Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 161(1): 207-28, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20718751

ABSTRACT

BACKGROUND AND PURPOSE: Recently identified antagonists of the urotensin-II (U-II) receptor (UT) are of limited utility for investigating the (patho)physiological role of U-II due to poor potency and limited selectivity and/or intrinsic activity. EXPERIMENTAL APPROACH: The pharmacological properties of two novel UT antagonists, GSK1440115 and GSK1562590, were compared using multiple bioassays. KEY RESULTS: GSK1440115 (pK(i)= 7.34-8.64 across species) and GSK1562590 (pK(i)= 9.14-9.66 across species) are high affinity ligands of mammalian recombinant (mouse, rat, cat, monkey, human) and native (SJRH30 cells) UT. Both compounds exhibited >100-fold selectivity for UT versus 87 distinct mammalian GPCR, enzyme, ion channel and neurotransmitter uptake targets. GSK1440115 showed competitive antagonism at UT in arteries from all species tested (pA(2)= 5.59-7.71). In contrast, GSK1562590 was an insurmountable UT antagonist in rat, cat and hUT transgenic mouse arteries (pK(b)= 8.93-10.12 across species), but a competitive antagonist in monkey arteries (pK(b)= 8.87-8.93). Likewise, GSK1562590 inhibited the hU-II-induced systemic pressor response in anaesthetized cats at a dose 10-fold lower than that of GSK1440115. The antagonistic effects of GSK1440115, but not GSK1562590, could be reversed by washout in rat isolated aorta. In ex vivo studies, GSK1562590 inhibited hU-II-induced contraction of rat aorta for at least 24 h following dosing. Dissociation of GSK1562590 binding was considerably slower at rat than monkey UT. CONCLUSIONS AND IMPLICATIONS: Whereas both GSK1440115 and GSK1562590 represent high-affinity/selective UT antagonists suitable for assessing the (patho)physiological role of U-II, only GSK1562590 exhibited sustained UT residence time and improved preclinical efficacy in vivo.


Subject(s)
Benzamides/pharmacology , Benzoxazines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Urotensins/metabolism , Animals , Arteries/drug effects , Arteries/physiology , Benzamides/chemistry , Benzoates/chemistry , Benzoates/pharmacology , Benzoxazines/chemistry , Cats , Cell Line , Dose-Response Relationship, Drug , Haplorhini , Humans , Male , Mice , Mice, Transgenic , Molecular Structure , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/agonists , Tachykinins , Vasoconstriction
2.
Biomarkers ; 12(1): 87-112, 2007.
Article in English | MEDLINE | ID: mdl-17438656

ABSTRACT

The assessment of target organ damage is important in defining the optimal treatment of hypertension and blood pressure-related cardiovascular disease. The aims of the present study were (1) to investigate candidate biomarkers of target organ damage, osteopontin (OPN) and plasminogen activator inhibitor-1 (PAI-1), in models of malignant hypertension with well characterized end-organ pathology; and (2) to evaluate the effects of chronic treatment with a p38 MAPK inhibitor. Gene expression, plasma concentrations, and renal immunohistochemical localization of OPN and PAI-1 were measured in stroke-prone spontaneously hypertensive rats on a salt-fat diet (SFD SHR-SP) and in spontaneously hypertensive rats receiving N(omega)-nitro-L-arginine methyl ester (L-NAME SHR). Plasma concentrations of OPN and PAI-1 increased significantly in SFD SHR-SP and L-NAME SHR as compared with controls, (2.5-4.5-fold for OPN and 2.0-9.0-fold for PAI-1). The plasma levels of OPN and PAI-1 were significantly correlated with the urinary excretion of albumin (p < 0.0001). Elevations in urinary albumin, plasma OPN and PAI-1 were abolished by chronic treatment (4-8 weeks) with a specific p38 MAPK inhibitor, SB-239063AN. OPN immunoreactivity was localized predominantly in the apical portion of tubule epithelium, while PAI-1 immunoreactivity was robust in glomeruli, tubules and renal artery endothelium. Treatment with the p38 MAPK inhibitor significantly reduced OPN and PAI-1 protein expression in target organs. Kidney gene expression was increased for OPN (4.9- and 7.9-fold) and PAI-1 (2.8- and 11.5-fold) in SFD SHR-SP and L-NAME SHR, respectively. In-silico pathway analysis revealed that activation of p38 MAPK was linked to OPN and PAI-1 via SPI, c-fos and c-jun; suggesting that these pathways may play an important role in p38 MAPK-dependent hypertensive renal dysfunction. The results suggest that enhanced OPN and PAI-1 expression reflects end-organ damage in hypertension and that suppression correlates with end-organ protection regardless of overt antihypertensive action.


Subject(s)
Biomarkers/analysis , Hypertension/metabolism , Osteopontin/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Enzyme-Linked Immunosorbent Assay , Hypertension/physiopathology , Immunohistochemistry , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred SHR
3.
J Cardiovasc Pharmacol ; 38(4): 606-17, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11588531

ABSTRACT

Evidence suggests that endothelin receptor antagonists may have therapeutic potential for the chronic treatment of heart failure. In the current study, the effects of an orally active mixed endothelin-A/endothelin-B (ETA /ETB ) receptor antagonist (enrasentan) were assessed in a model of cardiac hypertrophy and dysfunction (spontaneously hypertensive stroke prone rats) maintained on a high-salt/high-fat diet. Echocardiography was used to quantify cardiac performance and left ventricular dimensions. Enrasentan (1,200 and 2,400 parts per million in the high-salt/high-fat diet) had no significant effects on body weight and systolic blood pressure. However, increases in heart rate were not observed in the enrasentan-treated groups at 12 weeks (p < 0.05). Enrasentan-treated groups exhibited significantly improved survival (90-95% vs. 30% [control rats] at 18 weeks; p < 0.001). Enrasentan treatments also increased stroke volume (at 8, 12, and 16 weeks) and cardiac index (at 8 and 16 weeks) 33-50% and 45-63%, respectively. Enrasentan treatments reduced the relative wall thickness (14-27% at 8 and 12 weeks), ratio of left ventricular mass to body weight (20% at 12 weeks), and ratio of terminal heart weight to body weight (16-23%, p < 0.05). Finally, circulating aldosterone concentration (54-57%) and proANF fragment (33%) were reduced in enrasentan-treated groups (54-57% and 33%, respectively). Mixed ETA /ETB receptor antagonism improves cardiac performance and attenuates ventricular remodeling and premature mortality in an aggressive hypertension model.


Subject(s)
Carboxylic Acids/therapeutic use , Hypertension/drug therapy , Hypertrophy, Left Ventricular/drug therapy , Indans/therapeutic use , Ventricular Remodeling/drug effects , Aldosterone/blood , Animals , Atrial Natriuretic Factor/blood , Blood Pressure/drug effects , Blood Pressure/physiology , Endothelin Receptor Antagonists , Heart Rate/drug effects , Heart Rate/physiology , Hypertension/blood , Hypertension/mortality , Hypertrophy, Left Ventricular/blood , Hypertrophy, Left Ventricular/mortality , Male , Myocardium/metabolism , Protein Precursors/blood , Rats , Rats, Inbred SHR , Survival Rate , Ventricular Remodeling/physiology
4.
Circulation ; 104(11): 1292-8, 2001 Sep 11.
Article in English | MEDLINE | ID: mdl-11551882

ABSTRACT

BACKGROUND: Numerous pathological mediators of cardiac hypertrophy (eg, neurohormones, cytokines, and stretch) have been shown to activate p38 MAPK. The purpose of the present study was to examine p38 MAPK activation and the effects of its long-term inhibition in a model of hypertensive cardiac hypertrophy/dysfunction and end-organ damage. METHODS AND RESULTS: In spontaneously hypertensive stroke-prone (SP) rats receiving a high-salt/high-fat diet (SFD), myocardial p38 MAPK was activated persistently during the development of cardiac hypertrophy and inactivated during decompensation. Long-term oral treatment of SFD-SP rats with a selective p38 MAPK inhibitor (SB239063) significantly enhanced survival over an 18-week period compared with the untreated group (100% versus 50%). Periodic echocardiographic analysis revealed a significant reduction in LV hypertrophy and dysfunction in the SB239063-treatment groups. Little or no difference in blood pressure was noted in the treatment or vehicle groups. Basal and stimulated (lipopolysaccharide) plasma tumor necrosis factor-alpha concentrations were reduced in the SB239063-treatment groups. In vitro vasoreactivity studies demonstrated a significant preservation of endothelium-dependent relaxation in animals treated with the p38 MAPK inhibitor without effects on contraction or NO-mediated vasorelaxation. Proteinuria and the incidence of stroke (53% versus 7%) were also reduced significantly in the SB239063-treated groups. CONCLUSIONS: These results demonstrate a crucial role for p38 MAPK in hypertensive cardiac hypertrophy and end-organ damage. Interrupting its function with a specific p38 MAPK inhibitor halts clinical deterioration.


Subject(s)
Cardiomegaly/physiopathology , Hypertension/physiopathology , Mitogen-Activated Protein Kinases/metabolism , Animals , Cardiomegaly/enzymology , Cardiomegaly/mortality , Disease Models, Animal , Dose-Response Relationship, Drug , Echocardiography , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Activation , Heart/drug effects , Heart/physiopathology , Hemodynamics/drug effects , Imidazoles/pharmacology , Kidney/drug effects , Kidney/physiopathology , Lipopolysaccharides/pharmacology , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Myocardium/metabolism , Myocardium/pathology , Phosphorylation , Proteinuria/prevention & control , Proteinuria/urine , Pyrimidines/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/pathology , Stroke/prevention & control , Survival Rate , Time Factors , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vasodilation/drug effects , p38 Mitogen-Activated Protein Kinases
6.
Cardiovasc Res ; 50(3): 525-37, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11376628

ABSTRACT

OBJECTIVE: Eprosartan is a selective angiotensin II type I receptor antagonist approved for the treatment of hypertension. In the present studies, eprosartan's ability to provide end-organ protection was evaluated in a model of cardiomyopathy and renal failure in stroke-prone rats (SP). METHODS: SP were fed a high fat (24.5% in food) and high salt (1% in water) diet (SFD). Eprosartan (60 mg/kg/day) or vehicle (saline control) (n = 25/group) was administered by intraperitoneally-implanted minipumps to these SP on the SFD for 12 weeks. Normal diet fed SP and WKY rats (n = 25/group) were also included for comparison (i.e. served as normal controls). Mortality, hemodynamics, and both renal and cardiac function and histopathology were monitored in all treatment groups. RESULTS: Eprosartan decreased the severely elevated arterial pressure (-12%; P < 0.05) produced by SFD but did not affect heart rate. Vehicle-treated SP-SFD control rats exhibited significant weight loss (-13%; P < 0.05) and marked mortality (50% by week 6 and 95% by week 9; P < 0.01). Eprosartan-treated SP-SFD rats maintained normal weight, and exhibited zero mortality at week 12 and beyond. Eprosartan prevented the increased urinary protein excretion (P < 0.05) that was observed in vehicle-treated SP-SFD rats. Echocardiographic (i.e. 2-D guided M-mode) evaluation indicated that SP-SFD vehicle control rats exhibited increased septal (+22.2%) and posterior left ventricular wall (+30.0%) thickness, and decreased left ventricular chamber diameter (-15.9%), chamber volume (-32.7%), stroke volume (-48.7%) and ejection fraction (-22.3%), and a remarkable decrease in cardiac output (-59.3%) compared to controls (all P < 0.05). These same parameters in eprosartan-treated SP-SFD rats were normal and differed markedly and consistently from vehicle-treated SP-SFD rats (i.e. treatment prevented pathology; all P < 0.05). Cardiac-gated MRI data confirmed the ability of eprosartan to prevent cardiac pathology/remodeling (P < 0.05). Histopathological analysis of hearts and kidneys indicated that eprosartan treatment significantly reduced end-organ damage (P < 0.01) and provided corroborative evidence that eprosartan reduced remodeling of these organs. Vehicle-treated SP-SFD rats exhibited a 40% increase in the plasma level of pro-atrial natiuretic factor that was reduced to normal by eprosartan (P < 0.05). CONCLUSION: These data demonstrate that eprosartan, at a clinically relevant dose, provides significant end-organ protection in the severely hypertensive stroke-prone rat. It preserves cardiac and renal structural integrity, reduces cardiac hypertrophy and indices of heart failure, maintains normal function of the heart and kidneys, and eliminates premature mortality due to hypertension-induced end-organ failure.


Subject(s)
Acrylates/therapeutic use , Antihypertensive Agents/therapeutic use , Cardiomegaly/drug therapy , Hypertension/drug therapy , Imidazoles/therapeutic use , Thiophenes , Animals , Atrial Natriuretic Factor/blood , Blood Pressure/drug effects , Body Weight/drug effects , Heart Rate/drug effects , Kidney/pathology , Magnetic Resonance Imaging , Male , Myocardium/pathology , Natriuresis/physiology , Organ Size/drug effects , Peptide Fragments/blood , Protein Precursors/blood , Proteinuria/prevention & control , Rats , Rats, Inbred SHR , Stroke/prevention & control , Survival Rate , Ventricular Remodeling/drug effects
7.
J Pharmacol Exp Ther ; 296(3): 768-75, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181905

ABSTRACT

Calcitonin gene-related peptide (CGRP), a potent vasodilatory and cardiotonic peptide, has a potential role for CGRP in diverse physiologic and pathophysiologic situations such as congestive heart failure, diabetes, migraine, and neurogenic inflammation. Although a peptide CGRP receptor antagonist, CGRP(8-37,) is available, its utility presents significant limitations for these indications. Here, we describe the properties of SB-(+)-273779 [N-methyl-N-(2-methylphenyl)-3-nitro-4-(2-thiazolylsulfinyl)nitrobenzanilide], a selective nonpeptide antagonist of CGRP(1) receptor. SB-(+)-273779 inhibited (125)I-labeled CGRP binding to SK-N-MC (human neuroblastoma cells) and human cloned CGRP(1) receptor with K(i) values of 310 +/- 40 and 250 +/-15 nM, respectively. SB-(+)-273779 also inhibited CGRP (3 nM)-activated adenylyl cyclase in these systems with IC(50) values of 390 +/-10 nM (in SK-N-MC) and 210 +/-16 nM (recombinant human CGRP receptors). Prolonged treatment (>30 min) of SK-N-MC cells with SB-(+)-273779 followed by extensive washing resulted in reduction in maximum CGRP-mediated adenylyl cyclase activity, suggesting that this compound has irreversible binding characteristics. In addition, SB-(+)-273779 antagonized CGRP-mediated 1) stimulation of intracellular Ca(2+) in recombinant CGRP receptors in HEK-293 cells, 2) inhibition of insulin-stimulated [(14)C]deoxyglucose uptake in L6 cells, 3) vasodilation in rat pulmonary artery, and 4) decrease in blood pressure in anesthetized rats. SB-(+)-273779 tested at 3 microM had no significant affinity for calcitonin, endothelin, angiotensin II, and alpha-adrenergic receptors under standard ligand binding assays. SB-(+)-273779 also did not inhibit forskolin and pituitary adenylate cyclase-activating polypeptide. These results suggest that SB-(+)-273779 is a valuable tool for studying CGRP-mediated functional responses in complex biological systems.


Subject(s)
Anilides/pharmacology , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Thiazoles/pharmacology , Anilides/chemistry , Animals , Blood Pressure/drug effects , Calcitonin Gene-Related Peptide/metabolism , Humans , Male , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship , Thiazoles/chemistry , Tumor Cells, Cultured , Vasodilation/drug effects
8.
J Cardiovasc Pharmacol ; 36(5 Suppl 1): S163-6, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11078367

ABSTRACT

The observation that the novel G-protein-coupled receptor (GPCR) GPR14 and its cognate ligand, urotensin-II (U-II), are expressed within the mammalian vasculature raises the possibility that they may influence cardiohemodynamic homeostasis. To this end, this study examined the vasoactive properties of U-II in rodents, dogs and primates. In vitro, human U-II was a sustained vasoconstrictor with a potency (pD2s < or = 9) approximately an order of magnitude greater than that seen with endothelin-1 (ET-1), making it one of the most, if not the most, potent mammalian vasoconstrictor identified to date. However, in vitro responses exhibited significant anatomical and/or species-dependency, that is, human U-II was a selective 'aorto-coronary' vasoconstrictor in rats and dogs, inactive in mice and contracted all primate arteries studied. In vivo, this peptide evoked a complex, dose-dependent hemodynamic response in the anesthetized primate, culminating in severe myocardial depression and fatal circulatory collapse. As such, U-II may represent a novel neurohumoral regulator of mammalian cardiovascular physiology and pathology in particular disorders characterized by aberrant vascular smooth muscle and/or myocardial function.


Subject(s)
Urotensins/pharmacology , Vasoconstriction/drug effects , Animals , Dogs , Dose-Response Relationship, Drug , Hemodynamics/drug effects , Humans , In Vitro Techniques , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Species Specificity
9.
Br J Pharmacol ; 131(7): 1262-74, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090097

ABSTRACT

1. Urotensin-II (U-II) and its G-protein-coupled receptor, GPR14, are expressed within mammalian cardiac and peripheral vascular tissue and, as such, may regulate mammalian cardiovascular function. The present study details the vasoconstrictor profile of this cyclic undecapeptide in different vascular tissues isolated from a diverse range of mammalian species (rats, mice, dogs, pigs, marmosets and cynomolgus monkeys). 2. The vasoconstrictor activity of human U-II was dependent upon the anatomical origin of the vessel studied and the species from which it was isolated. In the rat, constrictor responses were most pronounced in thoracic aortae and carotid arteries: -log[EC(50)]s 9.09+/-0.19 and 8.84+/-0.21, R(max)s 143+/-21 and 67+/-26% 60 mM KCl, respectively (compared, for example, to -log[EC(50)] 7.90+/-0.11 and R(max) 142+/-12% 60 mM KCl for endothelin-1 [ET-1] in thoracic aortae). Responses were, however, absent in mice aortae (-log[EC(50)] <6.50). These findings were further contrasted by the observation that U-II was a 'coronary-selective' spasmogen in the dog (-log[EC(50)] 9.46+/-0.11, R(max) 109+/-23% 60 mM KCl in LCX coronary artery), yet exhibited a broad spectrum of vasoconstrictor activity in arterial tissue from Old World monkeys (-log[EC(50)]s range from 8.96+/-0.15 to 9.92+/-0.13, R(max)s from 43+/-16 to 527+/-135% 60 mM KCl). Interestingly, significant differences in reproducibility and vasoconstrictor efficacy were seen in tissue from pigs and New World primates (vessels which responded to noradrenaline, phenylephrine, KCl or ET-1 consistently). 3. Thus, human U-II is a potent, efficacious vasoconstrictor of a variety of mammalian vascular tissues. Although significant species/anatomical variations exist, the data support the hypothesis that U-II influences the physiological regulation of mammalian cardiovascular function.


Subject(s)
Blood Vessels/drug effects , Urotensins/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/physiology , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Arteries/drug effects , Arteries/physiology , Blood Vessels/physiology , Callithrix , Carotid Arteries/drug effects , Carotid Arteries/physiology , Coronary Vessels/drug effects , Coronary Vessels/physiology , Dogs , Dose-Response Relationship, Drug , Femoral Artery/drug effects , Femoral Artery/physiology , Humans , In Vitro Techniques , Macaca fascicularis , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Pulmonary Veins/drug effects , Pulmonary Veins/physiology , Rats , Swine , Trachea/drug effects , Trachea/physiology , Veins/drug effects , Veins/physiology
10.
J Neurosci Res ; 59(2): 238-46, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10650882

ABSTRACT

A short duration of ischemia (i.e., ischemic preconditioning) was shown to result in significant tolerance to subsequent ischemic injury. Since previous reports suggest that interleukin-1beta (IL-1beta) may be involved in both ischemic damage and neuroprotection, the present work examined the expression of IL-1beta mRNA in cortical brain tissue after an established preconditioning (PC) stimulus known to produce significant brain tolerance to focal stroke after 1-7 days. Significant induction of IL-1beta mRNA was observed in the ipsilateral cortex at 6 hr (87+/-9 copies of the mRNA per microgram of brain tissue compared to 16+/-5 copies in sham-operated samples, P < 0.001, n = 4) and 8 hr (46+/-4 copies, P < 0.01, n = 4) after PC by means of real-time Taqman polymerase chain reaction (PCR). The peak expression of IL-1beta mRNA after PC was significantly (P < 0.01) lower than that after permanent occlusion of the middle cerebral artery (MCAO), i.e., 87+/-9 and 546+/-92 copies of RNA per microgram tissue at peak levels for PC and focal stroke, respectively. Immunohistochemistry studies revealed a parallel induction of IL-1beta in the ipsilateral cortex after PC. The maximal expression of IL-1beta was observed during the first week post-PC, showing marked parallelism with the duration of ischemic tolerance. These data suggest that the significant but low levels of IL-1beta induction after PC may contribute to ischemic brain tolerance.


Subject(s)
Brain Ischemia/genetics , Brain Ischemia/physiopathology , Interleukin-1/genetics , Polymerase Chain Reaction/methods , Animals , Astrocytes/physiology , Cerebral Cortex/blood supply , Cerebral Cortex/chemistry , Cerebral Cortex/physiology , DNA Primers , Fluorescent Antibody Technique , Gene Expression/genetics , Interleukin-1/analysis , Ischemic Preconditioning , RNA, Messenger/analysis , Rats , Stroke/genetics , Stroke/physiopathology , Taq Polymerase , Time Factors
11.
Nature ; 401(6750): 282-6, 1999 Sep 16.
Article in English | MEDLINE | ID: mdl-10499587

ABSTRACT

Urotensin-II (U-II) is a vasoactive 'somatostatin-like' cyclic peptide which was originally isolated from fish spinal cords, and which has recently been cloned from man. Here we describe the identification of an orphan human G-protein-coupled receptor homologous to rat GPR14 and expressed predominantly in cardiovascular tissue, which functions as a U-II receptor. Goby and human U-II bind to recombinant human GPR14 with high affinity, and the binding is functionally coupled to calcium mobilization. Human U-II is found within both vascular and cardiac tissue (including coronary atheroma) and effectively constricts isolated arteries from non-human primates. The potency of vasoconstriction of U-II is an order of magnitude greater than that of endothelin-1, making human U-II the most potent mammalian vasoconstrictor identified so far. In vivo, human U-II markedly increases total peripheral resistance in anaesthetized non-human primates, a response associated with profound cardiac contractile dysfunction. Furthermore, as U-II immunoreactivity is also found within central nervous system and endocrine tissues, it may have additional activities.


Subject(s)
GTP-Binding Proteins/agonists , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/agonists , Receptors, G-Protein-Coupled , Urotensins/pharmacology , Vasoconstrictor Agents/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary , GTP-Binding Proteins/genetics , Humans , Macaca fascicularis , Male , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Tissue Distribution , Urotensins/metabolism , Vasoconstrictor Agents/metabolism
12.
J Vasc Res ; 36(2): 120-5, 1999.
Article in English | MEDLINE | ID: mdl-10213907

ABSTRACT

Bone morphogenetic proteins (BMPs) and their serine/threonine kinase receptors have been identified in atherosclerotic arteries and vascular smooth muscle cells, respectively. Thus, BMPs (the largest subfamily of the TGF-beta superfamily) have been implicated in the pathogenesis of atherosclerosis. However, the origins of BMP biosynthesis and the functional roles of BMP in blood vessels are unclear. The present study explored BMP-2 gene expression in various human blood vessels and vascular cell types. Functional in vitro studies were also performed to determine the effects of recombinant human BMP-2 on migration (transwell assay) and proliferation ([3H]-thymidine incorporation) of human aortic vascular smooth muscle cells (HASMC). RT-PCR experiments revealed BMP-2 gene expression in normal and atherosclerotic human arteries as well as cultured human aortic and coronary vascular smooth muscle cells, human umbilical vein endothelial cells (HUVECs) and human macrophages. In cellular migration studies, incubation with BMP-2 produced efficacious (

Subject(s)
Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Gene Expression/physiology , Muscle, Smooth, Vascular/drug effects , Transforming Growth Factor beta , Aorta/cytology , Aorta/drug effects , Aorta/physiology , Bone Morphogenetic Protein 2 , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Drug Synergism , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Platelet-Derived Growth Factor/pharmacology , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/physiology
13.
J Pharmacol Exp Ther ; 289(1): 48-53, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10086986

ABSTRACT

Clinical studies conducted with carvedilol suggest that beta-adrenoceptor antagonism is an effective therapeutic approach to the treatment of heart failure. However, many beta-adrenoceptor antagonists are weak partial agonists and possess significant intrinsic sympathomimetic activity (ISA), which may be problematic in the treatment of heart failure. In the present study, the ISAs of bucindolol, xamoterol, bisoprolol, and carvedilol were evaluated and compared in normal rats [Sprague-Dawley (SD)], in rats with confirmed heart failure [spontaneously hypertensive heart failure (SHHF)], and in isolated neonatal rat cardiomyocytes. At equieffective beta1-adrenolytic doses, the administration of xamoterol and bucindolol produced a prolonged, equieffective, and dose-related increase in heart rate in both pithed SD rats (ED50 = 5 and 40 microgram/kg, respectively) and SHHF rats (ED50 = 6 and 30 microgram/kg, respectively). The maximum effect of both compounds in SHHF rats was approximately 50% of that observed in SD rats. In contrast, carvedilol and bisoprolol had no significant effect on resting heart rate in the pithed SD or SHHF rat. The maximum increase in heart rate elicited by xamoterol and bucindolol was inhibited by treatment with propranolol, carvedilol, and betaxolol (beta1-adrenoceptor antagonist) but not by ICI 118551 (beta2-adrenoceptor antagonist) in neonatal rat. When the beta-adrenoceptor-mediated cAMP response was examined in cardiomyocytes, an identical partial agonist/antagonist response profile was observed for all compounds, demonstrating a strong correlation with the in vivo results. In contrast, GTP-sensitive ligand binding and tissue adenylate cyclase activity were not sensitive methods for detecting beta-adrenoceptor partial agonist activity in the heart. In summary, xamoterol and bucindolol, but not carvedilol and bisoprolol, exhibited direct beta1-adrenoceptor-mediated ISA in normal and heart failure rats.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Heart Failure/physiopathology , Heart Rate/drug effects , Adrenergic beta-Agonists/pharmacology , Animals , Animals, Newborn , Bisoprolol/pharmacology , Carbazoles/pharmacology , Carvedilol , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Decerebrate State , Dose-Response Relationship, Drug , Heart Failure/metabolism , Iodocyanopindolol , Ligands , Male , Myocardium/metabolism , Propanolamines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Xamoterol/pharmacology
14.
Hypertension ; 33(1 Pt 2): 402-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9931137

ABSTRACT

-Responsiveness to beta-adrenergic stimulation is reduced in the failing human myocardium. This results principally from reduced beta-adrenergic receptor (betaAR) density, elevated beta-adrenergic receptor kinase 1 (betaARK1) levels, and functional uncoupling of remaining receptors. The temporal nature of changes in the human myocardial beta-adrenergic system relative to onset of symptomatic heart failure (HF) has been difficult to discern. A relatively new model of HF, the spontaneously hypertensive heart failure (SHHF) rat spontaneously and reproducibly develops left ventricular hypertrophy (LVH) and progresses to HF, thus enabling longitudinal studies to examine the cellular and molecular bases for hypertension-induced cardiac hypertrophy and subsequent HF. The purpose of this study was to examine age-dependent changes in the betaAR system in this model. Lean male SHHF rats at 3, 7, 14, and 20 months were compared with age-matched Sprague-Dawley (SD) control rats ([C]; 4 animals/group). At all ages the SHHF rats had elevated blood pressures and left ventricular end-diastolic pressure relative to the SD control rats (P<0.05). Compared with age-matched SD control rats, LVH was evident by 3 months in SHHF rats; 20-month-old SHHF rats had significantly greater LVH compared with the other SHHF rat groups. beta-adrenergic responsiveness (maximal heart rate to isoproterenol) was reduced only in 20-month-old SHHF rats. betaARK1 protein levels and activity were elevated at 14 months (162+/-10% and 195+/-20% C, respectively), and betaARK1 protein remained elevated at 20 months (140+/-14% C). In contrast, G protein-coupled receptor kinase 5, a second receptor kinase in the heart, remained unchanged at all ages. betaAR density did not change with age in the SD control rats and was similar in the SHHF rats until 20 months of age when the receptor number was reduced (30+/-1%). These data indicate that cardiac dysfunction is coincident with reduced betaAR density. Importantly, cardiac dysfunction was preceded by elevated betaARK1 levels and activity, thus suggesting that betaARK1 may be a precipitating factor in the transition from hypertension-induced compensatory cardiac hypertrophy to HF. Furthermore, these results indicate that the SHHF rat is a powerful model for use in examination of the mechanisms involved in alterations of beta-adrenergic signaling that occur in human HF.


Subject(s)
Heart Failure/physiopathology , Heart/physiopathology , Hemodynamics , Hypertension/physiopathology , Receptors, Adrenergic, beta/metabolism , Aging/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Heart Failure/genetics , Heart Rate/drug effects , Heart Ventricles , Humans , Hypertension/genetics , Isoproterenol/pharmacology , Male , Myocardial Contraction , Myocardium/metabolism , Organ Size , Rats , Rats, Inbred Strains , Rats, Sprague-Dawley , Ventricular Function, Left , beta-Adrenergic Receptor Kinases
15.
Circ Res ; 83(3): 264-9, 1998 Aug 10.
Article in English | MEDLINE | ID: mdl-9710118

ABSTRACT

During the development of atherosclerotic lesions, lipoprotein(a) [Lp(a)], a highly atherogenic lipoprotein, accumulates within fibrin clots attached to blood vessel walls. As Lp(a) accumulates within the fibrin clot with time, fatty streaks are formed that develop into occlusive atherosclerotic plaques. It is not understood, however, which mechanisms are involved in the binding of Lp(a) to fibrin and, hence, the stable incorporation of Lp(a) into the fibrin clot. The results of the present study demonstrate that factor XIIIa, a transglutaminase that catalyzes the formation of amide bonds between endo-gamma-glutaminyl and endo-epsilon-lysyl residues of proteins, is capable of cross-linking Lp(a) to fibrinogen, the soluble precursor of fibrin. Biochemical assays were conducted to demonstrate that factor XIIIa cross-links Lp(a) with fibrinogen in a time- and concentration-dependent manner. Additionally, immunohistochemical studies revealed that factor XIII protein expression colocalizes with Lp(a) expression in human atherosclerotic plaques. It is proposed that factor XIIIa-mediated cross-linking of Lp(a) to fibrin effectively increases the local concentration of Lp(a) within a fibrin clot. The accumulation of Lp(a) within the blood vessel promotes an antifibrinolytic environment, foam cell formation, the generation of a fatty streak, and an increase in smooth muscle cell content, all of which may contribute to the pathogenesis of atherosclerosis.


Subject(s)
Arteriosclerosis/metabolism , Fibrinogen/metabolism , Lipoprotein(a)/metabolism , Transglutaminases/metabolism , Blotting, Western , Cross-Linking Reagents/metabolism , Enzyme-Linked Immunosorbent Assay , Fibrin/metabolism , Humans , In Vitro Techniques
16.
J Cardiovasc Pharmacol ; 31 Suppl 1: S149-57, 1998.
Article in English | MEDLINE | ID: mdl-9595425

ABSTRACT

Some endothelin (ET) receptor antagonists have been reported to elevate plasma immunoreactive endothelin-1 (irET-1). However, there is no information regarding the effects of ET receptor antagonists on cerebrospinal fluid (CSF) levels. To better understand the regulation of circulating and CSF ET-1, the effects of several nonpeptide antagonists with high, intermediate, or low affinity at the ETB receptor, as well as the potent ETB selective agonist sarafotoxin 6c (S6c), were characterized and compared. The effects of SB209670 (Ki ETA = 0.2 nM; Ki ETB = 12 nM), SB217242 (Ki ETA = 1.1 nM; Ki ETB = 111 nM), SB234551 (Ki ETA = 0.1 nM; Ki ETB = 500 nM), SB247083 (Ki ETA = 0.4 nM; Ki ETB = 467 nM), and S6c (Ki ETA = 950 nM; Ki ETB = 1 nM) on plasma irET-1 were determined by ELISA in the anesthetized dog after i.v. administration. Systemic administration of equivalent doses of the nonpeptide ET receptor antagonists produced dose-related elevations in plasma irET-1 which were correlated (p = 0.019) with affinity at the ETB receptor. There was no significant correlation with affinity at the ETA receptor. In addition, the plasma irET-1 and ET antagonist concentrations were linearly correlated (r = 0.98) throughout the time course after antagonist administration. There was no evidence of densensitization after three bolus administrations performed at 2-h intervals (SB209670, 1 and 3 mg/kg i.v.). Elevations in plasma irET-1 (four- to fivefold) were also observed after systemic administration of S6c (1 nmol/kg i.v.). The administration of L-NAME (200 micrograms/kg/min for 30 min), an inhibitor of nitric oxide (NO) synthase, increased blood pressure (33%) but did not alter plasma irET-1. In contrast, systemic administration of the ET receptor antagonists had little or no effect on the on irET-1 in the CSF. However, intracerebroventricular (i.c.v.) administration of SB209670 produced a dose-related (3-100 micrograms) increase in cisternal CSF levels of irET-1 without altering plasma irET-1. Systemic administration of ETB receptor antagonists and agonists rapidly increased plasma irET-1. These ETB receptor antagonist effects correlate linearly with affinity at the cloned human ETB receptor, do not exhibit desensitization, and do not appear to reflect inhibition of ETB-mediated NO production. The endothelial ETB receptor may represent a high-capacity storage/clearance site for circulating ET-1. ET receptor antagonists may also act extravascularly/abluminally to increase irET-1 in the CNS.


Subject(s)
Endothelin Receptor Antagonists , Endothelin-1/metabolism , Animals , Benzofurans/pharmacokinetics , Benzofurans/pharmacology , Carboxylic Acids/pharmacokinetics , Carboxylic Acids/pharmacology , Dogs , Endothelin-1/blood , Endothelin-1/cerebrospinal fluid , Enzyme Inhibitors/pharmacology , Indans/pharmacokinetics , Indans/pharmacology , Injections, Intravenous , Injections, Intraventricular , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Propionates/pharmacokinetics , Propionates/pharmacology , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Endothelin/metabolism , Vasoconstrictor Agents/pharmacology , Viper Venoms/pharmacology
17.
J Cardiovasc Pharmacol ; 31 Suppl 1: S277-83, 1998.
Article in English | MEDLINE | ID: mdl-9595459

ABSTRACT

The effects of a nonpeptide, orally active mixed endothelin (ET) ETA/ETB receptor antagonist, SB 217242, and an angiotensin-converting enzyme (ACE) inhibitor, ramipril, were evaluated after inter-renal aortic banding in the rat. Separate sham, vehicle, and treatment groups were compared in each study. In vehicle-treated animals in the ramipril group, aortic banding for 4 weeks produced significant cardiac hypertrophy (247 +/- 5 mg/100 g bw vs. 305 +/- 11 mg/100 g bw; p < 0.001), right (upstream) renal hypertrophy (380 +/- 6 mg/100 g bw vs. 559 +/- 28 mg/100 g bw; p < 0.001), and significant left (downstream) renal atrophy (405 +/- 4 mg/100 g bw vs. 192 +/- 25 mg/100 g bw; p < 0.001). Continuous ramipril treatment (1 mg/kg p.o. once daily), begun 3 days before aortic banding, inhibited cardiac hypertrophy (305 +/- 11 mg/100 g bw vs. 266 +/- 7 mg/100 g bw; p < 0.05) but did not alter renal hypertrophy or atrophy. In a similarly designed study, SB 217242 (30 mg/kg p.o. b.i.d.) had no effect on the development of cardiac hypertrophy (298 +/- 7 mg/100 g bw vs. 310 +/- 12 mg/100 g bw) or renal hypertrophy (561 +/- 15 mg/100 g bw vs. 575 +/- 19 mg/100 g bw), but abolished the development of renal atrophy (158 +/- 16 mg/100 g bw vs. 395 +/- 19 mg/100 g bw; p < 0.001). [125I]ET-1 radioligand binding experiments indicated that the density of both ETA and ETB receptors was increased dramatically (three- to fourfold) in the atrophic kidney cortex compared to sham or hypertrophic kidneys. In situ hybridization studies indicate an upregulation of ETB receptor mRNA in the glomeruli of atrophic kidneys within 5 days of aortic banding. In conclusion, an angiotensin-dependent mechanism may mediate cardiac hypertrophy associated with aortic banding, whereas ET-dependent mechanisms may mediate an atrophic response in the hypoperfused kidney, perhaps through an interaction with upregulated ETA and/or ETB receptors.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cardiomegaly/pathology , Endothelin Receptor Antagonists , Kidney/pathology , Ramipril/pharmacology , Animals , Hypertrophy , In Situ Hybridization , Kidney/drug effects , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Male , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Endothelin/genetics
18.
Pharmacology ; 56(1): 30-6, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9467185

ABSTRACT

Many beta-adrenoceptor antagonists are weak partial agonists, possessing significant intrinsic sympathomimetic activity (ISA). Under certain conditions, ISA may be deleterious through stimulation of beta 1- and/or beta 2-adrenoceptors in the heart. Drugs with ISA are particularly problematic in the treatment of congestive heart failure since agents that activate cardiac beta-adrenoceptors, such as xamoterol, have been associated with increases in the incidence of arrhythmia and mortality. Carvedilol was recently approved for the treatment of congestive heart failure, and bucindolol is currently in large clinical trials for this indication. In the present study, the ISA of bucindolol and carvedilol was evaluated in a standard model used to investigate ISA, the pithed rat. Both compounds produced dose-dependent inhibition of the positive-chronotropic effects of the non-selective beta-adrenoceptor agonist, isoproterenol, confirming that these drugs are beta-adrenoceptor antagonists. However, cumulative administration of bucindolol (10-1,000 micrograms/kg i.v.) in the pithed rat produced a significant dose-related increase in heart rate. The maximal increase in heart rate produced by bucindolol was 44% of that obtained with isoproterenol (90 +/- 6vs. 205 +/- 11 bpm, respectively). In marked contrast, cumulative administration of carvedilol (10-1,000 micrograms/kg i.v.) had no significant effect on resting heart rate in the pithed rat. The maximal increase in heart rate elicited by bucindolol (1,000 micrograms/kg i.v.) was inhibited by treatment with the competitive beta-adrenoceptor antagonist, propranolol (99 +/- 8.7 vs. 26 +/- 2.6 bpm), confirming that the ISA observed with bucindolol was mediated through stimulation of myocardial beta-adrenoceptors. Carvedilol, which had no ISA, antagonized the ISA of bucindolol, and was as effective as propranolol in blocking the ISA of bucindolol (99 +/- 8.7 vs. 27 +/- 2.3 bpm). In summary, bucindolol and carvedilol are both potent beta-adrenoceptor antagonists in the pithed rat: however, only bucindolol possesses beta-adrenoceptor-mediated ISA.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Carbazoles/therapeutic use , Heart Failure/drug therapy , Propanolamines/therapeutic use , Sympathomimetics/therapeutic use , Adrenergic beta-Antagonists/adverse effects , Animals , Carbazoles/adverse effects , Carvedilol , Decerebrate State , Drug Evaluation, Preclinical , Evaluation Studies as Topic , Male , Propanolamines/adverse effects , Rats , Rats, Sprague-Dawley , Sympathomimetics/adverse effects
19.
Stroke ; 28(6): 1233-44, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9183357

ABSTRACT

BACKGROUND AND PURPOSE: Tumor necrosis factor-alpha (TNF-alpha) is a pleiotropic cytokine that rapidly upregulates in the brain after injury. The present study was designed to explore the pathophysiological significance of brain TNF-alpha in the ischemic brain by systematically evaluating the effects of lateral cerebroventricular administration of exogenous TNF-alpha and agents that block the effects of TNF-alpha on focal stroke and by examining the potential direct toxic effects of TNF-alpha on cultured neurons to better understand how TNF-alpha might mediate stroke injury. METHODS: TNF-alpha (2.5 or 25 pmol) was administered intracerebroventricularly to spontaneously hypertensive rats 24 hours before permanent or transient (80 minutes and 160 minutes) middle cerebral artery occlusion (MCAO). Animals were examined 24 hours later for neurological deficits and ischemic hemisphere necrosis and swelling. In some of these studies, neutralizing anti-TNF-alpha monoclonal antibody (mAb) (60 pmol) was injected intracerebroventricularly 30 minutes before exogenous TNF-alpha (25 pmol). In addition, the effects of blocking endogenous TNF-alpha on permanent focal ischemic injury were determined with the use of either mAb (60 pmol) or soluble TNF receptor I (sTNF-RI) (0.3 or 0.7 nmol) administered intracerebroventricularly 30 minutes before and 3 and 6 hours after MCAO. Finally, the direct neurotoxic effects of TNF-alpha were studied in cultured rat cerebellar granule cells exposed to TNF-alpha (10 to 2000 U/mL for 6 to 24 hours), and neurotransmitter release, glutamate toxicity, and oxygen radical toxicity were studied. RESULTS: TNF-alpha increased the percent hemispheric infarct induced by permanent MCAO in a dose-related manner from 13.1 +/- 1.3% (vehicle) to 18.9 +/- 1.7% at 2.5 pmol (P < .05) and 27.1 +/- 1.3% at 25 pmol (P < .0001). The high dose of TNF-alpha increased ischemia-induced forelimb deficits from 1.6 +/- 0.2 to 2.3 +/- 0.2 (P < 0.1). TNF-alpha (2.5 pmol) also increased the infarction induced by 80 or 160 minutes of transient MCAO from 1.9 +/- 0.9% to 4.3 +/- 0.4% (P < .01) and from 14.2 +/- 1.3% to 21.6 +/- 2.2% (P < .05), respectively. The exacerbation of infarct size, swelling, and neurological deficit after exogenous TNF-alpha was reversed by preinjection of 60 pmol mAb. Blocking endogenous TNF-alpha also significantly reduced focal ischemic brain injury. Treatment with 60 pmol mAb before and after permanent MCAO significantly reduced infarct size compared with control (nonimmune) antibody treatment by 20.2% (P < .05). Reduced brain infarction also was produced by brain administration of 0.3 nmol (decreased 18.2%) or 0.7 nmol (decreased 26.1%, P < .05) sTNF-RI before and after focal stroke. The intracerebroventricular administration of TNF-alpha or sTNF-RI did not alter brain or body temperature, blood gases or pH, blood pressure, blood glucose, or general blood chemistry. In cultured cerebellar granule cells, the application of TNF-alpha did not directly affect neurotransmitter release or glutamate or oxygen free radical toxicity. CONCLUSIONS: These studies demonstrate that exogenous TNF-alpha exacerbates focal ischemic injury and that blocking endogenous TNF-alpha is neuroprotective. The specificity of the action(s) of TNF-alpha was demonstrated by antagonism of its effects with specific anti-TNF-alpha tools (ie, mAb and sTNF-RI). TNF-alpha toxicity does not appear to be due to a direct effect on neurons or modulation of neuronal sensitivity to glutamate or oxygen radicals and apparently is mediated through nonneuronal cells. These data suggest that inhibiting TNF-alpha may represent a novel pharmacological strategy to treat ischemic stroke.


Subject(s)
Brain Ischemia/physiopathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies, Monoclonal , Cells, Cultured , Cerebellum/drug effects , Cerebral Infarction/physiopathology , Dose-Response Relationship, Drug , Male , Neurons/drug effects , Rats , Rats, Inbred SHR , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/physiology
20.
J Cereb Blood Flow Metab ; 17(2): 210-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9040501

ABSTRACT

The role of the phosphodiesterase type IV isozyme (PDE IV) in the regulation of cerebrovascular tone was investigated in the canine basilar artery in vitro and in vivo. The PDE isozymes extracted from the canine basilar artery were isolated by diethylaminoethanol (DEAE)-Sepharose affinity chromatography and identified based on sensitivity to isozyme-selective PDE inhibitors. [3H]cAMP hydrolysis was observed in one major and one minor peak of activity. The predominant peak was inhibited by the addition of cGMP (25%), siguazodan (26%), rolipram (39%), and the combination of siguazodan and rolipram (95%). Selective PDE IV inhibitors BRL 61063, rolipram, and denbufylline were equieffective inhibitors of [3H]-ccAMP hydrolysis mediated by PDE IV isolated from the canine basilar artery [concentrations producing 50% inhibition (IC50S) = 0.21 +/- 0.05 microM, 0.67 +/- 0.23 microM, and 0.73 +/- 0.16 microM, respectively]. In precontracted isolated ring segments of the canine basilar artery, selective PDE IV inhibitors produced potent and complete relaxation (IC50S < 150 nM). In contrast, zaprinast (a selective PDE V inhibitor) and siguazodan (a selective PDE III inhibitor) produced only weak relaxation of the basilar artery (IC50S = 4.5 microM and > 10 microM, respectively). Vasorelaxation produced by PDE IV inhibitors was not altered by removing the endothelium, 1-NAME, or adenosine receptor antagonism. In a canine model of acute cerebral vasospasm, all three selective PDE IV inhibitors reversed basilar artery spasm produced by autologous blood without altering mean arterial blood pressure. In contrast, prolonged treatment with BRL 61063 failed to alter the development of basilar spasm in the two hemorrhage canine models of chronic cerebral vasospasm. Denbufylline-induced relaxation in vitro was also significantly impaired in basilar arteries obtained from the model of chronic vasospasm. In conclusion, PDE IV appears to be the predominant isozyme regulating vascular tone mediated by cAMP hydrolysis in cerebral vessels. In addition, vasorelaxation modulated by PDE IV is compromised in chronic cerebral vasospasm associated with subarachnoid hemorrhage.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases , Cerebral Arteries/enzymology , Isoenzymes/physiology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Basilar Artery , Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dogs , Drug Evaluation, Preclinical , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/enzymology , Isoenzymes/isolation & purification , Male , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/isolation & purification , Prostaglandin Endoperoxides, Synthetic/pharmacology , Prostaglandin Endoperoxides, Synthetic/toxicity , Pyrrolidinones/pharmacology , Pyrrolidinones/therapeutic use , Rolipram , Second Messenger Systems , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/etiology , Subarachnoid Hemorrhage/prevention & control , Thromboxane A2/analogs & derivatives , Thromboxane A2/pharmacology , Thromboxane A2/toxicity , Vasoconstrictor Agents/pharmacology , Vasoconstrictor Agents/toxicity , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use , Xanthines/pharmacology , Xanthines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...