Vasodilator Agents/therapeutic use
3.
J Am Heart Assoc ; 12(4): e028056, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36752268

ABSTRACT

Background Deep vein thrombosis (DVT) is the primary cause of pulmonary embolism and the third most life-threatening cardiovascular disease in North America. Post-DVT anticoagulants, such as warfarin, heparin, and direct oral anticoagulants, reduce the incidence of subsequent venous thrombi. However, all currently used anticoagulants affect bleeding time at various degrees, and there is therefore a need for improved therapeutic regimens in DVT. It has recently been shown that mast cells play a crucial role in a DVT murine model. The underlying mechanism involved in the prothrombotic properties of mast cells, however, has yet to be identified. Methods and Results C57BL/6 mice and mouse mast cell protease-4 (mMCP-4) genetically depleted mice (mMCP-4 knockout) were used in 2 mouse models of DVT, partial ligation (stenosis) and ferric chloride-endothelial injury model of the inferior vena cava. Thrombus formation and impact of genetically repressed or pharmacologically (specific inhibitor TY-51469) inhibited mMCP-4 were evaluated by morphometric measurements of thrombi immunochemistry (mouse and human DVT), color Doppler ultrasound, bleeding times, and enzymatic activity assays ex vivo. Recombinant chymases, mMCP-4 (mouse) and CMA-1 (human), were used to characterize the interaction with murine and human plasmin, respectively, by mass spectrometry and enzymatic activity assays. Inhibiting mast cell-generated mMCP-4, genetically or pharmacologically, resolves and prevents venous thrombus formation in both DVT models. Inferior vena cava blood flow obstruction was observed in the stenosis model after 6 hours of ligation, in control- but not in TY-51469-treated mice. In addition, chymase inhibition had no impact on bleeding times of healthy or DVT mice. Furthermore, endogenous chymase limits plasmin activity in thrombi ex vivo. Recombinant mouse or human chymase degrades/inactivates purified plasmin in vitro. Finally, mast cell-containing immunoreactive chymase was identified in human DVT. Conclusions This study identified a major role for mMCP-4, a granule-localized protease of chymase type, in DVT formation. These findings support a novel pharmacological strategy to resolve or prevent DVT without affecting the coagulation cascade through the inhibition of chymase activity.


Subject(s)
Fibrinolysin , Venous Thrombosis , Mice , Humans , Animals , Chymases/metabolism , Bleeding Time , Disease Models, Animal , Constriction, Pathologic , Mice, Inbred C57BL , Venous Thrombosis/prevention & control , Anticoagulants
4.
Respir Res ; 23(1): 272, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36183104

ABSTRACT

BACKGROUND: Oxidative stress associated with severe cardiopulmonary diseases leads to impairment in the nitric oxide/soluble guanylate cyclase signaling pathway, shifting native soluble guanylate cyclase toward heme-free apo-soluble guanylate cyclase. Here we describe a new inhaled soluble guanylate cyclase activator to target apo-soluble guanylate cyclase and outline its therapeutic potential. METHODS: We aimed to generate a novel soluble guanylate cyclase activator, specifically designed for local inhaled application in the lung. We report the discovery and in vitro and in vivo characterization of the soluble guanylate cyclase activator mosliciguat (BAY 1237592). RESULTS: Mosliciguat specifically activates apo-soluble guanylate cyclase leading to improved cardiopulmonary circulation. Lung-selective effects, e.g., reduced pulmonary artery pressure without reduced systemic artery pressure, were seen after inhaled but not after intravenous administration in a thromboxane-induced pulmonary hypertension minipig model. These effects were observed over a broad dose range with a long duration of action and were further enhanced under experimental oxidative stress conditions. In a unilateral broncho-occlusion minipig model, inhaled mosliciguat decreased pulmonary arterial pressure without ventilation/perfusion mismatch. With respect to airway resistance, mosliciguat showed additional beneficial bronchodilatory effects in an acetylcholine-induced rat model. CONCLUSION: Inhaled mosliciguat may overcome treatment limitations in patients with pulmonary hypertension by improving pulmonary circulation and airway resistance without systemic exposure or ventilation/perfusion mismatch. Mosliciguat has the potential to become a new therapeutic paradigm, exhibiting a unique mode of action and route of application, and is currently under clinical development in phase Ib for pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary , Acetylcholine , Animals , Guanylate Cyclase/metabolism , Guanylate Cyclase/therapeutic use , Nitric Oxide/metabolism , Rats , Soluble Guanylyl Cyclase/metabolism , Soluble Guanylyl Cyclase/therapeutic use , Swine , Swine, Miniature/metabolism , Thromboxanes/therapeutic use , Vasodilator Agents
5.
J Med Chem ; 64(9): 5323-5344, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33872507

ABSTRACT

Herein we describe the discovery, mode of action, and preclinical characterization of the soluble guanylate cyclase (sGC) activator runcaciguat. The sGC enzyme, via the formation of cyclic guanosine monophoshphate, is a key regulator of body and tissue homeostasis. sGC activators with their unique mode of action are activating the oxidized and heme-free and therefore NO-unresponsive form of sGC, which is formed under oxidative stress. The first generation of sGC activators like cinaciguat or ataciguat exhibited limitations and were discontinued. We overcame limitations of first-generation sGC activators and identified a new chemical class via high-throughput screening. The investigation of the structure-activity relationship allowed to improve potency and multiple solubility, permeability, metabolism, and drug-drug interactions parameters. This program resulted in the discovery of the oral sGC activator runcaciguat (compound 45, BAY 1101042). Runcaciguat is currently investigated in clinical phase 2 studies for the treatment of patients with chronic kidney disease and nonproliferative diabetic retinopathy.


Subject(s)
Drug Design , Enzyme Activators/chemistry , Soluble Guanylyl Cyclase/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , Dogs , Enzyme Activators/metabolism , Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Half-Life , Heart Rate/drug effects , Hemodynamics/drug effects , Hypertension/drug therapy , Hypertension/pathology , Molecular Dynamics Simulation , Rats , Rats, Inbred SHR , Solubility , Soluble Guanylyl Cyclase/metabolism , Structure-Activity Relationship
6.
J Cardiovasc Pharmacol ; 74(1): 44-52, 2019 07.
Article in English | MEDLINE | ID: mdl-31274842

ABSTRACT

Increased plasma vasopressin levels have been shown to be associated with the progression of congestive heart failure. Vasopressin mediates water retention by renal tubular V2 receptor activation as well as vasoconstriction, cardiac hypertrophy, and fibrosis through V1a receptor activation. Therefore, we developed a novel, dual-acting vasopressin receptor antagonist, BAY 1753011, with almost identical Ki-values of 0.5 nM at the human V1a receptor and 0.6 nM at the human V2 receptor as determined in radioactive binding assays. Renal V2 antagonism by BAY 1753011 was compared with the loop diuretic furosemide in acute diuresis experiments in conscious rats. Similar diuretic efficacy was found with 300-mg/kg furosemide (maximal diuretic response) and 0.1-mg/kg BAY 1753011. Furosemide dose-dependently induced plasma renin and angiotensin I levels, while an equiefficient diuretic BAY 1753011 dose did not activate the renin-angiotensin system. BAY 1753011 dose-dependently decreased the vasopressin-induced expression of the profibrotic/hypertrophic marker plasminogen activator inhibitor-1 and osteopontin in rat cardiomyocytes, while the selective V2 antagonist satavaptan was without any effect. The combined vascular V1a-mediated and renal V2-mediated properties as well as the antihypertrophic/antifibrotic activity enable BAY 1753011 to become a viable treatment option for oral chronic treatment of congestive heart failure.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/pharmacology , Heart Failure/drug therapy , Receptors, Vasopressin/drug effects , Animals , Aorta/drug effects , Aorta/metabolism , Arterial Pressure/drug effects , CHO Cells , Cricetulus , Diuresis/drug effects , Fibrosis , Furosemide/pharmacology , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Osteopontin/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Rats , Rats, Wistar , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/pharmacology
7.
J Med Chem ; 60(12): 5146-5161, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28557445

ABSTRACT

The first-in-class soluble guanylate cyclase (sGC) stimulator riociguat was recently introduced as a novel treatment option for pulmonary hypertension. Despite its outstanding pharmacological profile, application of riociguat in other cardiovascular indications is limited by its short half-life, necessitating a three times daily dosing regimen. In our efforts to further optimize the compound class, we have uncovered interesting structure-activity relationships and were able to decrease oxidative metabolism significantly. These studies resulting in the discovery of once daily sGC stimulator vericiguat (compound 24, BAY 1021189), currently in phase 3 trials for chronic heart failure, are now reported.


Subject(s)
Heart Failure/drug therapy , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Soluble Guanylyl Cyclase/metabolism , Structure-Activity Relationship , Administration, Intravenous , Administration, Oral , Animals , Blood Pressure/drug effects , Chemistry Techniques, Synthetic , Dogs , Hepatocytes/drug effects , Heterocyclic Compounds, 2-Ring/administration & dosage , Humans , Male , NG-Nitroarginine Methyl Ester/adverse effects , Pyrimidines/administration & dosage , Rats, Transgenic , Rats, Wistar , Soluble Guanylyl Cyclase/genetics
8.
BJU Int ; 119(2): 325-332, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27763717

ABSTRACT

OBJECTIVES: To evaluate the expression of the Rho/Rho-associated protein kinase (ROCK) pathway in the corpus cavernosum of patients with severe erectile dysfunction (ED) compared with healthy human corpus cavernosum, and to test the functional effects of two Rho kinase inhibitors (RKIs) on erectile tissue of patients with severe ED, which did not respond to phosphodiesterase type 5 inhibitors (PDE5Is). PATIENTS AND METHODS: Human corpus cavernosum samples were obtained after consent from men undergoing penile prosthesis implantation (n = 7 for organ bath experiments, n = 17 for quantitative PCR [qPCR]). Potent control subjects (n = 5) underwent penile needle biopsy. qPCR was performed for the expression of RhoA and ROCK subtypes 1 and 2. Immunohistochemistry staining against ROCK and α smooth muscle actin (αSMA) was performed on the corpus cavernosum of patients with ED. Tissue strips were precontracted with phenylephrine and incubated with 1 µm of the PDE5I vardenafil or with DMSO (control). Subsequently, increasing concentrations of the RKIs azaindole or Y-27632 were added, and relaxation of tissue was quantified. RESULTS: The expression of ROCK1 was unchanged (P > 0.05), while ROCK2 (P < 0.05) was significantly upregulated in patients with ED compared with controls. ROCK1 and ROCK2 protein colocalized with αSMA, confirming the presence of this kinase in cavernous smooth muscle cells and/or myofibroblasts. After incubation with DMSO, 10 µm azaindole and 10 µm Y-27632 relaxed precontracted tissues with 49.5 ± 7.42% (P = 0.1470 when compared with vehicle) and 85.9 ± 10.3% (P = 0.0016 when compared with vehicle), respectively. Additive effects on relaxation of human corpus cavernosum were seen after preincubation with 1 µm vardenafil. CONCLUSION: The RKI Y-27632 causes a significant relaxation of corpus cavernosum in tissue strips of patients with severe ED. The additive effect of vardenafil and Y-27632 shows that a combined inhibition of Rho-kinase and phosphodiesterase type 5 could be a promising orally administered treatment for severe ED.


Subject(s)
Amides/pharmacology , Enzyme Inhibitors/pharmacology , Erectile Dysfunction/drug therapy , Penis/drug effects , Phosphodiesterase 5 Inhibitors/therapeutic use , Pyridines/pharmacology , Vardenafil Dihydrochloride/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Drug Synergism , Humans , Male , Middle Aged , Treatment Failure
9.
PLoS One ; 10(10): e0141477, 2015.
Article in English | MEDLINE | ID: mdl-26509272

ABSTRACT

Urinary colics from calculosis are frequent and intense forms of pain whose current pharmacological treatment remains unsatisfactory. New and more effective drugs are needed to control symptoms and improve stone expulsion. Recent evidence suggested that the Nitric Oxide (NO) / cyclic guanosine monophosphate (cGMP)/phosphodiesterase type 5 (PDE5) system may contribute to ureteral motility influencing stone expulsion. We investigated if PDE5 inhibitors and sGC stimulators influence ureteral contractility, pain behaviour and stone expulsion in a rat model of ureteral calculosis. We investigated: a) the sex-specific PDE5 distribution in the rat ureter; b) the functional in vitro effects of vardenafil and sildenafil (PDE5 inhibitors) and BAY41-2272 (sGC stimulator) on induced ureteral contractility in rats and c) the in vivo effectiveness of vardenafil and BAY41-2272, alone and combined with ketoprofen, vs hyoscine-N-butylbromide alone or combined with ketoprofen, on behavioural pain indicators and stone expulsion in rats with artificial calculosis in one ureter. PDE5 was abundantly expressed in male and female rats' ureter. In vitro, both vardenafil and BAY41-2272 significantly relaxed pre-contracted ureteral strips. In vivo, all compounds significantly reduced number and global duration of "ureteral crises" and post-stone lumbar muscle hyperalgesia in calculosis rats. The highest level of reduction of the pain behaviour was observed with BAY41-2272 among all spasmolytics administered alone, and with the combination of ketoprofen with BAY41-2272. The percentage of stone expulsion was maximal in the ketoprofen+BAY41-2272 group. The NO/cGMP/PDE5 pathway is involved in the regulation of ureteral contractility and pain behaviour in urinary calculosis. PDE5 inhibitors and sGC stimulators could become a potent new option for treatment of urinary colic pain.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Enzyme Activators/pharmacology , Guanylate Cyclase/metabolism , Lithiasis/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Ureteral Calculi/metabolism , Animals , Autopsy , Behavior, Animal , Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry , Disease Models, Animal , Enzyme Activators/administration & dosage , Female , Gene Expression , Gene Expression Profiling , Guanylate Cyclase/genetics , Lithiasis/drug therapy , Lithiasis/genetics , Lithiasis/pathology , Male , Muscle Contraction/drug effects , Pain , Phosphodiesterase 5 Inhibitors/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Ureter/drug effects , Ureteral Calculi/drug therapy , Ureteral Calculi/genetics , Ureteral Calculi/pathology
10.
Thromb Haemost ; 110(1): 162-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23636219

ABSTRACT

Rivaroxaban is an oral, direct factor Xa inhibitor for the management of thromboembolic disorders. Despite its short half-life, the ability to reverse rivaroxaban anticoagulation could be beneficial in life-threatening emergencies. The potential of prothrombin complex concentrate (PCC; Beriplex®), activated PCC (aPCC; FEIBA®) or recombinant activated factor VII (rFVIIa; NovoSeven®) to reverse rivaroxaban in rats and baboons was investigated. Anaesthetised rats pre-treated with intravenous rivaroxaban (2 mg/kg) received intravenous rFVIIa (100/400 µg/kg), PCC (25/50 U/kg) or aPCC (50/100 U/kg) after initiation of bleeding. Clotting times and bleeding times (BTs) were recorded. Rivaroxaban was administered as an intravenous 0.6 mg/kg bolus followed by continuous 0.6 mg/kg/hour infusion in baboons. Animals received intravenous aPCC 50 U/kg (2 U/kg/minute) or rFVIIa 210 µg/kg. BT and clotting parameters were measured. In rats pretreated with high-dose rivaroxaban, PCC 50 U/kg, aPCC 100 U/kg and rFVIIa 400 µg/kg significantly reduced BT vs rivaroxaban alone (5.4 ± 1.4-fold to 1.5 ± 0.4-fold [p<0.05]; 3.0 ± 0.4-fold to 1.4 ± 0.1-fold [p<0.001]; and 3.5 ± 0.7-fold to 1.7 ± 0.2-fold [p<0.01] vs baseline, respectively). In baboons pre-infused with rivaroxaban and then given aPCC, BT increased by 2.0 ± 0.2-fold and aPCC returned BT to baseline for the duration of its infusion. rFVIIa reduced BT from 2.5 ± 0.3-fold over baseline to 1.7 ± 0.3-fold over baseline. Prolongation of prothrombin time was reduced by PCC, aPCC and rFVIIa in both species. Rivaroxaban reduced thrombin-antithrombin levels; application of PCC and aPCC, but not rFVIIa, increased these levels. In conclusion, PCC, aPCC or rFVIIa have the potential to reverse the anticoagulant and anti-haemostatic effects of rivaroxaban.


Subject(s)
Anticoagulants/administration & dosage , Morpholines/administration & dosage , Thiophenes/administration & dosage , Thromboembolism/drug therapy , Animals , Bleeding Time , Blood Coagulation/drug effects , Blood Coagulation Factors/administration & dosage , Factor VIIa/administration & dosage , Humans , Male , Primates , Prothrombin Time , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Rivaroxaban
11.
J Sex Med ; 10(5): 1268-77, 2013 May.
Article in English | MEDLINE | ID: mdl-23421435

ABSTRACT

INTRODUCTION: Overall efficacy rates of phosphodiesterase type 5 inhibitors (PDE5-i) for erectile dysfunction (ED) are 60-70%. PDE5-i treatment failures currently have to resort to invasive treatment options for restoration of erectile function. AIMS.: To assess changes in the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP)/protein kinase (PKG) pathway in human corpus cavernosum (HCC) of PDE5-i nonresponders compared with healthy controls. To evaluate the effects of BAY 60-4552, a stimulator of soluble guanylate cyclase (sGC), and vardenafil on relaxation of HCC strips from PDE5-i nonresponders. MAIN OUTCOME MEASURES: mRNA expression, morphological localization of the NO/cGMP/PKG pathway, and relaxant capacity of both compounds alone or combined. Analysis of variance, t-test or Mann-Whitney test based upon number of groups and normality of data. METHODS: HCC tissues were harvested after consent from individuals undergoing penile prosthesis implantation (patients) and potent patients undergoing transurethral surgery (healthy controls, needle biopsy). HCC tissues of patients were compared with those of healthy controls for the expression of mRNA coding for PDE5A, eNOS, PKGα1, PKG2, sGCα1, sGCα2, sGCß1, sGCß2, α-smooth muscle actin (aSMA) and ß-actin by quantitative polymerase chain reaction (qPCR). The respective proteins were localized using immunofluorescence. Tissue strips of patients were precontracted with phenylepinephrine followed by incubation with 1 µM of either vardenafil or BAY 60-4552, or both simultaneously. RESULTS: The main targets in the NO/cGMP/sGC pathway were downregulated in PDE5-i nonresponders. The pathway was morphologically located to HCC smooth muscle, of which the overall content was preserved in ED patients based on aSMA expression. BAY 60-4552 and vardenafil have synergistic effects on relaxation of HCC of PDE5-i nonresponders. The main limitation is the small amount of control tissue precluding functional testing on these samples. CONCLUSION: Despite downregulation of the NO/cGMP/PKG pathway, combining BAY 60-4552 with vardenafil significantly enhanced relaxation HCC strips of PDE5-i nonresponders.


Subject(s)
Erectile Dysfunction/physiopathology , Imidazoles/pharmacology , Penile Erection/drug effects , Penis/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Actins/metabolism , Cyclic GMP/metabolism , Drug Synergism , Guanylate Cyclase/metabolism , Humans , Male , Middle Aged , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Penis/physiopathology , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Sulfones/pharmacology , Triazines/pharmacology , Vardenafil Dihydrochloride
12.
J Am Coll Cardiol ; 59(4): 410-7, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22261164

ABSTRACT

OBJECTIVES: This study investigated the effects of 17ß-estradiol (E2) on gene regulation in human cardiac tissues. We hypothesized that a candidate E2 effect is cardiomyocyte (CM)- and sex-specific, conserved between humans and mice, and that E2 impairs contractile function in male CMs only. BACKGROUND: Both men and women produce E2 locally from androgenic precursors. E2 regulates cardiovascular function, but specific mechanisms, protective or harmful, are not fully understood. METHODS: We performed genome-wide expression profiling of E2-treated cardiac tissues from men and women, and studied gene expression and function in CMs from hearts of male and female E2-treated mice. RESULTS: We found 36 E2-dependent genes regulated in a sex-specific manner. Of these, after E2 exposure, the myosin regulatory light chain interacting protein (MYLIP) gene was induced in tissues of men only. Focusing on Mylip and employing isolated mouse CMs, we confirmed our hypotheses that the E2 effect is CM- and sex-specific and conserved between humans and mice. The E2-treatment led to impaired contractile function in male CMs only, which was characterized by increased Mylip mRNA and protein levels, and decreased myosin regulatory light chain (Mrlc) protein. Our report is the first to our knowledge to show that cardiac Mrlc is an in vivo substrate for Mylip, leading to augmented Mrlc ubiquitination. Of relevance, we found that MYLIP expression levels rise with increasing age in hearts of men. CONCLUSIONS: E2 directly influences cardiac gene regulation, and E2 actions may be different between the sexes. Since E2 levels rise in older and/or obese men, pharmacological targeting of MYLIP in men with elevated E2 levels could possibly decrease their risk for the development or progression of cardiovascular disease.


Subject(s)
Estradiol/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Sex Characteristics , Ubiquitin-Protein Ligases/metabolism , Adult , Aging/metabolism , Animals , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myosin Light Chains/metabolism , Transcriptome , Ubiquitination
13.
J Cardiovasc Pharmacol ; 50(1): 75-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17666919

ABSTRACT

BACKGROUND: The antihypertensive and organ-protective effects of the combination of the angiotensin II type 1 receptor blocker losartan and the calcium channel blocker nifedipine were examined in Dahl salt-sensitive rats. METHODS: The rats fed with a high-salt diet developed hypertension accompanied by aorta and heart hypertrophy, and impaired renal function. The animals were treated with losartan (30 mg/kg/day), nifedipine (7.8 mg/kg/day) or with a combination of both drugs for 8 weeks. At the end of the study systolic blood pressure, kidney function, organ weight, and mRNA expression were investigated. RESULTS: Losartan reduced significantly the systolic blood pressure as well as the aorta and left ventricular hypertrophy. Nifedipine and its combination with losartan had similar effects on the systolic blood pressure, aorta and left ventricular hypertrophy but only the combination treatment reduced the expression of transforming growth factor-beta1 in aorta and brain natriuretic peptide in left ventricle significantly. Nifedipine and the combination therapy reduced proteinuria and improved urine creatinine excretion. The expression of collagen III and IV in the kidney was significantly reduced by the combination therapy. CONCLUSION: These results indicate that although losartan and nifedipine were effective in lowering blood pressure and showed moderate organ protection, additional benefits can be expected by combination therapy with both compounds.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Calcium Channel Blockers/pharmacology , Hypertension/drug therapy , Losartan/pharmacology , Nifedipine/pharmacology , Animals , Aorta/metabolism , Blood Pressure/drug effects , Drug Therapy, Combination , Gene Expression , Hypertension/chemically induced , Hypertrophy, Left Ventricular/complications , Hypertrophy, Left Ventricular/drug therapy , Kidney/metabolism , Male , Natriuretic Peptide, Brain/metabolism , Organ Size , RNA, Messenger/metabolism , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary , Transforming Growth Factor beta1/metabolism
14.
BJU Int ; 98(6): 1259-63, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16956354

ABSTRACT

OBJECTIVE: To evaluate the potential of sildenafil, vardenafil and tadalafil, all phosphodiesterase-5 (PDE-5) inhibitors used for treating erectile dysfunction, for treating benign prostatic hyperplasia (BPH) and lower urinary tract symptoms (LUTS). MATERIALS AND METHODS: The mRNA expression of the PDE-5 was determined in rat LUT tissues. The PDE-5 inhibitors were also tested in organ-bath experiments and in a partial bladder outlet obstruction (BOO) rat model in vivo. RESULTS: The highest PDE-5 mRNA expression was in the bladder, followed by the urethra and prostate. PDE-5 inhibitors dose-dependently reduced the contraction of the isolated bladder, urethral and prostate strips. The rank order of potency was vardenafil > sildenafil > tadalafil. In human prostate stromal cells vardenafil inhibited cell proliferation and was more effective than tadalafil and sildenafil. In the BOO model, there was a reduction in the non-voiding contractions after bolus intravenous administration of 3 mg/kg sildenafil and vardenafil. CONCLUSION: These results show that PDE-5 is expressed in LUT tissues. PDE-5 inhibitors induced significant relaxation of these tissues, inhibited the proliferation of human prostate stromal cells and reduced the irritative symptoms of BPH/LUTS in vivo. Therefore, PDE-5 inhibitors could be used as an effective treatment for BPH/LUTS.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Phosphodiesterase Inhibitors/therapeutic use , Prostatic Hyperplasia/drug therapy , Prostatism/drug therapy , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Animals , Carbolines/therapeutic use , Cyclic Nucleotide Phosphodiesterases, Type 5 , Imidazoles/therapeutic use , Male , Piperazines/therapeutic use , Prostatic Hyperplasia/complications , Prostatism/etiology , Purines , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Sulfones/therapeutic use , Tadalafil , Treatment Outcome , Triazines/therapeutic use , Vardenafil Dihydrochloride
15.
Am J Physiol Regul Integr Comp Physiol ; 286(5): R942-51, 2004 May.
Article in English | MEDLINE | ID: mdl-14715486

ABSTRACT

To investigate regional aspects of hypoxic regulation of adrenomedullin (AM) in kidneys, we mapped the distribution of AM in the rat kidney after hypoxia (normobaric hypoxic hypoxia, carbon monoxide, and CoCl(2) for 6 h), anemia (hematocrit lowered by bleeding) and after global transient ischemia for 1 h (unilateral renal artery occlusion and reperfusion for 6 and 24 h) and segmental infarct (6 and 24 h). AM expression and localization was determined in normal human kidneys and in kidneys with arterial stenosis. Hypoxia stimulated AM mRNA expression significantly in rat inner medulla (CO 13 times, 8% O(2) 6 times, and CoCl(2) 8 times), followed by the outer medulla and cortex. AM mRNA level was significantly elevated in response to anemia and occlusion-reperfusion. Immunoreactive AM was associated with the thin limbs of Henle's loop, distal convoluted tubule, collecting ducts, papilla surface epithelium, and urothelium. AM labeling was prominent in the inner medulla after CO and in the outer medulla after occlusion-reperfusion. The infarct border zone was strongly labeled for AM. In cultured inner medullary collecting duct cells, AM mRNA was significantly increased by hypoxia. AM mRNA was equally distributed in human kidney and AM was localized as in the rat kidney. In human kidneys with artery stenosis, AM mRNA was not significantly enhanced compared with controls, but AM immunoreactivity was observed in tubules, vessels, and glomerular cells. In summary, AM expression was increased in the rat kidney in response to hypoxic and ischemic hypoxia in keeping with oxygen gradients. AM was widely distributed in the human kidney with arterial stenosis. AM may play a significant role to counteract hypoxia in the kidney.


Subject(s)
Hypoxia/physiopathology , Ischemia/physiopathology , Kidney/physiology , Peptides/genetics , Renal Artery Obstruction/physiopathology , Adrenomedullin , Anemia/metabolism , Anemia/physiopathology , Animals , Cells, Cultured , Gene Expression , Humans , Hypertension, Renal/metabolism , Hypertension, Renal/physiopathology , Hypoxia/metabolism , Immunohistochemistry , Ischemia/metabolism , Kidney/blood supply , Kidney/cytology , Male , Peptides/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Renal Artery Obstruction/metabolism
16.
Pflugers Arch ; 443(5-6): 754-61, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11889573

ABSTRACT

Changes of cell volume and intracellular calcium concentration ([Ca(2+)](i)) in immortalized thick ascending limb of Henle's loop (TALH) cells were monitored using confocal laser scanning microscopy and fura-2 fluorescence, respectively. Reduction of the extracellular osmolarity from 600 to 300 mosmol/l induced cell swelling followed by regulatory volume decrease (RVD). Simultaneously, the [Ca(2+)](i) increased transiently. The calcium rise was not observed in calcium-free solution or in the presence of nifedipine, indicating that the change was, in the first place, due to the activation of a calcium influx. Application of ATP or caffeine in isotonic solutions increased transiently the [Ca(2+)](i) which revealed the existence of stores in TALH cells sensitive to inositol-1,4,5 trisphosphate (IP(3)) and ryanodine. To examine the possibility that the calcium influx might induce calcium release, manganese quenching experiments were performed. In hypotonic calcium-free solutions, the decay of the calcium-insensitive and calcium-sensitive fluorescence occurred simultaneously. In the presence of extracellular calcium however, the calcium-sensitive wavelength revealed initial calcium influx followed by a calcium release from intracellular stores. Thus, the calcium influx was a prerequisite for the calcium release. We conclude that calcium-induced calcium release participates in global calcium signalling during RVD of TALH cells.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Loop of Henle/cytology , Loop of Henle/metabolism , Water-Electrolyte Balance/physiology , Animals , Calcium/pharmacology , Calcium Signaling/drug effects , Cell Line , Hypotonic Solutions/pharmacology , Osmotic Pressure , Rabbits
17.
Pflugers Arch ; 443(5-6): 779-90, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11889576

ABSTRACT

We studied the effects of hypertonic stress on ion transport and cell volume regulation (regulatory volume increase; RVI) in the human tumor cell-line HepG2. Ion conductances were monitored in intracellular current-clamp measurements with rapid ion-substitutions and in whole-cell patch-clamp recordings; intracellular pH buffering capacity and activation of Na(+)/H(+) antiport were determined fluorometrically; the rates of Na(+)-K(+)-2Cl(-) symport and Na(+)/K(+)-ATPase were quantified on the basis of time-dependent and furosemide- or ouabain-sensitive (86)Rb(+) uptake, respectively; changes in cell volume were recorded by means of confocal laser-scanning microscopy. It was found that hypertonic conditions led to the activation of a cation conductance that was inhibited by Gd(3+), flufenamate as well as amiloride, but not by benzamil or ethyl-isopropyl-amiloride (EIPA). Most likely, this cation conductance was non-selective for Na(+) over K(+). Hypertonic stress did not change K(+) conductance, whereas possible changes in Cl(-) conductance remain ambiguous. The contribution of Na(+)/H(+)antiport to the RVI process appeared to be minor. Under hypertonic conditions an approximately 3.5-fold stimulation of Na(+)-K(+)-2Cl(-)symport was observed but this transporter did not significantly contribute to the overall RVI process. Hypertonic stress did not increase the activity of Na(+)/K(+)-ATPase, which even under isotonic conditions appeared to be working at its limit. It is concluded that the main mechanism in the RVI of HepG2 cells is the activation of a novel non-selective cation conductance. In contrast, there is little if any contribution of K(+) conductance, Na(+)/H(+) antiport, Na(+)-K(+)-2Cl(-) symport, and Na(+)/K(+)-ATPase to this process.


Subject(s)
Carcinoma, Hepatocellular , Hepatocytes/metabolism , Liver Neoplasms , Water-Electrolyte Balance/physiology , Alkalies/metabolism , Amiloride/pharmacology , Diuretics/pharmacology , Hepatocytes/cytology , Humans , Hydrogen-Ion Concentration , Hypertonic Solutions/pharmacology , Osmotic Pressure , Patch-Clamp Techniques , Potassium/metabolism , Sodium/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Tumor Cells, Cultured , Water-Electrolyte Balance/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...