Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Cyst Fibros ; 22(4): 656-664, 2023 07.
Article in English | MEDLINE | ID: mdl-37121795

ABSTRACT

BACKGROUND: MRT5005, a codon-optimized CFTR mRNA, delivered by aerosol in lipid nanoparticles, was designed as a genotype-agnostic treatment for CF lung disease. METHODS: This was a randomized, double-blind, placebo-controlled Phase 1/2 study performed in the US. Adults with 2 severe class I and/or II CFTR mutations and baseline ppFEV1 values between 50 and 90% were randomized 3:1 (MRT5005: placebo). Six dose levels of MRT5005 (4, 8, 12, 16, 20, and 24 mg) or placebo (0.9% Sodium Chloride) were administered by nebulization. The single ascending dose cohort was treated over a range from 8 to 24 mg; the multiple ascending dose cohort received five weekly doses (range 8-20 mg); and the daily dosing cohort received five daily doses (4 mg). RESULTS: A total of 42 subjects were assigned to MRT5005 [31] or placebo [11]. A total of 14 febrile reactions were observed in 10 MRT5005-treated participants, which were mild [3] or moderate [11] in severity; two subjects discontinued related to these events. Additionally, two MRT5005-treated patients experienced hypersensitivity reactions, which were managed conservatively. The most common treatment emergent adverse events were cough and headache. No consistent effects on FEV1 were noted. CONCLUSIONS: MRT5005 was generally safe and well tolerated through 28 days of follow-up after the last dose, though febrile and hypersensitivity reactions were noted. The majority of these reactions resolved within 1-2 days with supportive care allowing continued treatment with MRT5005 and careful monitoring. In this small first-in-human study, FEV1 remained stable after treatment, but no beneficial effects on FEV1 were observed.


Subject(s)
Cystic Fibrosis , Adult , Humans , Cystic Fibrosis/diagnosis , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , RNA, Messenger , Respiratory Aerosols and Droplets , Mutation , Double-Blind Method
2.
Mol Genet Metab ; 110(3): 303-10, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23988379

ABSTRACT

In the pivotal phase II/III trial of idursulfase administered intravenously to treat mucopolysaccharidosis II, approximately half of the patients developed antibodies to idursulfase. This post-hoc analysis of data from the phase II/III trial and extension study examined the relationship between antibody status and outcomes. A total of 63 treatment-naïve patients received 0.5 mg/kg of intravenous idursulfase weekly for two years. Thirty-two patients (51%) were positive for anti-idursulfase IgG antibodies, 23 of whom (37%) became persistently positive. All patients who developed an antibody response did so by their scheduled Week 27 study visit. Positive antibody status appeared to have no statistically significant effect upon changes in six-minute walk test distance, percent predicted forced vital capacity, or liver and spleen volume. All patients showed significant decreases in urinary GAG levels, although the antibody positive group maintained somewhat higher urinary GAG levels than their antibody-negative counterparts at the end of study (138.7 vs. 94.7 µg/mg creatinine, p = 0.001). Antibody positivity was not associated with a higher event rate for serious adverse events. Among patients who had no prior infusion-related reactions, antibody positive patients were 2.3 times more likely to have a first infusion-related reaction than those who would remain negative (p = 0.017); the risk increased to 2.5 times more likely for those who were persistently positive (p = 0.009). These differences in risk disappeared among patients with a previous infusion-related reaction, likely because of preventive measures. A genotype analysis for the 36 patients with available data found that patients with nonsense or frameshift mutations may be more likely to develop antibodies, to experience infusion-related reactions, and to have a reduced uGAG response than those with missense mutations, suggesting the possibility that antibodies are not a driver of clinical outcomes but rather a marker for genotype.


Subject(s)
Antibodies/immunology , Enzyme Replacement Therapy , Iduronate Sulfatase/immunology , Iduronate Sulfatase/therapeutic use , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis II/immunology , Administration, Intravenous , Adolescent , Adult , Child , Child, Preschool , Enzyme Replacement Therapy/adverse effects , Genotype , Glycoproteins/genetics , Glycosaminoglycans/urine , Humans , Iduronate Sulfatase/administration & dosage , Iduronate Sulfatase/adverse effects , Liver/metabolism , Liver/pathology , Mucopolysaccharidosis II/genetics , Organ Size , Spleen/metabolism , Spleen/pathology , Treatment Outcome , Young Adult
3.
CNS Neurol Disord Drug Targets ; 6(1): 31-43, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17305552

ABSTRACT

The role of histaminergic neurotransmission in the promotion of waking has been extensively studied in preclinical species. Appreciation for the role of histamine continues to expand with increasing understanding of the interaction of histamine within the broad network of neuromodulators that regulate sleep and wake. The effects of histamine on waking are transduced through the H(1) and the H(3) receptors in the central nervous system. Brain penetrant over-the-counter antihistamines comprised of antagonist actions at H(1) receptors as well as varying degrees of antimuscarinic properties are marketed as sleep aids, based on their well-known daytime drowsiness side effects. The data supporting their use as sedatives, however, are not consistent. H(3) receptors are presynaptic receptors that limit histamine release as well as that of monoamine neurotransmitters thought to participate in the maintenance of waking. In this review, we discuss the existing studies on various antihistamines and antagonists of the H(1) receptor in the regulation of sleep in preclinical studies, normal subjects and in subjects with sleep disorders. In addition, we review the current data available on the use of ligands at H(3) receptors for the modulation of sleep and wake.


Subject(s)
Circadian Rhythm/physiology , Histamine Agents/therapeutic use , Histamine/physiology , Sleep Disorders, Circadian Rhythm/drug therapy , Animals , Circadian Rhythm/drug effects , Histamine Agents/pharmacology , Humans , Receptors, Histamine H3/physiology
4.
Br J Pharmacol ; 143(5): 649-61, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15466448

ABSTRACT

1 1-[4-(3-piperidin-1-yl-propoxy)-benzyl]-piperidine (JNJ-5207852) is a novel, non-imidazole histamine H3 receptor antagonist, with high affinity at the rat (pKi=8.9) and human (pKi=9.24) H3 receptor. JNJ-5207852 is selective for the H3 receptor, with negligible binding to other receptors, transporters and ion channels at 1 microm. 2 JNJ-5207852 readily penetrates the brain tissue after subcutaneous (s.c.) administration, as determined by ex vivo autoradiography (ED50 of 0.13 mg kg(-1) in mice). In vitro autoradiography with 3H-JNJ-5207852 in mouse brain slices shows a binding pattern identical to that of 3H-R-alpha-methylhistamine, with high specific binding in the cortex, striatum and hypothalamus. No specific binding of 3H-JNJ-5207852 was observed in brains of H3 receptor knockout mice. 3 In mice and rats, JNJ-5207852 (1-10 mg kg(-1) s.c.) increases time spent awake and decreases REM sleep and slow-wave sleep, but fails to have an effect on wakefulness or sleep in H3 receptor knockout mice. No rebound hypersomnolence, as measured by slow-wave delta power, is observed. The wake-promoting effects of this H3 receptor antagonist are not associated with hypermotility. 4 A 4-week daily treatment of mice with JNJ-5207852 (10 mg kg(-1) i.p.) did not lead to a change in body weight, possibly due to the compound being a neutral antagonist at the H3 receptor. 5 JNJ-5207852 is extensively absorbed after oral administration and reaches high brain levels. 6 The data indicate that JNJ-5207852 is a novel, potent and selective H3 antagonist with good in vitro and in vivo efficacy, and confirm the wake-promoting effects of H3 receptor antagonists.


Subject(s)
Histamine Antagonists/pharmacology , Piperidines/pharmacology , Receptors, Histamine H3/drug effects , Wakefulness/drug effects , Administration, Oral , Animals , Autoradiography , Body Temperature/drug effects , Body Weight/drug effects , Cyclic AMP/metabolism , Electrodes , Electroencephalography/drug effects , Electromyography/drug effects , Histamine Antagonists/administration & dosage , Histamine Antagonists/pharmacokinetics , Humans , Injections, Intravenous , Male , Mice , Mice, Knockout , Mice, Obese , Motor Activity/drug effects , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Polysomnography , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/genetics , Sleep/drug effects , Transducers
5.
J Biol Chem ; 274(20): 14067-73, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318821

ABSTRACT

Binding of epidermal growth factor (EGF) to its receptor (EGFR) augments the tyrosine kinase activity of the receptor and autophosphorylation. Exposure of some tissues and cells to EGF also stimulates adenylyl cyclase activity and results in an increase in cyclic AMP (cAMP) levels. Because cAMP activates the cAMP-dependent protein kinase A (PKA), we investigated the effect of PKA on the EGFR. The purified catalytic subunit of PKA (PKAc) stoichiometrically phosphorylated the purified full-length wild type (WT) and kinase negative (K721M) forms of the EGFR. PKAc phosphorylated both WT-EGFR as well as a mutant truncated form of EGFR (Delta1022-1186) exclusively on serine residues. Moreover, PKAc also phosphorylated the cytosolic domain of the EGFR (EGFRKD). Phosphorylation of the purified WT as well as EGFRDelta1022-1186 and EGFRKD was accompanied by decreased autophosphorylation and diminished tyrosine kinase activity. Pretreatment of REF-52 cells with the nonhydrolyzable cAMP analog, 8-(4-chlorophenylthio)-cAMP, decreased EGF-induced tyrosine phosphorylation of cellular proteins as well as activation of the WT-EGFR. Similar effects were also observed in B82L cells transfected to express the Delta1022-1186 form of EGFR. Furthermore, activation of PKAc in intact cells resulted in serine phosphorylation of the EGFR. The decreased phosphorylation of cellular proteins and diminished activation of the EGFR in cells treated with the cAMP analog was not the result of altered binding of EGF to its receptors or changes in receptor internalization. Therefore, we conclude that PKA phosphorylates the EGFR on Ser residues and decreases its tyrosine kinase activity and signal transduction both in vitro and in vivo.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , ErbB Receptors/physiology , Animals , Catalytic Domain , Cattle , Cell Line , Humans , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Serine/metabolism , Tyrosine/metabolism
6.
Proc Natl Acad Sci U S A ; 94(18): 9602-7, 1997 Sep 02.
Article in English | MEDLINE | ID: mdl-9275169

ABSTRACT

Using the full-length and two engineered soluble forms (C1-C2 and Cla-C2) of type V adenylyl cyclase (ACV), we have investigated the role of an intramolecular interaction in ACV that modulates the ability of the alpha subunit of the stimulatory GTP-binding protein of AC (Gsalpha) to stimulate enzyme activity. Concentration-response curves with Gsalpha suggested the presence of high and low affinity sites on ACV, which interact with the G protein. Activation of enzyme by Gsalpha interaction at these two sites was most apparent in the C1a-C2 form of ACV, which lacks the C1b region (K572-F683). Yeast two-hybrid data demonstrated that the C1b region interacted with the C2 region and its 64-aa subdomain, C2I. Using peptides corresponding to the C2I region of ACV, we investigated the role of the C1b/C2I interaction on Gsalpha-mediated stimulation of C1-C2 and full-length ACV. Our data demonstrate that a 10-aa peptide corresponding to L1042-T1051 alters the profile of the activation curves of full-length and C1-C2 forms of ACV by different Gsalpha concentrations to mimic the activation profile observed with C1a-C2 ACV. The various peptides used in our studies did not alter forskolin-mediated stimulation of full-length and C1-C2 forms of ACV. We conclude that the C1b region of ACV interacts with the 10-aa region (L1042-T1051) in the C2 domain of the enzyme to modulate Gsalpha-elicited stimulation of activity.


Subject(s)
Adenylyl Cyclases/metabolism , GTP-Binding Proteins/metabolism , Adenylyl Cyclases/chemistry , Amino Acid Sequence , Enzyme Activation , Escherichia coli , Molecular Sequence Data , Protein Binding , Recombinant Proteins/chemistry
7.
Proc Natl Acad Sci U S A ; 94(7): 2915-20, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9096321

ABSTRACT

Type V adenylyl cyclase (ACV) belongs to the family of Ca2+-inhibited cyclases. We have generated two soluble forms of the enzyme containing the C1 or C1a region (which lacks the C-terminal 112 amino acids) linked to the C2 domain and compared their regulation with the full-length ACV. All three forms of ACV were stimulated by the alpha subunit of the stimulatory G protein Gs (G(s alpha)) and forskolin. However, the synergistic stimulation by both these activators was markedly enhanced in the soluble enzymes. Moreover, the alpha subunit of the inhibitory G protein Gi (G(i alpha)) inhibited all forms of the enzyme, indicating that the regions for G(s alpha) and G(i alpha) interaction are preserved in the soluble forms. Ca2+ inhibited forskolin-stimulated adenylyl cyclase (AC) activity of the full-length and C1-C2 forms of ACV but did not alter the activity of the C1a-C2 form. Maximal stimulation of AC activity by combination of G(s alpha) and forskolin obliterated the Ca2+-mediated inhibition of the full-length and C1-C2 forms of ACV. In 45Ca2+ overlay experiments, the C1-C2 but not the C1a-C2 soluble ACV bound Ca2+. Moreover, proteins corresponding to the C1a and C2 domains did not bind calcium. On the other hand, the proteins corresponding to C1 and its C-terminal 112 amino acids (C1b) bound 45Ca2+. To our knowledge, this is the first report of nonchimeric soluble forms of AC in which regulation by G(s alpha) and G(i alpha) is preserved. Moreover, we demonstrate that the 112 amino acid C1b region of ACV is responsible for the binding of Ca2+ and inhibition of enzyme activity.


Subject(s)
Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Cell Line , GTP-Binding Proteins/metabolism , Recombinant Proteins/metabolism , Solubility , Spodoptera
8.
Eur J Pharmacol ; 276(1-2): 41-7, 1995 Mar 24.
Article in English | MEDLINE | ID: mdl-7540140

ABSTRACT

The breakdown of the relaxation-inducing second messengers cAMP and cGMP is mediated by phosphodiesterases. Inhibitors of functionally present phosphodiesterases can be expected to induce relaxation by increasing the basic amount of cAMP and/or cGMP. In the cat gastric fundus, vinpocetine, which has some selectivity for phosphodiesterase type I, only induced contractions, but the inhibitors of type III [5-(4-acetimidophenyl)pyrazin-(1H)-one; SKF 94120], type IV (rolipram) and type V (zaprinast) phosphodiesterase all caused concentration-dependent relaxation, as did the non-specific phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (IBMX). The most potent relaxant agent was rolipram (EC50 9 +/- 5 x 10(-7) M and 3 +/- 1 x 10(-7) M in longitudinal and circular smooth muscle strips, respectively). These results suggest that type III, IV and V phosphodiesterases are functionally present in the cat gastric fundus and are involved in the regulation of tone. The possible influence of the phosphodiesterase inhibitors on non-adrenergic non-cholinergic (NANC) relaxation induced by nitric oxide (NO), vasoactive intestinal polypeptide (VIP) and train and sustained electrical field stimulation was then tested. Rolipram (3 x 10(-8) M), SKF 94120 (10(-5) M) and IBMX (10(-6) M) did not potentiate any of the relaxant stimuli studied. Zaprinast (10(-5) M), the cGMP specific type V phosphodiesterase inhibitor, caused a significant increase of the relaxation induced by exogenous NO and by train electrical field stimulation. These stimuli are thought to induce relaxation via an increase of intracellular cGMP.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Gastric Fundus/drug effects , Muscle Relaxation/drug effects , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Cats , Cyclic GMP/physiology , Dose-Response Relationship, Drug , Female , Gastric Fundus/physiology , In Vitro Techniques , Isoenzymes/physiology , Male , Nitric Oxide/pharmacology , Purinones/pharmacology , Pyrrolidinones/pharmacology , Rolipram , Vasoactive Intestinal Peptide/pharmacology
9.
Br J Pharmacol ; 111(4): 1280-6, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8032615

ABSTRACT

1. The relaxant responses of S-nitroso-L-cysteine (CysNO), S-nitroso-N-acetyl-D,L-penicillamine (SNAP), S-nitroso-N-acetyl-L-cysteine (SNAC) and S-nitrosoglutathione (GSNO) in the rat gastric fundus (forestomach) were studied and compared to the relaxant responses obtained in response to nitric oxide (NO) and electrical field stimulation (EFS, 10 s strains) of non-adrenergic non-cholinergic (NANC) nerves. 2. CysNO (10(-7)-3 x 10(-4) M) caused transient relaxation of the precontracted rat gastric fundus, comparable to the response to NO (10(-6)-10(-4) M) and EFS. SNAP, SNAC and GSNO elicited more sustained relaxations. 3. The cyclic GMP-specific phosphodiesterase inhibitor, zaprinast (3 x 10(-5) M) increased the relaxant effect of CysNO, SNAP and GSNO while the NO-synthase inhibitor, NG-nitro-L-arginine (L-NOARG, 3 x 10(-4) M) had no influence. 4. In the presence of LY 83583 (10(-5) M), which releases superoxide anions, the relaxant response to NO and CysNO was decreased, whereas that to all other stimuli was unaltered. The inhibitory effect of LY 83583 on CsNO-induced relaxations was prevented by superoxide dismutase (SOD, 1000 u ml-1). 5. Tissues incubated for 1 h with 5.5 x 10(-4) M nitroglycerin (GTN) became tolerant to GTN. In this condition, the relaxant response to 10(-5) M NO was maintained, while the relaxations by EFS (8 Hz) and 3 x 10(-5) M SNAP were significantly decreased. The reduction of the response to the other S-nitrosothiols was not significant. 6. The combination of nitrate tolerance and 10-5 M LY 83583 caused a significantly larger inhibition of the relaxant response to EFS (8 Hz) than nitrate tolerance alone. The combination of LY 83583 and GTN tolerance reduced the relaxant effect of 10-5 M NO to a similar extent to LY 83583 alone, while the relaxant response to 10-4 M GTN was reduced to the same extent as after 1 h exposure to 5.5 x 10-4 M GTN alone.7. It is concluded that S-nitrosothiols potently relax the rat gastric fundus, possibly by a cyclic GMP-dependent mechanism and S-nitrosothiols such as SNAC and GSNO may be involved in NANC neurotransmission.


Subject(s)
Cysteine/analogs & derivatives , Gastric Fundus/drug effects , Glutathione/analogs & derivatives , Nitroglycerin/pharmacology , Nitroso Compounds/pharmacology , Penicillamine/analogs & derivatives , S-Nitrosothiols , Aminoquinolines/pharmacology , Animals , Cyclic GMP/physiology , Cysteine/pharmacology , Drug Tolerance , Electric Stimulation , Female , Gastric Fundus/innervation , Gastric Fundus/physiology , Glutathione/pharmacology , In Vitro Techniques , Male , Nitric Oxide/pharmacology , Penicillamine/pharmacology , Rats , Rats, Wistar , S-Nitroso-N-Acetylpenicillamine , S-Nitrosoglutathione
10.
J Pharmacol Exp Ther ; 266(1): 172-8, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8331556

ABSTRACT

Vasoactive intestinal polypeptide (VIP) is a neurotransmitter of nonadrenergic noncholinergic (NANC) nerves in the cat gastric fundus. A possible additional role for nitric oxide (NO) was investigated in circular and longitudinal muscle strips from this tissue. Incubation with the inhibitor of NO-synthesis, NG-nitro-L-arginine methyl ester (L-NAME, 3 x 10(-4) M), inhibited the relaxant response to both short- and long-lasting electrical field stimulation. The substrate for NO synthesis, L-arginine (2 x 10(-3) M), prevented this inhibition. In experiments with long-lasting electrical field stimulation, trypsin (3 x 10(-6) M) and L-NAME inhibited the beginning of the NANC relaxation to the same extent, but the inhibition by trypsin was more pronounced at the end of the stimulation period. The inhibitory effect of L-NAME and trypsin was additive. L-NAME did not influence the relaxations induced by VIP (10(-7) M), NO (10(-5) M) and isoprenaline (3 x 10(-6) M). NG-nitro-L-arginine (3 x 10(-4) M) also did not change the response to VIP. The relaxation induced by 10(-5) M NO was not transient but was sustained. The plateau phase of this relaxation was reduced by trypsin but not by 3 x 10(-6) M tetrodotoxin. It is concluded that NO is involved in NANC relaxation of the cat gastric fundus. During sustained NANC relaxation in the cat gastric fundus, cotransmission of NO and VIP is possible.


Subject(s)
Adrenergic Fibers/physiology , Arginine/physiology , Cholinergic Fibers/physiology , Gastric Fundus/innervation , Muscle Relaxation/physiology , Muscle, Smooth/innervation , Neural Pathways/physiology , Nitric Oxide/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cats , Electric Stimulation , Female , Gastric Fundus/physiology , In Vitro Techniques , Isoproterenol/pharmacology , Male , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , NG-Nitroarginine Methyl Ester , Nitric Oxide/pharmacology , Time Factors , Trypsin/pharmacology , Vasoactive Intestinal Peptide/pharmacology
11.
Eur J Pharmacol ; 219(2): 331-4, 1992 Aug 25.
Article in English | MEDLINE | ID: mdl-1425959

ABSTRACT

In the rat gastric fundus, the reported cGMP-lowering agent LY 83583 (10(-5) M) inhibited the relaxation induced by nitric oxide (NO), without altering the response to isoprenaline, vasoactive intestinal polypeptide, sodium nitroprusside or electrical field stimulation of inhibitory non-adrenergic non-cholinergic neurones, which are thought to release NO. Incubation with superoxide dismutase partially prevented the effect of LY 83583. When added during a relaxation maintained by continuous NO infusion, LY 83583 reversed the relaxation. It is concluded that LY 83583 inactivates exogenous NO through the generation of superoxide anions.


Subject(s)
Aminoquinolines/pharmacology , Gastric Fundus/drug effects , Muscle, Smooth/drug effects , SRS-A/antagonists & inhibitors , Sympathetic Nervous System/physiology , Animals , Electric Stimulation , Female , Gastric Fundus/innervation , Male , Muscle Relaxation/drug effects , Nitric Oxide/pharmacology , Rats , Rats, Wistar
12.
Br J Pharmacol ; 106(1): 173-9, 1992 May.
Article in English | MEDLINE | ID: mdl-1504726

ABSTRACT

1. The influence of NG-nitro-L-Arginine (L-NNA) on non-adrenergic non-cholinergic (NANC) relaxations induced by electrical field stimulation was investigated in circular muscle strips of the guinea-pig gastric fundus. 2. In the presence of 10(-6) M atropine and 4 x 10(-6) M guanethidine, electrical field stimulation (40 V, 1 ms, 0.125-16 Hz) with 10 s trains at 5 min intervals induced short-lasting, frequency-dependent relaxations. Continuous stimulation, with cumulative increase of the stimulation frequency, induced sustained frequency-dependent relaxations. Both types of response were abolished by 3 x 10(-6) M tetrodotoxin. 3. L-NNA (10(-5) M and 10(-4) M) concentration-dependently reduced both types of NANC response. Pre-incubation with 2 x 10(-3) M L-arginine prevented the inhibitory action of 10(-5) M L-NNA and partially antagonized that of 10(-4) M L-NNA. D-arginine (2 x 10(-3) M) did not protect against the inhibitory effect of L-NNA. 4. L-NNA did not consistently influence the basal tone of the tissues. L-Arginine and D-arginine likewise did not influence basal tone; they also had no influence on the electrically-induced NANC relaxations. 5. NO (10(-6)-10(-4) M) induced short-lasting concentration-dependent relaxations, while vasoactive intestinal polypeptide (VIP, (10(-9)-10(-7) M) induced more sustained relaxations, that developed at a slower rate. The NO- and VIP-induced relaxations were not influenced by 10(-4) M L-NNA.6. These results suggest that NO is involved in NANC neurotransmission of the guinea-pig gastric fundus; its contribution to sustained NANC relaxation in the guinea-pig gastric fundus is much more important than in the rat.


Subject(s)
Arginine/analogs & derivatives , Autonomic Nervous System/drug effects , Gastric Fundus/drug effects , Muscle Relaxation/drug effects , Animals , Arginine/pharmacology , Atropine/pharmacology , Autonomic Nervous System/physiology , Dose-Response Relationship, Drug , Electric Stimulation , Female , Gastric Fundus/innervation , Gastric Fundus/physiology , Guanethidine/pharmacology , Guinea Pigs , Male , Muscle Contraction/drug effects , Nitric Oxide/pharmacology , Nitroarginine , Vasoactive Intestinal Peptide/pharmacology
13.
Eur J Pharmacol ; 210(3): 315-23, 1992 Jan 21.
Article in English | MEDLINE | ID: mdl-1377130

ABSTRACT

Vasoactive intestinal polypeptide (VIP) and nitric oxide (NO) have been proposed as inhibitory non-adrenergic non-cholinergic (NANC) neurotransmitters in the rat gastric fundus. The smooth muscle relaxant actions of VIP and NO are medaited by cAMP and cGMP, respectively; therefore the effect of inhibitors of phosphodiesterases responsible for cyclic nucleotide breakdown on relaxation induced by VIP, NO and electrical field stimulation was investigated. The non-specific phosphodiesterase inhibitor, 3-isobutyl-1-methylxanthine (IBMX), the cGMP-specific phosphodiesterase inhibitor, zaprinast, the adenylate cyclase activator, forskolin, and the cyclic nucleotide analog, 8-bromo cGMP, produced concentration-dependent relaxation of rat gastric fundus strips precontracted by PGF2 alpha. IBMX potentiated isoprenaline-induced relaxation but not relaxation induced by sodium nitroprusside, VIP, NO or electrical field stimulation. Zaprinast potentiated the relaxation induced by sodium nitroprusside, while having no influence on relaxation due to any other stimulus. The combination of both phosphodiesterase inhibitors did not significantly affect the electrically induced relaxation. It is concluded that both cAMP and cGMP mediate relaxation in the rat gastric fundus. Further research is needed to investigate the role of the cyclic nucleotides during NANC relaxation of this tissue.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Gastric Fundus/drug effects , Purinones/pharmacology , Animals , Colforsin/pharmacology , Drug Interactions , Electric Stimulation , Female , Male , Muscle Relaxation/drug effects , Rats , Rats, Inbred Strains
14.
J Auton Pharmacol ; 9(4): 279-91, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2549072

ABSTRACT

1. GABA induced concentration-dependent transient contractions of the guinea-pig duodenum, but only occasionally evoked small relaxatory responses. The GABA-induced contractions were blocked by atropine and tetrodotoxin but were not influenced by hexamethonium; during electrically evoked twitch contractions, GABA had a concentration-dependent inhibitory effect. 2. The concentration-response curve for the contractile effect of GABA was shifted to the right in a dose-dependent manner by bicuculline and picrotoxin, with a clear reduction of the maximal effect in the presence of picrotoxin. 3. Homotaurine and delta-aminovaleric acid but not baclofen mimicked the GABA-induced contractions; the responses induced by these GABAA receptor agonists were antagonized by atropine, tetrodotoxin and bicuculline. Baclofen concentration-dependently inhibited electrically evoked twitch contractions. 4. Ethylenediamine also had a GABA-like effect, and cross-desensitization developed between GABA and ethylenediamine. 5. The ethylenediamine-induced contractions were not antagonized by thiosemicarbazide; they were reduced by 3-mercaptopropionic acid but the GABA-induced contractions were reduced to the same extent. 6. It is concluded that GABA induces contraction of the guinea-pig duodenum by excitation of GABAA receptors on postganglionic cholinergic neurones; a GABAB receptor-mediated inhibitory effect can be observed during electrically evoked twitch contractions. Ethylenediamine mimicks the GABAA receptor-mediated effect probably by a direct effect on the GABAA receptors.


Subject(s)
Ethylenediamines/pharmacology , Muscle, Smooth/drug effects , gamma-Aminobutyric Acid/pharmacology , 3-Mercaptopropionic Acid/pharmacology , Animals , Bicuculline/pharmacology , Duodenum/drug effects , Electric Stimulation , Female , Guinea Pigs , In Vitro Techniques , Isometric Contraction/drug effects , Male , Muscle Contraction/drug effects , Picrotoxin/pharmacology , Receptors, GABA-A/drug effects , Taurine/analogs & derivatives , Taurine/pharmacology , Thiosemicarbazones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...