Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Pharmacol Rep ; 71(1): 139-148, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30550994

ABSTRACT

BACKGROUND: The G protein-coupled receptor 35 (GPR35), is considered important for nociceptive transmission, as suggested by accumulating evidence. This receptor was discovered in 1998; however, a lack of pharmacological tools prevented a complete understanding of its function and how to exploit it therapeutically. We studied the influence of CXCL17, kynurenic acid and zaprinast on nociceptive transmission in naïve and neuropathic mice. Additionally, we investigated the influence of kynurenic acid and zaprinast on morphine effectiveness in neuropathic pain. METHODS: The chronic constriction injury (CCI) of the sciatic nerve in Swiss mice was performed. The CXCL17, kynurenic acid, zaprinast and morphine were injected intrathecally into naive and CCI-exposed mice at day 14. To evaluate tactile and thermal hypersensitivity, the von Frey and cold plate tests were used, respectively. RESULTS: Our results have shown, for the first time, that administration of CXCL17 in naïve mice induced strong pain-related behaviours, as measured by von Frey and cold plate tests. Moreover, we demonstrated that kynurenic acid and zaprinast diminished CXCL17-evoked pain-related behaviours in both tests. Kynurenic acid and zaprinast reduced thermal and tactile hypersensitivity developed by sciatic nerve injury and strongly enhanced the effectiveness of morphine in neuropathy. CONCLUSIONS: Our study highlights the importance of GPR35 as a receptor involved in neuropathic pain development. Therefore, these results suggest that the modulation of GPR35 could become a potential strategy for the treatment of neuropathic pain.


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics/pharmacology , Behavior, Animal/drug effects , Chemokines, CXC/toxicity , Kynurenic Acid/pharmacology , Morphine/pharmacology , Pain Perception/drug effects , Pain Threshold/drug effects , Purinones/pharmacology , Sciatica/drug therapy , Spinal Cord/drug effects , Analgesics/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Chemokines, CXC/administration & dosage , Disease Models, Animal , Injections, Spinal , Kynurenic Acid/administration & dosage , Male , Mice , Morphine/administration & dosage , Purinones/administration & dosage , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Sciatica/chemically induced , Sciatica/physiopathology , Sciatica/psychology , Spinal Cord/metabolism , Spinal Cord/physiopathology
2.
Naunyn Schmiedebergs Arch Pharmacol ; 390(9): 939-948, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28660304

ABSTRACT

Renal fibrosis is an important factor for end-stage renal failure. However, only few therapeutic options for its treatment are established. Zaprinast, a phosphodiesterase 5 inhibitor, and serelaxin, the recombinant form of the naturally occurring hormone relaxin, are differently acting modulators of cyclic guanosine monophosphate (cGMP) signaling. Both agents enhance cGMP availability in kidney tissue. These substances alone or in combination might interfere with the development of kidney fibrosis. Therefore, we compared the effects of combination therapy with the effects of monotherapy on renal fibrosis. Renal fibrosis was induced by unilateral ureteral obstruction (UUO) for 7 days in wild-type (WT) and cGKI knockout (KO) mice. Renal antifibrotic effects were assessed after 7 days. In WT, zaprinast and the combination of zaprinast and serelaxin significantly reduced renal interstitial fibrosis assessed by α-SMA, fibronectin, collagen1A1, and gelatinases (MMP2 and MMP9). Intriguingly in cGKI-KO, mRNA and protein expression of fibronectin and collagen1A1 were reduced by zaprinast, in contrast to serelaxin. Gelatinases are not regulated by zaprinast. Although both substances showed similar antifibrotic properties in WT, they distinguished in their effect mechanisms. In contrast to serelaxin which acts both on Smad2 and Erk1, zaprinast did not significantly diminish Erk1/2 phosphorylation. Interestingly, the combination of serelaxin/zaprinast achieved no additive antifibrotic effects compared to the monotherapy. Due to antifibrotic effects of zaprinast in cGKI-KO, we hypothesize that additional cGKI-independent mechanisms are supposed for antifibrotic signaling of zaprinast.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic GMP/metabolism , Kidney Diseases/prevention & control , Purinones/pharmacology , Relaxin/pharmacology , Animals , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Drug Therapy, Combination , Fibrosis , Kidney/drug effects , Kidney/pathology , Kidney Diseases/pathology , Mice , Mice, Knockout , Phosphodiesterase 5 Inhibitors/administration & dosage , Phosphodiesterase 5 Inhibitors/pharmacology , Purinones/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Relaxin/administration & dosage , Signal Transduction/drug effects , Ureteral Obstruction/complications
3.
Med Sci Monit Basic Res ; 20: 105-11, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25057848

ABSTRACT

BACKGROUND: Phosphodiesterase (PDE) inhibitors in the central nervous system have been shown to stimulate neuronal functions and increase neurogenesis in Alzheimer disease (AD) patients. MATERIAL/METHODS: The aim of this study was to investigate the effects of zaprinast, a PDE5 inhibitor, and rolipram, a PDE4 inhibitor, on learning and memory in elevated plus maze (EPM) and passive avoidance (PA) tests in naive mice. Male Balb-c mice received short-term treatment with zaprinast (3 and 10 mg/kg) and rolipram (0.05 and 0.1 mg/kg) before the acquisition trial of the EPM and PA tests. The exploratory activity of the animals was also investigated in the Hughes box test. RESULTS: Both zaprinast (10 mg/kg) and rolipram (0.1 mg/kg) significantly decreased second-day latency compared to the control group in the EPM test, while only rolipram (0.1 mg/kg) significantly increased second-day latency in the PA test. Both zaprinast (10 mg/kg) and rolipram (0.1 mg/kg) significantly decreased the number of entries to new areas and time spent in new areas in the Hughes box test. CONCLUSIONS: Our study revealed that both zaprinast and rolipram enhanced spatial memory in EPM, while rolipram seemed to have more emotional memory-enhancing effects in the PA test compared to zaprinast. Both zaprinast and rolipram diminished exploratory activity in the Hughes box test, which can be attributed to the drugs' anxiogenic effects.


Subject(s)
Emotions/drug effects , Exploratory Behavior/drug effects , Maze Learning/drug effects , Purinones/pharmacology , Rolipram/pharmacology , Spatial Memory/drug effects , Animals , Avoidance Learning/drug effects , Male , Mice, Inbred BALB C , Purinones/administration & dosage , Rolipram/administration & dosage
4.
Biomed Res Int ; 2013: 397391, 2013.
Article in English | MEDLINE | ID: mdl-23956981

ABSTRACT

The edema formation in nephrotic syndrome (NS) is associated with a blunted response to atrial natriuretic peptide (ANP). The natriuretic effects of ANP have been related to renal dopamine D1-receptors (D1R). We examined the interaction between ANP and renal D1R in rats with puromycin aminonucleoside-induced NS (PAN-NS). Urinary sodium, cyclic guanosine monophosphate (cGMP) excretion, and D1R protein expression and localization in renal tubules were evaluated in PAN-NS and control rats before and during volume expansion (VE). The effects of zaprinast (phosphodiesterase type 5 inhibitor), alone or in combination with Sch-23390 (D1R antagonist), were examined in both groups. The increased natriuresis and urinary cGMP excretion evoked by acute VE were blunted in PAN-NS despite increased levels of circulating ANP. This was accompanied in PAN-NS by a marked decrease of D1R expression in the renal tubules. Infusion of zaprinast in PAN-NS resulted in increased urinary excretion of cGMP and sodium to similar levels of control rats and increased expression of D1R in the plasma membrane of renal tubular cells. Combined administration of Sch-23390 and zaprinast prevented natriuresis and increased cGMP excretion induced by zaprinast alone. We conclude that D1R may play a major role in the ANP resistance observed in PAN-NS.


Subject(s)
Atrial Natriuretic Factor/metabolism , Natriuresis/drug effects , Receptors, Dopamine D1/biosynthesis , Sodium/metabolism , Animals , Benzazepines/administration & dosage , Cyclic GMP/urine , Glomerular Filtration Rate , Homeostasis , Kidney/metabolism , Kidney/pathology , Male , Nephrotic Syndrome/chemically induced , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Purinones/administration & dosage , Puromycin Aminonucleoside/toxicity , Rats , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/metabolism
5.
Exp Dermatol ; 20(7): 568-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21410772

ABSTRACT

We previously demonstrated that exposure to red light (550-670 nm) accelerates epidermal permeability barrier recovery after barrier disruption. Furthermore, we showed that photosensitive proteins, originally found in retina, are also expressed in epidermis. In retina, transducin and phosphodiesterase 6 play key roles in signal transmission. In this study, we evaluate the role of phosphodiesterese 6 in the acceleration by red light of epidermal permeability barrier recovery. Immunohistochemical study and reverse transcription-PCR assays confirmed the expression of both transducin and phosphodiesterase 6 in epidermal keratinocytes. Topical application of 3-isobutyl-1-methylxanthine, a non-specific phosphodiesterase inhibitor, blocked the acceleration of the barrier recovery by red light. Topical application of zaprinast, a specific inhibitor of phosphodiesterases 5 and 6, also blocked the acceleration, whereas T0156, a specific inhibitor of phosphodiesterase 5, had no effect. Red light exposure reduced the epidermal hyperplasia induced by barrier disruption under low humidity, and the effect was blocked by pretreatment with zaprinast. Our results indicate phosphodiesterase 6 is involved in the recovery-accelerating effect of red light on the disrupted epidermal permeability barrier.


Subject(s)
Phosphodiesterase Inhibitors/pharmacology , Phototherapy , Skin/metabolism , Wound Healing/drug effects , Wound Healing/radiation effects , 1-Methyl-3-isobutylxanthine/administration & dosage , 1-Methyl-3-isobutylxanthine/pharmacology , Acetone/pharmacology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Epidermis/radiation effects , Epidermis/ultrastructure , Gene Expression/genetics , Hyperplasia/chemically induced , Hyperplasia/prevention & control , Hyperplasia/radiotherapy , Male , Mice , Mice, Hairless , Naphthyridines/administration & dosage , Naphthyridines/pharmacology , Permeability , Phosphodiesterase Inhibitors/administration & dosage , Purinones/administration & dosage , Purinones/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Skin/drug effects , Skin/pathology , Skin/radiation effects , Skin/ultrastructure , Transducin/genetics , Transducin/metabolism , Water/metabolism
6.
Psychopharmacology (Berl) ; 201(3): 361-71, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18758756

ABSTRACT

RATIONALE: Ethanol and caffeine are two of the most widely consumed drugs in the world, often used in the same setting. Animal models may help to understand the conditions under which incidental memories formed just before ethanol intoxication might be lost or become difficult to retrieve. OBJECTIVES: Ethanol-induced retrograde amnesia was investigated using a new odor-recognition test. MATERIALS AND METHODS: Rats thoroughly explored a wood bead taken from the cage of another rat, and habituated to this novel odor (N1) over three trials. Immediately following habituation, rats received saline, 25 mg/kg pentylenetetrazol (a seizure-producing agent known to cause retrograde amnesia) to validate the test, 1.0 g/kg ethanol, or 3.0 g/kg ethanol. The next day, they were presented again with N1 and also a bead from a new rat's cage (N2). RESULTS: Rats receiving saline or the lower dose of ethanol showed overnight memory for N1, indicated by preferential exploration of N2 over N1. Rats receiving pentylenetetrazol or the higher dose of ethanol appeared not to remember N1, in that they showed equal exploration of N1 and N2. Caffeine (5 mg/kg), delivered either 1 h after the higher dose of ethanol or 20 min prior to habituation to N1, negated ethanol-induced impairment of memory for N1. A combination of a phosphodiesterase-5 inhibitor and an adenosine A(2A) antagonist, mimicking two major mechanisms of action of caffeine, likewise prevented the memory impairment, though either drug alone had no such effect. Binge alcohol can induce retrograde, caffeine-reversible disruption of social odor memory storage or recall.


Subject(s)
Amnesia, Retrograde/chemically induced , Amnesia, Retrograde/prevention & control , Caffeine/therapeutic use , Central Nervous System Depressants/toxicity , Central Nervous System Stimulants/therapeutic use , Ethanol/toxicity , Adenosine A2 Receptor Antagonists , Animals , Caffeine/administration & dosage , Caffeine/pharmacology , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Exploratory Behavior/drug effects , Habituation, Psychophysiologic , Injections, Intraperitoneal , Male , Odorants , Pentylenetetrazole/toxicity , Phosphodiesterase 5 Inhibitors , Purinones/administration & dosage , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Recognition, Psychology , Seizures/chemically induced , Smell , Time Factors , Triazines/administration & dosage , Triazoles/administration & dosage , Wood
7.
Pharmacology ; 82(4): 245-9, 2008.
Article in English | MEDLINE | ID: mdl-18818509

ABSTRACT

GPR35 is a Gi/o- and G16-coupled receptor abundantly expressed in gastrointestinal tissues and immune cells. Kynurenic acid (a tryptophan metabolite and ionotropic glutamate receptor antagonist) and zaprinast (a phosphodiesterase inhibitor) are GPR35 agonists. Here, we show that the chloride channel blocker 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB) is also a GPR35 agonist. NPPB activates the GPR35-Gi/o and GPR35-G16 pathways in human embryonic kidney 293 (HEK293) cells and induces intracellular calcium mobilization in a concentration-dependent manner in HEK293 cells coexpressing human, rat or mouse GPR35 and the chimeric G protein G(qi5). These results suggest a novel pharmacological activity of NPPB and will provide useful information to search for more potent and selective GPR35 agonists.


Subject(s)
Nitrobenzoates/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line , Chloride Channels/antagonists & inhibitors , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Mice , Nitrobenzoates/administration & dosage , Purinones/administration & dosage , Purinones/pharmacology , Rats
8.
Am J Respir Crit Care Med ; 176(11): 1138-45, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17872487

ABSTRACT

RATIONALE: Nitric oxide-independent agonists of soluble guanylate cyclase (sGC) have been developed. OBJECTIVES: We tested whether inhalation of novel dry-powder microparticle formulations containing sGC stimulators (BAY 41-2272, BAY 41-8543) or an sGC activator (BAY 58-2667) would produce selective pulmonary vasodilation in lambs with acute pulmonary hypertension. We also evaluated the combined administration of BAY 41-8543 microparticles and inhaled nitric oxide (iNO). Finally, we examined whether inhaling BAY 58-2667 microparticles would produce pulmonary vasodilation when the response to iNO is impaired. METHODS: In awake, spontaneously breathing lambs instrumented with vascular catheters and a tracheostomy tube, U-46619 was infused intravenously to increase mean pulmonary arterial pressure to 35 mm Hg. MEASUREMENTS AND MAIN RESULTS: Inhalation of microparticles composed of either BAY 41-2272, BAY 41-8543, or BAY 58-2667 and excipients (dipalmitoylphosphatidylcholine, albumin, lactose) produced dose-dependent pulmonary vasodilation and increased transpulmonary cGMP release without significant effect on mean arterial pressure. Inhalation of microparticles containing BAY 41-8543 or BAY 58-2667 increased systemic arterial oxygenation. The magnitude and duration of pulmonary vasodilation induced by iNO were augmented after inhaling BAY 41-8543 microparticles. Intravenous administration of 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), which oxidizes the prosthetic heme group of sGC, markedly reduced the pulmonary vasodilator effect of iNO. In contrast, pulmonary vasodilation and transpulmonary cGMP release induced by inhaling BAY 58-2667 microparticles were greatly enhanced after treatment with ODQ. CONCLUSIONS: Inhalation of microparticles containing agonists of sGC may provide an effective novel treatment for patients with pulmonary hypertension, particularly when responsiveness to iNO is impaired by oxidation of sGC.


Subject(s)
Benzoates/administration & dosage , Morpholines/administration & dosage , Pulmonary Circulation/drug effects , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Receptors, Cytoplasmic and Nuclear/agonists , Vasodilation , Administration, Inhalation , Aerosols , Animals , Benzoates/pharmacology , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Guanylate Cyclase , Injections, Intravenous , Lung/drug effects , Lung/metabolism , Morpholines/pharmacology , Nitric Oxide/administration & dosage , Nitric Oxide/pharmacology , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacology , Particle Size , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/pharmacology , Powders , Purinones/administration & dosage , Purinones/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinoxalines/administration & dosage , Quinoxalines/pharmacology , Sheep , Soluble Guanylyl Cyclase , Vasodilation/drug effects
9.
Behav Brain Res ; 169(1): 181-5, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16472878

ABSTRACT

A weakly reinforced variant of the single trial passive avoidance task developed for the day-old chick typically fails to consolidate long-term memory. However, administration of zaprinast, a phosphodiesterase (PDE) type 5 inhibitor, (ic; 10 microl/side) immediately post-training resulted in a dose-dependent increase in retention at test 180 min post-training. Further, 100 microM zaprinast resulted in high levels of retention at test 180 min post-training when administered from 10 min before training to 10 min after training. Finally, 100 microM zaprinast, when administered immediately post-training, resulted in the consolidation of long-term memory at a number of times of test extending as late as 24 h post-training. Inhibition of PDE type 5 is known to increase cellular cGMP levels. Previous investigations using a strongly reinforced variant of this task have suggested a role for cGMP in memory retrieval, we now postulate that cGMP is also necessary for memory formation in chicks trained using passive avoidance.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Avoidance Learning/drug effects , Nootropic Agents/administration & dosage , Purinones/administration & dosage , Retention, Psychology/drug effects , Analysis of Variance , Animals , Chickens , Dose-Response Relationship, Drug , Drug Administration Schedule , Reinforcement, Psychology
10.
Eur J Anaesthesiol ; 23(1): 65-70, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16390569

ABSTRACT

BACKGROUND AND OBJECTIVE: The cyclic guanosine monophosphate level, which causes an antinociception, is increased in cells as a direct result of phosphodiesterase inhibition. This study used a nociceptive test to examine the nature of the pharmacological interaction between intrathecal zaprinast, a phosphodiesterase inhibitor, and morphine. METHODS: Catheters were inserted into the intrathecal space through an incision in the atlantooccipital membrane of male Sprague-Dawley rats. As a nociceptive model, 50 microL of a 5% formalin solution was injected into the hind paw. After observing the effect of zaprinast (37, 111, 369 nmol) and morphine (1, 4, 10, 40 nmol) alone, the interactions of their combination were examined by an isobolographic analysis. RESULTS: Intrathecal zaprinast (P < 0.05) and morphine (P < 0.05) dose-dependently suppressed the flinching observed during phase 1 and phase 2 in the formalin test. The ED50 values (95% confidence intervals) of zaprinast and morphine in phase 1 were 161.9 (87.9-298.3) and 11.6 nmol (4.8-27.9 nmol), respectively. The phase 2 ED50 values (95% confidence intervals) of zaprinast and morphine were 229.9 (142.5-370.9) and 3.9 nmol (1.9-7.6 nmol), respectively. Isobolographic analysis revealed a synergistic interaction after intrathecal delivery a zaprinast-morphine mixture in both phases. The ED50 values of (95% confidence intervals) zaprinast in the combination of zaprinast with morphine in phase 1 and phase 2 were 14.2 (4.9-40.6) and 10.4 nmol (3-35.9 nmol), respectively. CONCLUSIONS: Intrathecal zaprinast and morphine are effective against acute pain and facilitated pain state. Zaprinast interacts synergistically with morphine.


Subject(s)
Analgesics, Opioid/pharmacology , Formaldehyde , Morphine/pharmacology , Pain Measurement/drug effects , Pain/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Purinones/pharmacology , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Synergism , Injections, Spinal , Male , Morphine/administration & dosage , Muscle Tonus/drug effects , Phosphodiesterase Inhibitors/administration & dosage , Purinones/administration & dosage , Rats , Rats, Sprague-Dawley , Reflex/drug effects
11.
Eur J Pharmacol ; 528(1-3): 176-82, 2005 Dec 28.
Article in English | MEDLINE | ID: mdl-16316644

ABSTRACT

The insulinotropic activity of KCP256 [(R)-8-benzyl-2-cyclopentyl-7, 8-dihydro-4-propyl-1H-imidazo[2,1-i]purin-5(4H)-one hydrochloride] was examined using MIN6 cells (a pancreatic beta-cell line) and pancreatic islets isolated from rats. Unlike sulfonylurea anti-diabetic drugs, KCP256 dose-dependently (0.1-10 microM) enhanced insulin secretion from MIN6 cells and its insulinotropic effect was exerted only at high concentrations of glucose (8.3-22 mM) but not at low concentrations of glucose (3.3-5.5 mM). Furthermore, the action mechanism of KCP256 was different because, unlike sulfonylurea drugs, KCP256 did not displace the binding of [3H]glibenclamide, and did not inhibit the 86Rb+ efflux nor K(ATP) channel activity. In isolated islets, KCP256 also enhanced insulin secretion in a dose- and a glucose-concentration-dependent manner. Plasma levels of insulin after glucose challenge in KCP256-administrated rats were higher than those in vehicle-administrated animals, indicating that KCP256 can enhance insulin secretion in vivo. Since the insulinotropic activity of KCP256 only occurs at high concentrations of glucose, this novel drug may exhibit a decreased risk of drug-induced hypoglycemia compared with sulfonylurea drugs when treating patients with diabetes.


Subject(s)
Glucose/pharmacology , Hypoglycemic Agents/pharmacology , Imidazoles/pharmacology , Insulin/metabolism , Purines/pharmacology , Purinones/pharmacology , Animals , Blood Glucose/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Glucose Tolerance Test , Hypoglycemic Agents/administration & dosage , In Vitro Techniques , Insulin/blood , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Purinones/administration & dosage , Rats , Rats, Wistar
12.
Neurol Res ; 27(6): 638-43, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16157016

ABSTRACT

BACKGROUND: Guanosine 3', 5'-cyclic monophosphate (cGMP) acts as a relaxant second messenger in the cerebral vessels. cGMP-specific phosphodiesterase type 5 (PDE5) inhibitor increases intracellular cGMP levels. This study investigated the effect of the PDE5 inhibitor on the ischemic brain. METHODS: Regional cerebral blood flow (rCBF), cGMP concentration, and infarction volume were measured in the rat middle cerebral artery occlusion model. Ten minutes after ischemia, the animals received an intravenous (i.v.) infusion of vehicle (phosphate-buffered saline), PDE5 inhibitor, zaprinast (10 mg/kg), or nitric oxide donor, S-nitroso-N-acetyl-penicillamine (SNAP, 100 microg/kg). rCBF was measured continuously by laser-Doppler flowmetry in the ischemic penumbra of the ischemic and contralateral sides under continuous blood pressure monitoring. cGMP concentrations were determined using the enzyme immunoassay and infarct volumes were estimated by 2,3,5-triphenyltetrazolium chloride staining. RESULTS: The administration of zaprinast significantly increased rCBF in the ischemic brain compared with the pre-drug control value despite the decreased mean blood pressure, whereas it did not affect rCBF in the contralateral side. The cGMP concentration was significantly higher in the ischemic cortex compared with the contralateral side. SNAP infusion increased the cGMP concentration in the bilateral cortices to a similar extent. The volume of cerebral infarction was significantly decreased by zaprinast administration. CONCLUSIONS: The PDE5 inhibitor zaprinast may selectively increase CBF in the ischemic brain via increased cGMP levels, thus providing a new strategy against acute cerebral infarction.


Subject(s)
Brain Ischemia/prevention & control , Cerebrovascular Circulation/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Phosphodiesterase Inhibitors/administration & dosage , Purinones/administration & dosage , Regional Blood Flow/drug effects , Analysis of Variance , Animals , Blood Circulation Time/drug effects , Blood Pressure/drug effects , Cyclic GMP/metabolism , Disease Models, Animal , Functional Laterality , Immunoenzyme Techniques/methods , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Laser-Doppler Flowmetry/methods , Male , Nitric Oxide Donors/administration & dosage , Penicillamine/administration & dosage , Penicillamine/analogs & derivatives , Rats , Rats, Wistar , Tetrazolium Salts , Time Factors
13.
Neurosci Lett ; 390(2): 114-7, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16115733

ABSTRACT

Zaprinast is a phosphodiesterase inhibitor that is active in various models of pain when administered locally. In addition, the antinociception of zaprinast is involved in the nitric oxide (NO)-cGMP pathway. However, the effect of zaprinast administered spinally has not been examined. Therefore, this study examined the effect of zaprinast on the formalin-induced nociception at the spinal level. Next, the role of the NO-cGMP-potassium channel pathway on the effect of zaprinast was further clarified. Catheters were inserted into the intrathecal space of male Sprague-Dawley (SD) rats. Pain was induced by applying 50 microl of a 5% formalin solution to the hindpaw. The change in the zaprinast-induced effect was examined after an intrathecal pretreatment with a NO synthase inhibitor (l-NMMA), a guanylyl cyclase inhibitor (ODQ) or a potassium channel blocker (glibenclamide). Zaprinast produced an antinociceptive effect during phase 1 and phase 2 in the formalin test. Intrathecal l-NMMA, ODQ and glibenclamide did not reverse the antinociception of zaprinast in either phase of the formalin test. These results suggest that zaprinast is effective against both acute pain and the facilitated pain state at the spinal level. However, the NO-sensitive cGMP-potassium channel pathway is not contributable to the antinociceptive mechanism of zaprinast in the spinal cord.


Subject(s)
Analgesics/pharmacology , Cyclic GMP/metabolism , Nitric Oxide/metabolism , Pain Measurement/drug effects , Potassium Channels/metabolism , Purinones/pharmacology , Signal Transduction/physiology , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Analgesics/administration & dosage , Analgesics/metabolism , Animals , Formaldehyde/adverse effects , Glyburide/metabolism , Injections, Spinal , Male , Oxadiazoles/metabolism , Pain/chemically induced , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/pharmacology , Purinones/administration & dosage , Purinones/metabolism , Quinoxalines/metabolism , Rats , Rats, Sprague-Dawley , omega-N-Methylarginine/metabolism
14.
Am J Physiol Lung Cell Mol Physiol ; 280(5): L923-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11290516

ABSTRACT

We studied the role of cGMP in nitric oxide (NO)-induced changes in lung liquid production (J(v)) in chronically instrumented fetal sheep. Forty-five studies were done in which J(v) was measured by a tracer dilution technique. Left pulmonary arterial flow (Q(lpa)) was measured by a Doppler flow probe. There were two series of experiments. In the first, we gave 8-bromo-cGMP, a cGMP analog, by either the pulmonary vascular or intraluminal route; in the second, we used agents to inhibit or enhance endogenous cGMP activity. When infused directly into the pulmonary circulation, 8-bromo-cGMP significantly increased Q(lpa) but had no effect on J(v). Conversely, when instilled into the lung liquid, 8-bromo-cGMP had no effect on Q(lpa) but significantly reduced J(v). Inhibition of guanylate cyclase activity with methylene blue totally blocked, whereas phosphodiesterase inhibition with Zaprinast significantly enhanced, the effect of instilled NO on J(v). Thus the reduction in lung liquid caused by NO appears to be mediated by cGMP, perhaps through a direct effect on the pulmonary epithelium.


Subject(s)
Cyclic GMP/metabolism , Lung/metabolism , Nitric Oxide/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , 8-Bromo Cyclic Adenosine Monophosphate/administration & dosage , Animals , Blood Gas Analysis , Blood Pressure/drug effects , Drug Administration Routes , Enzyme Inhibitors/administration & dosage , Fetus , Guanylate Cyclase/antagonists & inhibitors , Lung/blood supply , Lung/drug effects , Lung/embryology , Methylene Blue/administration & dosage , Nitric Oxide/administration & dosage , Pulmonary Circulation/drug effects , Purinones/administration & dosage , Sheep , Vascular Resistance/drug effects
15.
Crit Care Med ; 28(7): 2420-4, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10921573

ABSTRACT

OBJECTIVE: To determine whether the combination of the phosphodiesterase 5 inhibitor zaprinast and inhaled nitric oxide (NO) decreases hypoxic pulmonary hypertension in the rat. DESIGN: Prospective, experimental study. SETTING: Animal laboratory of a university medical center. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Anesthetized rats were mechanically ventilated and instrumented for measurement of mean systemic arterial pressure, pulmonary arterial pressure, and cardiac output. In group 1, four acute hypoxic challenges (FIO2 = 0.17 for 5 mins) were performed: initial, during 40 ppm inhaled NO, immediately after discontinuation of 5 mins of inhaled NO, and final. In group 2 rats, an initial hypoxic challenge was performed and rats then received zaprinast (3 mg/kg bolus followed by 0.3 mg/kg/min infusion). Four hypoxic challenges analogous to group 1 were then performed during zaprinast administration. MEASUREMENTS AND MAIN RESULTS: Initial hypoxic challenge produced similar increases in pulmonary arterial pressure in both groups. In group 1, inhaled NO either only before or only during hypoxia decreased the pulmonary hypertensive response to hypoxia. In group 2, zaprinast administration did not alter hemodynamics. Zaprinast alone decreased the pulmonary hypertensive response to hypoxia. The combination of zaprinast and inhaled NO (either before or during hypoxia) abolished the pulmonary hypertensive response to hypoxia. CONCLUSIONS: Treatment with inhaled NO for 5 mins before but not during hypoxia is as effective as inhaled NO during hypoxia. Inhaled NO and zaprinast both decrease the pulmonary hypertensive response to hypoxia, and the combination abolishes the response. The combination of a phosphodiesterase 5 inhibitor and inhaled NO may have clinical applicability in the treatment of pulmonary hypertension.


Subject(s)
Bronchodilator Agents/therapeutic use , Hemodynamics/drug effects , Hypertension, Pulmonary/drug therapy , Nitric Oxide/therapeutic use , Phosphodiesterase Inhibitors/therapeutic use , Purinones/therapeutic use , Administration, Inhalation , Analysis of Variance , Animals , Bronchodilator Agents/administration & dosage , Drug Interactions , Drug Therapy, Combination , Hypoxia/drug therapy , Infusions, Intravenous , Male , Nitric Oxide/administration & dosage , Phosphodiesterase Inhibitors/administration & dosage , Purinones/administration & dosage , Rats , Rats, Sprague-Dawley
16.
Anesthesiology ; 93(2): 422-30, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10910492

ABSTRACT

BACKGROUND: Inhaled nitric oxide (No) selectively dilates the pulmonary vasculature and improves gas exchange in acute respiratory distress syndrome. Because of the very short half-life of NO, inhaled NO is administered continuously. Intravenous Zaprinast (2-o-propoxyphenyl-8-azapurin-6-one), a cyclic guanosine monophosphate phosphodiesterase inhibitor, increases the efficacy and prolongs the duration of action of inhaled NO in models of acute pulmonary hypertension. Its efficacy in lung injury models is uncertain. The authors hypothesized that the use of intravenous Zaprinast would have similar beneficial effects when used in combination with inhaled NO to improve oxygenation and dilate the pulmonary vasculature in a diffuse model of acute lung injury. METHODS: The authors studied two groups of sheep with lung injury produced by saline lavage. In the first group, 0, 5, 10, and 20 ppm of inhaled NO were administered in a random order before and after an intravenous Zaprinast infusion (2 mg/kg bolus followed by 0.1 mg. kg-1. min-1). In the second group, inhaled NO was administered at the same concentrations before and after an intravenous infusion of Zaprinast solvent (0.05 m NaOH). RESULTS: After lavage, inhaled NO decreased pulmonary arterial pressure and resistance with no systemic hemodynamic effects, increased arterial oxygen partial pressure, and decreased venous admixture (all P < 0.05). The intravenous administration of Zaprinast alone decreased pulmonary artery pressure but worsened gas exchange (P < 0.05). Zaprinast infusion abolished the beneficial ability of inhaled NO to improve pulmonary gas exchange and reduce pulmonary artery pressure (P < 0. 05 vs. control). CONCLUSIONS: This study suggests that nonselective vasodilation induced by intravenously administered Zaprinast at the dose used in our study not only worsens gas exchange, but also abolishes the beneficial effects of inhaled NO.


Subject(s)
Bronchodilator Agents/therapeutic use , Hemodynamics/drug effects , Nitric Oxide/therapeutic use , Phosphodiesterase Inhibitors/therapeutic use , Pulmonary Gas Exchange/drug effects , Purinones/therapeutic use , Respiratory Distress Syndrome/drug therapy , Administration, Inhalation , Analysis of Variance , Animals , Bronchodilator Agents/administration & dosage , Dose-Response Relationship, Drug , Drug Synergism , Infusions, Intravenous , Models, Biological , Nitric Oxide/administration & dosage , Phosphodiesterase Inhibitors/administration & dosage , Purinones/administration & dosage , Sheep
17.
Am J Respir Crit Care Med ; 160(5 Pt 1): 1500-6, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10556112

ABSTRACT

Inhalation of aerosolized prostaglandin I(2) (PGI(2)) causes selective pulmonary vasodilation, but the effect rapidly levels off after termination of nebulization. In experimental pulmonary hypertension in intact rabbits, provoked by continuous infusion of the stable thromboxane mimetic U46619, the impact of intravenous phosphodiesterase (PDE) inhibitors on pulmonary and systemic hemodynamics was investigated in the absence and the presence of aerosolized PGI(2). We employed the monoselective inhibitors motapizone (PDE 3), rolipram (PDE 4), and zaprinast (PDE 5), as well as the dual-selective blockers zardaverine and tolafentrine (both PDE 3/4). All PDE inhibitors dose-dependently reduced the pulmonary artery pressure (Ppa), with doses for an approximately 20% decrease in pulmonary vascular resistance being 5 microgram/kg for motapizone, 25 microgram/kg for rolipram, 500 microgram/kg for zardaverine, 1 mg/kg for zaprinast, and 1 mg/kg for tolafentrine. Additive efficacy was noted when combining the monoselective 3 plus 4, 3 plus 5, and 4 plus 5 inhibitors. In parallel with the pulmonary vasorelaxant effect, all PDE inhibitors caused a decrease in systemic arterial pressure and an increase in cardiac output. Nebulized PGI(2) (56 ng/kg. min) reduced the U46619-evoked increase in Ppa by approximately 30%. This vasorelaxant effect was fully lost within 10 min after termination of PGI(2) nebulization. Coapplication of subthreshold doses of intravenous PDE inhibitors, which per se did not affect pulmonary and systemic hemodynamics, resulted in a marked prolongation of the post-PGI(2) decrease in Ppa for all blockers (motapizone at 2.2 microgram/kg, rolipram at 5.5 microgram/kg, zaprinast at 100 microgram/kg). The most effective agents, zardaverine (50 microgram/kg) and tolafentrine (100 microgram/kg), augmented the maximum Ppa drop during nebulization by approximately 30-50% and prolonged the post-PGI(2) pulmonary vasodilation to > 30 min, without affecting systemic arterial pressure and arterial oxygenation. We conclude that subthreshold systemic doses of monoselective PDE 3, 4, and 5 inhibitors and in particular dual-selective PDE 3/4 inhibitors cause significant amplification of the pulmonary vasodilatory response to inhaled PGI(2), while limiting the hypotensive effect to the pulmonary circulation. Combining nebulized PGI(2) with low-dose systemic PDE inhibitors may thus offer a therapeutic strategy to achieve selective pulmonary vasodilation in acute and chronic pulmonary hypertension.


Subject(s)
Antihypertensive Agents/administration & dosage , Epoprostenol/administration & dosage , Hypertension, Pulmonary/physiopathology , Phosphodiesterase Inhibitors/administration & dosage , Pulmonary Circulation/drug effects , Vasodilation/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Administration, Inhalation , Aerosols , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Cardiac Output/drug effects , Dose-Response Relationship, Drug , Drug Therapy, Combination , Epoprostenol/pharmacology , Hypertension, Pulmonary/chemically induced , Naphthyridines/administration & dosage , Naphthyridines/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Purinones/administration & dosage , Purinones/pharmacology , Pyridazines/administration & dosage , Pyridazines/pharmacology , Rabbits , Rolipram/administration & dosage , Rolipram/pharmacology , Vasoconstrictor Agents
18.
Cancer Res ; 58(5): 914-20, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9500450

ABSTRACT

Intracarotid infusion of bradykinin selectively increases the delivery of compounds into brain tumors. This study sought to determine the role of cyclic GMP in increased permeability across the blood-tumor barrier (BTB) after infusion of bradykinin. In permeability studies, 186 Wistar rats with RG2 gliomas and C6 gliomas were used. Transport across the BTB was quantified by autoradiography and reported as a unidirectional transport, Ki, for [14C]dextran (Mr 70,000) and [14C]aminoisobutyric acid (Mr 103,000), with or without inhibition of cyclic GMP-specific phosphodiesterase or soluble guanylate cyclase. We also determined cyclic GMP levels in tumors and normal brain, with or without intracarotid bradykinin infusion, using RIA. Intracarotid infusion of bradykinin selectively increased permeability in RG2 tumors and C6 tumors for both tracers. Simultaneous infusion of bradykinin and a cyclic GMP-specific phosphodiesterase inhibitor, zaprinast (20 mg/kg), resulted in significantly increased permeability across the BTB, compared to intracarotid bradykinin infusion alone. Zaprinast also significantly prolonged the permeability effects of bradykinin. Pretreatment using i.v. infusion of the soluble guanylate cyclase inhibitor, LY-83583 (125 microg/kg), significantly attenuated the bradykinin effect of opening the BTB. Cyclic GMP levels in RG2 and C6 tumors were significantly increased after intracarotid bradykinin infusion (2.8- and 2.2-fold, respectively). Cyclic GMP levels in normal brain were not increased by bradykinin infusion. These results show that increasing cyclic GMP in tumor microvessels can increase permeability in response to bradykinin.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Bradykinin/administration & dosage , Brain Neoplasms/metabolism , Cell Membrane Permeability/drug effects , Glioma/metabolism , Phosphodiesterase Inhibitors/administration & dosage , Purinones/administration & dosage , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Animals , Biological Transport/drug effects , Brain Neoplasms/pathology , Drug Synergism , Female , Glioma/pathology , Rats , Rats, Wistar
19.
Anesthesiology ; 88(2): 410-6, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9477062

ABSTRACT

BACKGROUND: Zaprinast, an inhibitor of guanosine-3',5'-cyclic monophosphate (cGMP)-selective phosphodiesterase, augments smooth muscle relaxation induced by endothelium-dependent vasodilators (including inhaled nitric oxide [NO]). The present study was designed to examine the effects of inhaled nebulized zaprinast, alone, and combined with inhaled NO. METHODS: Eight awake lambs with U46619-induced pulmonary hypertension sequentially breathed two concentrations of NO (5 and 20 ppm), followed by inhalation of aerosols generated from solutions containing four concentrations of zaprinast (10, 20, 30, and 50 mg/ml). The delivered doses of nebulized zaprinast at each concentration (mean +/- SD) were 0.23 +/- 0.06, 0.49 +/- 0.14, 0.71 +/- 0.24, and 1.20 +/- 0.98 mg x kg(-1) x min(-1), respectively. Each lamb also breathed NO (5 and 20 ppm) and zaprinast (0.23 +/- 0.06 mg x kg[-1] x min[-1]) in combination after a 2-h recovery period. RESULTS: Inhaled NO selectively dilated the pulmonary vasculature. Inhaled zaprinast selectively dilated the pulmonary circulation and potentiated and prolonged the pulmonary vasodilating effects of inhaled NO. The net transpulmonary release of cGMP was increased by inhalation of NO, zaprinast, or both. The duration of the vasodilation induced by zaprinast inhalation was greater than that induced by NO inhalation. CONCLUSIONS: Aerosolization of a cGMP-selective phosphodiesterase inhibitor alone or combined with NO may be a useful noninvasive therapeutic method to treat acute or chronic pulmonary hypertension.


Subject(s)
Lung/drug effects , Nitric Oxide/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Purinones/pharmacology , Vasodilation/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/adverse effects , Administration, Inhalation , Aerosols , Animals , Cyclic GMP/blood , Dose-Response Relationship, Drug , Drug Combinations , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Nitric Oxide/administration & dosage , Phosphodiesterase Inhibitors/administration & dosage , Pulmonary Circulation/drug effects , Purinones/administration & dosage , Sheep , Vasoconstrictor Agents/adverse effects
20.
Clin Exp Pharmacol Physiol ; 21(8): 607-14, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7813120

ABSTRACT

1. We tested the hypothesis that increasing myocardial cyclic GMP levels would reduce myocardial O2 consumption and areas of low O2 supply/consumption balance, using zaprinast, a selective cyclic GMP-phosphodiesterase inhibitor. 2. The study was conducted in three groups (vehicle, 10(-3) and 3 x 10(-3) mol/L zaprinast) of anaesthetized open-chest New Zealand white rabbits (n = 24). Coronary blood flow (radioactive microspheres), arterial and venous O2 saturation (microspectrophotometry), O2 consumption, cyclic GMP content (competitive binding) and cyclic GMP-phosphodiesterase activity (conversion of 3H-cyclic GMP to 3H-GMP) were determined. 3. Agents were applied to a patch on the myocardial surface and did not cause significant haemodynamic changes, except for bradycardia in the vehicle and low dose group. 4. The total myocardial cyclic GMP-phosphodiesterase activity was 148 +/- 14 while the zaprinast (10 mumol/L) inhibitable activity averaged 63 +/- 8 pmol/mg protein per min. Cyclic GMP content was increased with increasing doses of zaprinast (vehicle, 4.308 +/- 0.349 pmol/g; low dose zaprinast, 4.803 +/- 0.279 and high dose zaprinast, 7.938 +/- 1.304 pmol/g). 5. Coronary blood flow was not different after treatment (198 +/- 11, 209 +/- 10 and 153 +/- 9 mL/min per 100 g for the vehicle, low and high dose zaprinast, respectively). 6. Under control conditions, 48% of the small veins had O2 saturations below 50%. With zaprinast, this value was reduced to 19% for the low and 24% for the high dose. 7. Average venous O2 saturation increased with zaprinast (49 +/- 2%, 61 +/- 3% and 59 +/- 1%).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Cyclic GMP/metabolism , Myocardium/enzymology , Oxygen/metabolism , Purinones/pharmacology , Animals , Binding, Competitive , Blood Pressure/drug effects , Coronary Circulation/drug effects , Dose-Response Relationship, Drug , Heart/drug effects , Heart Rate/drug effects , Microspheres , Oxygen/blood , Oxygen Consumption/drug effects , Purinones/administration & dosage , Rabbits , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...