Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Physiol ; 550(Pt 1): 149-58, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12879865

ABSTRACT

Delta9-tetrahydrocannabinol (delta9-THC) is an effective anti-emetic; however, other potential gastrointestinal therapeutic effects of delta9-THC are less well-known. Here, we report a role of delta9-THC in a vago-vagal reflex that can result in gastro-oesophageal reflux, that is, gastric distension-evoked lower oesophageal sphincter (LOS) relaxation. Oesophageal, LOS and gastric pressures were measured using a miniaturized, manometric assembly in decerebrate, unanaesthetized ferrets.Gastric distension (30 ml) evoked LOS relaxation (70 +/- 8% decrease from baseline). Delta9-THC administered systemically (0.2 mg kg-1, iv.) or directly to the dorsal hindbrain surface (0.002 mg),significantly attenuated the nadir of the gastric distention-evoked LOS relaxation, and time to reach maximal response. Similar increases to maximal effect were observed after treatment with the cannabinoid receptor agonist WIN 55,212-2 (0.2 mg kg-1 iv.). The effect of systemic delta9-THC on gastric distention-evoked LOS relaxation was reversed by a selective cannabinoid1 (CBI) receptor antagonist, SR141617A (1 mg kg-1 i.v.). Since this reflex is vagally mediated, we used a CB1 receptor antiserum and immunocytochemistry to determine its distribution in ferret vagal circuitry. CBI receptor staining was present in cell bodies within the area postrema, nucleus tractus solitarius (NTS) and nodose ganglion. Intense terminal-like staining was noted within the NTS and dorsal motor vagal nucleus (DMN). Neither nodose ganglionectomy nor vagotomy altered the CB1 receptor terminal-like staining in the dorsal vagal complex. Retrogradely labelled gastric- or LOS-projecting DMN neurones did not express CBI receptors within their soma. Therefore, CBI receptor staining in the NTS and DMN is not due to primary vagal afferents or preganglionic neurones. These novel findings suggest that delta9-THC can modulate reflex LOS function and that the most likely site of action is via the CBI receptor within the NTS. This effect of delta9-THC may have implications in treatment of gastro-oesophageal reflux and other upper gut disorders.


Subject(s)
Esophagogastric Junction/physiology , Fourth Ventricle/metabolism , Muscle Relaxation/physiology , Muscle, Smooth/physiology , Receptor, Cannabinoid, CB1/physiology , Vagus Nerve/physiology , Animals , Decerebrate State , Dilatation , Dronabinol/pharmacology , Esophagogastric Junction/drug effects , Ferrets , Fourth Ventricle/physiology , Immunohistochemistry , Male , Manometry , Pressure , Receptor, Cannabinoid, CB1/drug effects , Stomach/physiology
2.
Neurogastroenterol Motil ; 14(3): 295-304, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12061915

ABSTRACT

An understanding of the neural control of lower oesophageal sphincter (LOS) relaxation is clinically relevant because transient LOS relaxations (TLOSRs) are a mechanism of acid reflux into the oesophagus. Preganglionic motor neurones innervating the LOS are localized in the dorsal motor nucleus of the vagus (DMV). Based on a single study in cats, it is now widely accepted that these neurones are functionally organized into two separate populations, such that stimulation of the caudal and rostral DMV evokes LOS relaxation and contraction, respectively. Our goal was to map the functional LOS responses to chemical stimulation in the DMV and nucleus tractus solitarius (NTS) of ferrets, an animal model commonly used for conscious studies on TLOSRs, and to test whether DMV-evoked LOS relaxation is mediated through hexamethonium-sensitive vagal-inhibitory pathways to the LOS. We used miniaturized manometry with Dentsleeve to monitor LOS and oesophageal pressures in decerebrate unanaesthetized ferrets. LOS relaxation was evoked readily in response to gastric insufflation, which shows that the vago-vagal reflex was intact in this preparation. Microinjections of l-glutamate (12.5 nmol L-1 in 25 nL) were made into the DMV from approximately - 1.5 to + 2.0 mm relative to the obex. Microinjections into the caudal (- 1.5 to + 0.0 mm behind obex) and intermediate (+ 0.1 to + 1.0 mm rostral to obex) DMV both significantly decreased LOS pressure, and complete LOS relaxation was noted in 28/32 and 11/18 cases, respectively. LOS relaxation responses to DMV microinjection were highly reproducible and abolished by bilateral vagotomy or hexamethonium (15 mg kg-1 intravenously). A nitric oxide synthase inhibitor (l-NAME 100 mg kg-1 intramuscularly) significantly increased the time taken to reach the maximal response. Increases in LOS pressure (24 +/- 4 mmHg; n = 3) were obtained only when stimulation sites were located equal to greater than 1.5 mm rostral to the obex. LOS relaxation (- 78 +/- 10%; n = 6) was evoked by stimulation of the NTS but not immediately outside of the NTS (11 +/- 27%; n = 5). We conclude that there is a very extensive population of 'inhibitory' motor neurones in the DMV that may account for the predominant vagal-inhibitory tone in ferrets. As NTS stimulation evokes LOS relaxation and the predominant response to DMV stimulation is also LOS relaxation, this vago-vagal reflex may involve an excitatory interneurone between the NTS and DMV vagal inhibitory output.


Subject(s)
Esophagogastric Junction/physiology , Muscle Relaxation/physiology , Vagus Nerve/physiology , Animals , Esophagogastric Junction/drug effects , Ferrets , Glutamic Acid/pharmacology , Male , Medulla Oblongata/drug effects , Medulla Oblongata/physiology , Muscle Relaxation/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Solitary Nucleus/drug effects , Solitary Nucleus/physiology , Vagotomy , Vagus Nerve/drug effects
3.
Gastroenterology ; 120(7): 1749-62, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375956

ABSTRACT

BACKGROUND & AIMS: Stimulation of gamma-aminobutyric acid B metabotropic receptors (GBRs) by baclofen reduces the incidence of transient lower esophageal sphincter (LES) relaxations. The GBR effect may be a result of a central site of action in the dorsal vagal complex, where upper gastrointestinal vagal reflexes are integrated. Therefore, we first localized GBR immunostaining in the dorsal vagal complex. Next, we tested the hypothesis that baclofen modulates LES motor tone via GBR expressed by vagal efferent neurons. METHODS: An antibody against the human GBR1b isoform was characterized and used for immunocytochemistry in rats and ferrets. Functional studies involved microinjection of L-glutamate into the caudal dorsal motor nucleus of the vagus to evoke an LES relaxation in decerebrate unanesthetized ferrets. RESULTS: In both species, GBR1b was expressed in preganglionic motor neurons and, in ferrets, the receptor was highly expressed in identified LES-projecting preganglionic neurons. GBR1b immunostaining was also pronounced in the subnucleus centralis of the nucleus tractus solitarius. This distribution implicates GBR in control of the esophageal phase of swallowing at the level of the central program generator. In functional studies, centrally evoked LES relaxation (-73% +/- 8% mm Hg) was significantly attenuated after 7 micromol/kg intravenous baclofen (-37% +/- 10%; N = 5). CONCLUSIONS: These data all suggest that GBR agonists inhibit LES relaxation via a site of action associated with vagal motor outflow to the LES.


Subject(s)
Esophagogastric Junction/physiology , Receptors, GABA-B/physiology , Vagus Nerve/physiology , Amino Acid Sequence , Animals , Antibody Specificity , Baclofen/pharmacology , Blotting, Western , Esophagogastric Junction/chemistry , Esophagogastric Junction/innervation , Ferrets , Immunohistochemistry , Male , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/analysis
4.
J Comp Neurol ; 430(2): 222-34, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11135258

ABSTRACT

The motor control of the lower esophageal sphincter (LES) is critical for normal swallowing and emesis, as well as for the prevention of gastroesophageal reflux. However, there are surprisingly few data on the central organization and neurochemistry of LES-projecting preganglionic neurons. There are no such data in ferrets, which are increasingly being used to study LES relaxation. Therefore, we determined the location of preganglionic neurons innervating the ferret LES, with special attention to their relationship with gastric fundus-projecting neurons. The neurochemistry of LES-projecting neurons was also investigated using two markers of "nontraditional" neurotransmitters in vagal preganglionic neurons, nitric oxide synthase (NOS), and dopamine (tyrosine hydroxylase: TH). Injection of cholera toxin B subunit (CTB)-horseradish peroxidase (HRP) into the muscular wall of the LES-labeled profiles throughout the rostrocaudal extent of the dorsal motor nucleus of the vagus (DMN) The relative numbers of profiles in three regions of the DMN from caudal to rostral are, 43 +/- 5, 67 +/- 11, and 113 +/- 30). A similar rostrocaudal distribution occurred after injection into the gastric fundus. When CTB conjugated with different fluorescent tags was injected into the LES and fundus both labels were noted in 56 +/- 3% of LES-labeled profiles overall. This finding suggests an extensive coinnervation of both regions by vagal motor neurons. There were significantly fewer LES-labeled profiles that innervated the antrum (16 +/- 9%). In the rostral DMN, 15 +/- 4% of LES-projecting neurons also contained NADPH-diaphorase activity; however, TH immunoreactivity was never identified in LES-projecting neurons. This finding suggests that NO, but not catecholamine (probably dopamine), is synthesized by a population of LES-projecting neurons. We conclude that there are striking similarities between LES- and fundic-projecting preganglionic neurons in terms of their organization in the DMN, presence of NOS activity and absence of TH immunoreactivity. Coinnervation of the LES and gastric fundus is logical, because the LES has similar functions to the fundus, which relaxes to accommodate food during ingestion and preceding emesis, but has quite different functions from the antrum, which provides mixing and propulsion of contents for gastric emptying. The presence of NOS in some LES-projecting neurons may contribute to LES relaxation, as it does in the case of fundic relaxation. The neurologic linkage of vagal fundic and LES relaxation may have clinical relevance, because it helps explain why motor disorders of the LES and fundus frequently occur together.


Subject(s)
Esophagogastric Junction/innervation , Ferrets/physiology , Ganglia/physiology , Neurons/physiology , Vagus Nerve/physiology , Animals , Ganglia/metabolism , Gastric Fundus/physiology , Medulla Oblongata/cytology , Medulla Oblongata/enzymology , Medulla Oblongata/physiology , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Rhombencephalon , Synaptic Transmission , Tyrosine 3-Monooxygenase/metabolism , Vagus Nerve/metabolism
5.
Am J Med ; 108 Suppl 4a: 90S-98S, 2000 Mar 06.
Article in English | MEDLINE | ID: mdl-10718459

ABSTRACT

The lower esophageal sphincter is innervated by both parasympathetic (vagus) and sympathetic (primarily splanchnic) nerves; however, the vagal pathways are the ones that are essential for reflex relaxation of the lower esophageal sphincter (LES), such as that which occurs during transient LES relaxations. Vagal afferent sensory endings from the distal esophagus and LES terminate in the hindbrain nucleus tractus solitarius. The preganglionic motor innervation of the LES arises from the dorsal motor nucleus of the vagus. Together these nuclei comprise the dorsal vagal complex within which there is a neural network coordinating reflex control of the sphincter. Vagal efferent preganglionic neurons to the gastrointestinal tract are organized viscerotopically in the dorsal motor nucleus of the vagus. Stimulation of the dorsal motor nucleus of the vagus caudal to the opening of the fourth ventricle results in relaxations, whereas stimulation in the rostral portion of the nucleus evokes contractions of the LES. Few details are known about the neural circuitry that links sensory information from the stomach and esophagus within the nucleus tractus solitarius to these separate populations of neurons within the dorsal motor nucleus of the vagus. The motor vagal preganglionic output is primarily cholinergic, which ultimately stimulates excitatory or inhibitory motor neurons that control the smooth muscle tone. Excitatory neurons evoke muscarinic receptor-mediated muscle contraction. Inhibitory neurons evoke nitric oxide or vasoactive intestinal polypeptide-mediated relaxation of the lower esophageal sphincter. However, other neurotransmitters are found in vagal preganglionic neurons, including norepinephrine/dopamine and nitric oxide. A subpopulation of nitric oxide synthase-containing vagal preganglionic neurons innervate the upper gastrointestinal tract and mediate relaxation. The neurotransmitters and circuitry controlling lower esophageal sphincter pressure are important to characterize, because part of the dorsal vagal complex is outside of the blood-brain barrier and is a potential target for pharmacologic intervention in the treatment of such disorders as gastroesophageal reflux disease.


Subject(s)
Deglutition , Esophagogastric Junction/innervation , Esophagogastric Junction/metabolism , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/pathology , Vagus Nerve/anatomy & histology , Humans , Neurotransmitter Agents/metabolism , Vagus Nerve/metabolism , Vagus Nerve/pathology
6.
J Auton Nerv Syst ; 77(2-3): 83-9, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10580290

ABSTRACT

Vagal stimulation results in both gastric motor excitatory and non-adrenergic non-cholinergic (NANC) inhibitory responses. The NANC pathway involves preganglionic cholinergic neurons, which act through nicotinic receptors to ultimately evoke gastric smooth muscle relaxation via release of nitric oxide (NO) and other neurotransmitters. Within the dorsal motor nucleus of the vagus (DMN), some preganglionic neurons also contain NO synthase. The NO synthase-containing neurons innervate the gastric fundus where adaptive relaxation occurs. This study tests the hypothesis that chemical stimulation of vagal motor neurons in animals, in which nicotinic receptors are blocked, evokes an NO-dependent gastric relaxation. A cell body excitant, N-methyl-D-aspartate (NMDA, 0.03-3 nmol), was microinjected into the DMN in anesthetized rats while recording intragastric pressure (IgP). The first group received NMDA before and after administration of a ganglionic blocker, hexamethonium bromide (15 mg/kg, i.v.) and atropine (1.0 mg/kg). Significant dose-dependent increases in IgP and gastric motility occurred before hexamethonium after the 0.3 and 3 nmol doses of NMDA. After hexamethonium, 0.3 and 3 nmol NMDA evoked significant decreases in IgP. A second group of rats was hexamethonium-pretreated and received NMDA microinjection into the DMN before and after an NO synthase inhibitor, N(G)-nitro-L-arginine methyl ester (10 mg/kg, i.v.). The NMDA-evoked decrease in IgP was completely abolished by the NO synthase inhibitor. These data support the novel idea that NO synthase-containing preganglionic neurons mediate gastric relaxation that is independent of nicotinic receptors.


Subject(s)
Motor Neurons/physiology , Muscle Relaxation/physiology , Stomach/physiology , Vagus Nerve/physiology , Animals , Area Under Curve , Atropine/pharmacology , Dose-Response Relationship, Drug , Gastrointestinal Motility/drug effects , Glutamic Acid/administration & dosage , Hexamethonium/pharmacology , Male , Medulla Oblongata/physiology , Microinjections , Motor Neurons/drug effects , Muscle Relaxation/drug effects , N-Methylaspartate/administration & dosage , Nicotinic Antagonists/pharmacology , Nitric Oxide/physiology , Rats , Rats, Sprague-Dawley , Stimulation, Chemical , Stomach/drug effects , Stomach/innervation , Vagus Nerve/drug effects
7.
Eur J Pharmacol ; 368(2-3): 173-82, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10193653

ABSTRACT

Blockade of GABA(A) receptors in the dorsal vagal complex produces marked gastric motor excitation. This effect is abolished by a prior microinjection of a non-selective excitatory amino acid receptor antagonist. Here we present functional evidence for kainate and NMDA receptor-mediated gastric excitation in the dorsal vagal complex. Microinjections into the dorsal vagal complex were performed in alpha-chloralose-anesthetized rats using multi-barrelled glass micropipettes while recording intragastric pressure and motility. Kainic acid (30 and 100 pmol in 30 nl) and NMDA (100 and 300 pmol) produced dose-related increases in intragastric pressure and motility. The gastric responses to kainate (30 pmol) and NMDA were selectively abolished by prior microinjection 6,7-dinitroquinoxaline-2,3-dione (600 pmol, 60 nl) and DL-2-amino-5-phosphanopentanoic acid (2 nmol), respectively. Atropine (1 mg/kg, i.v.) pretreatment blocked kainate-, NMDA- and L-glutamate-induced gastric excitation. Thus, both kainate- and NMDA-receptors in the dorsal vagal complex can independently cause vagally-mediated gastric motor excitation.


Subject(s)
Gastrointestinal Motility/physiology , Vagus Nerve/physiology , Animals , Atropine/pharmacology , Blood Pressure/drug effects , Brain Stem , Bronchodilator Agents/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gastrointestinal Motility/drug effects , Glutamic Acid/pharmacology , Kainic Acid/pharmacology , Male , Microinjections , N-Methylaspartate/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Stomach/drug effects , Stomach/innervation , Stomach/physiology , Vagus Nerve/drug effects
8.
Physiol Behav ; 63(4): 629-34, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9523908

ABSTRACT

Cocaine increases the circulating levels of plasma catecholamines, presumably via the activation of the sympathoadrenal axis. However, a number of reports have shown that the predominant response to cocaine is a generalized decrease in sympathetic nerve activity. One possible explanation for the increase in plasma catecholamines may be that the adrenal sympathetic nerve is less sensitive to the sympathoinhibitory actions of cocaine than are other nerves. This study compared the effects of cocaine on adrenal and renal sympathetic nerve discharge (SND) recorded simultaneously in pentobarbital-anesthetized rats. Cocaine produced dose-related decreases in both renal and adrenal SND; however, the decreases in adrenal SND were significantly smaller than in renal SND. Cocaine also elicited pressor responses in these rats. The decreases in adrenal SND were similar in baroreceptor intact and sinoaortically denervated rats, indicating that pressor-mediated baroreceptor reflex activation was not responsible for the decrease in adrenal SND. In a separate group of rats, i.v. administration of desipramine decreased both adrenal and renal SND. As with cocaine, the decreases in adrenal SND after desipramine were smaller, suggesting that the differences in the neural responses did not reflect a differential local anesthetic effect of cocaine on the two nerves. In conclusion, these studies showed that cocaine decreases adrenal SND in pentobarbital-anesthetized rats. However, the adrenal sympathetic nerve is less sensitive than the renal nerve to the sympathoinhibitory actions of cocaine. Whether the adrenal SND remaining after cocaine contributes to the increase in plasma catecholamines produced by this drug remains to be determined.


Subject(s)
Adrenal Glands/innervation , Cocaine/pharmacology , Narcotics/pharmacology , Sympathetic Nervous System/drug effects , Adrenal Glands/drug effects , Anesthesia , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Desipramine/pharmacology , Dose-Response Relationship, Drug , Heart Rate/drug effects , Heart Rate/physiology , Male , Rats , Rats, Sprague-Dawley , Sinoatrial Node/physiology
9.
Am J Physiol ; 272(6 Pt 1): G1372-81, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9227472

ABSTRACT

Thyrotropin-releasing hormone (TRH) from the nucleus raphe obscurus (nROb) innervates the dorsal vagal complex (DVC) and activates gastric motor function. Assessment of the importance of TRH has been hampered by the lack of TRH receptor antagonists. To overcome this, rats were given intracisternal antisense oligonucleotides against the first 18 bases of TRH receptor mRNA, mismatch oligonucleotides, or saline. Rats were anesthetized, and L-glutamate (15 nmol), TRH (1 and 10 pmol), and saline were microinjected into the DVC and nROb while gastric motor function was monitored. Intracisternal TRH mRNA antisense oligonucleotides abolished the gastric excitatory affects of microinjection of TRH, but not L-glutamate, into the DVC, and the response to TRH recovered after 2 wk of no antisense treatment. Chemical stimulation of the nROb increased intragastric pressure in saline- and mismatch- but not antisense-treated animals. These studies demonstrate that intracisternal TRH receptor antisense oligonucleotides produce a selective and reversible "knockdown" of responsiveness to exogenous TRH in the DVC, as well as to excitation of an endogenous TRH pathway controlling gastric function. It also provides a new tool for assessment of TRH pathways in hindbrain control of gastric function.


Subject(s)
Gastrointestinal Motility/physiology , Oligonucleotides, Antisense/pharmacology , Receptors, Thyrotropin-Releasing Hormone/physiology , Thyrotropin-Releasing Hormone/pharmacology , Vagus Nerve/physiology , Animals , Base Sequence , Gastrointestinal Motility/drug effects , Glutamic Acid/pharmacology , Male , Oligonucleotides, Antisense/chemistry , RNA, Messenger , Raphe Nuclei/drug effects , Raphe Nuclei/physiology , Rats , Rats, Sprague-Dawley , Receptors, Thyrotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/antagonists & inhibitors , Vagus Nerve/drug effects
10.
J Pharmacol Exp Ther ; 279(2): 967-74, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8930206

ABSTRACT

The purpose of this study was to determine whether neurons in the rostral ventrolateral medulla play a role in the sympathoinhibitory response elicited by i.v. administration of cocaine and, if so, to identify the type(s) of receptors involved. Adrenergic antagonists were microinjected bilaterally into the rostral ventrolateral medulla in pentobarbital-anesthetized rats in an attempt to block the decrease in sympathetic nerve discharge (SND) elicited by cocaine (1 mg/kg i.v.). After the bilateral microinjection of saline, cocaine elicited a -56 +/- 5% (mean +/- S.E.) decrease in SND lasting 36 +/- 3 min. Cocaine also increased arterial pressure (21 +/- 3 mm Hg). Prior microinjection of the alpha-2 adrenergic antagonist idazoxan (0.3, 3 or 10 nmol) did not alter the magnitude of the sympathoinhibitory response to cocaine; however, the duration of the response was significantly reduced by all 3 doses (range 21 +/- 3 to 11 +/- 2 min). Similarly, microinjection of the alpha-2 adrenergic antagonist piperoxan (10 nmol) decreased the duration (from 45 +/- 8 to 23 +/- 4 min), but not the magnitude of the sympathoinhibitory response. Microinjection of either the alpha-1 adrenergic antagonist terazosin (0.24 nmol) or the beta adrenergic receptor antagonist propranolol (2 nmol) did not attenuate the decrease in SND elicited by cocaine. The cocaine-mediated pressor response was not affected by any of the antagonist treatments. These data show that the decrease in SND elicited by cocaine is mediated centrally and involves, at least in part, the activation of alpha-2 adrenergic receptors in the rostral ventrolateral medulla.


Subject(s)
Cocaine/pharmacology , Medulla Oblongata/physiology , Receptors, Adrenergic, alpha-2/physiology , Sympatholytics/pharmacology , Adrenergic alpha-2 Receptor Antagonists , Animals , Blood Pressure/drug effects , Idazoxan/pharmacology , Male , Medulla Oblongata/drug effects , Microinjections , Piperoxan/pharmacology , Prazosin/analogs & derivatives , Prazosin/pharmacology , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley
11.
J Auton Nerv Syst ; 58(3): 170-6, 1996 May 06.
Article in English | MEDLINE | ID: mdl-8738310

ABSTRACT

Recent studies have shown that cocaine decreases, rather than increases sympathetic nerve discharge (SND). Whether these sympathoinhibitory responses are the result of cocaine's actions on monoaminergic transmission (i.e. blockade of neuronal uptake or stimulation of transmitter release) or its local anesthetic actions is not known. The purpose of the present study was to determine the degree to which cocaine's actions on monoaminergic transmission are involved in mediating the sympathoinhibitory response to this drug. We examined the mean arterial pressure, heart rate and splanchnic sympathetic nerve responses elicited by cocaine (1 mg/kg, i.v.) in pentobarbital-anesthetized rats depleted of monoamines. Monoamines were depleted by administering reserpine (10 mg/kg, i.p.) either 24, or 48 and 24 h before the experiment. The rats were also given alpha-methyl-p-tyrosine (200 mg/kg, i.p.) 2 h before the experiment. Vehicle-treated rats served as controls. Depletion of monoamines markedly reduced resting arterial pressure and heart rate and significantly attenuated the pressor response and tachycardia elicited by tyramine (1 mg/kg, i.v.). In control rats, cocaine elicited marked (-64 +/- 4%) and prolonged (44 +/- 4 min) decreases in SND. The magnitude (-34 +/- 11%) and duration (23 +/- 6 min) of these responses were significantly attenuated after 1 day of monoamine depletion. After 2 days of depletion, the sympathoinhibitory response was abolished and was replaced by a small, brief increase in SND (10 +/- 3%). The pressor responses were similar in control and depleted rats, while the bradycardic response (-33 +/- 4 bpm) was significantly reduced after 1 and 2 days of monoamine depletion to -20 +/- 3 and -15 +/- 2 bpm, respectively. We conclude that a functionally intact monoaminergic system is essential for the sympathoinhibitory response to cocaine. Whether the pressor responses result from a non-monoaminergic or a reserpine and/or alpha-methyl-p-tyrosine resistant catecholaminergic mechanism is unknown.


Subject(s)
Adrenergic Fibers/drug effects , Adrenergic Fibers/metabolism , Biogenic Monoamines/metabolism , Cocaine/pharmacology , Splanchnic Nerves/drug effects , Adjuvants, Anesthesia/pharmacology , Animals , Blepharoptosis/chemically induced , Blood Pressure/physiology , Enzyme Inhibitors/pharmacology , Heart Rate/physiology , Injections, Intravenous , Male , Methyltyrosines/pharmacology , Neural Inhibition/drug effects , Pentobarbital/pharmacology , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology , Splanchnic Nerves/metabolism , Sympatholytics/pharmacology , Synaptic Transmission/drug effects , alpha-Methyltyrosine
12.
Clin Obstet Gynecol ; 39(1): 17-35, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8635298

ABSTRACT

This article reviews the basic principles of pharmacodynamics and pharmacokinetics, with a special emphasis on the pharmacologic considerations that must be taken into account when treating the patient with respiratory disease who is also pregnant or nursing the neonate. A description of the four classes of therapeutic agents used for COPD is given with a discussion of the scientific evidence for their safety during pregnancy. The understanding of asthma suggests that bronchodilators relieve the symptoms, while antiinflammatories suppress the disease. Direct administration to the target tissue by inhalation of the bronchodilators (beta-adrenoreceptor agonists and anticholinergics) and immunosuppressors (corticosteroids and cromolyn) leads to low systemic levels of these drugs, which reduces fetal drug exposure. Oral administration of beta-adrenoreceptor agonists, corticosteroids, and theophylline may be necessary to obtain sufficient maternal lung function and ensure adequate oxygenation of the fetus. This must be carefully weighed against the potential fetal and maternal risks involved with increased systemic levels of these drugs. A brief description of classes of drugs used for upper respiratory diseases (antihistamines, alpha-adrenergic agonists, corticosteroids, antitussives, and expectorants) and their safety during pregnancy is also given. There is concern that most alpha-adrenergic agonists increase blood pressure at therapeutic doses needed to relieve nasal congestion. Therefore, for pregnant patients requiring decongestants, opinion favors administration of pseudoephedrine, which has the most favorable therapeutic index, to reduce potential cardiovascular adverse reactions in the fetus. Intranasal administration of the newer corticosteroids, which have limited absorption, is useful for suppression of allergic rhinitis, while minimizing the risk of adverse reactions. The purpose of this article has been to provide pharmacologic/toxicologic information about commonly used respiratory drugs. This will to enable the clinician to make an educated decision regarding the choice of therapy for respiratory disorders to ensure that fetal and maternal outcomes are optimal.


Subject(s)
Bronchodilator Agents/pharmacology , Bronchodilator Agents/pharmacokinetics , Histamine H1 Antagonists/pharmacology , Histamine H1 Antagonists/pharmacokinetics , Lung Diseases, Obstructive/drug therapy , Pregnancy Complications/drug therapy , Asthma/drug therapy , Asthma/metabolism , Bronchodilator Agents/adverse effects , Female , Histamine H1 Antagonists/adverse effects , Humans , Lung Diseases, Obstructive/metabolism , Pregnancy , Pregnancy Complications/metabolism
13.
Physiol Behav ; 59(1): 109-15, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8848468

ABSTRACT

Recent evidence suggests that cocaine decreases rather than increases sympathetic nerve discharge (SND). The purpose of the present study was to provide the first complete characterization of the dose-response relationships of cocaine (0.005-3 mg/kg, IV) for arterial pressure, heart rate, and lumbar, splanchnic, or renal SND in pentobarbital-anesthetized rats. Cocaine was also tested in conscious rats. In pentobarbital-anesthetized rats cocaine elicited prolonged (lasting up to 56 min), dose-dependent decreases in SND on all three nerves. The splanchnic nerve was significantly more sensitive to the inhibitory actions of cocaine than was the lumbar nerve. Cocaine increased arterial pressure and elicited bradycardia at doses above 0.5 mg/kg. Comparison of the dose-response curves of cocaine for splanchnic SND in sham-operated and sinoaortically deafferentated (SAD) rats showed that the baroreceptor reflex made only a minor contribution to the magnitude of sympathoinhibitory response. However, the duration of the sympathoinhibitory response was significantly shorter in SAD than in sham animals. In conscious rats, cocaine (0.1 and 1.0 mg/kg) elicited a pattern of neural and cardiovascular responses similar to that seen in anesthetized rats, except that the prolonged sympathoinhibitory responses were preceded by a brief (lasting < 10 s) increase in SND. From these data we conclude that cocaine produces prolonged decreases in SND in conscious and anesthetized rats. These sympathoinhibitory responses do not appear to result from baroreceptor reflex activation and may involve a central mechanism of action.


Subject(s)
Cocaine/pharmacology , Sympathetic Nervous System/drug effects , Anesthesia , Animals , Baroreflex/drug effects , Blood Pressure/drug effects , Denervation , Dose-Response Relationship, Drug , Heart Rate/drug effects , Male , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Rats , Rats, Sprague-Dawley , Sinoatrial Node/physiology
14.
J Pharmacol Exp Ther ; 270(1): 424-32, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7913499

ABSTRACT

Recent studies have reported that microinjection of kynurenic acid (KYN 12.5 nmol), the nonselective Excitatory Amino acid (EAA) antagonist, into the rostral ventrolateral medulla of the cat decreases arterial blood pressure (BP) and inferior cardiac sympathetic nerve discharge. The purpose of our study was to confirm this finding and determine the subtypes of EAA receptor(s) responsible for mediating this effect. This was done by microinjecting various EAA antagonists bilaterally into the SRFN of chloralose-anesthetized animals while monitoring BP and HR. KYN (12.5 nmol; N = 5) produced a decrease in mean BP (31 +/- 9 mmHg, P < .05) with no significant change in HR. To determine the subtype of EAA receptor responsible for eliciting tonic sympathetic outflow from the SRFN, specific antagonists of N-methyl-D-aspartate (NMDA) and non-NMDA EAA receptors were tested. The NMDA receptor antagonist 3-(RS)-Carboxypiperazin-4-yl)-proyl- 1-phosphonic acid (CPP-2.25 nmol; N = 3) microinjected into the SRFN produced a small but significant decrease in BP (-13 +/- 1 mmHg; P < .05). This effect of CPP was significantly less than that seen with KYN. Two antagonists of the non-NMDA subtype of EAA receptor, 6-cyano-7-nitroquinoxaline-2,3-dione (0.05 nmol; N = 4) and gamma-D-glutamylaminomethyl sulphonic acid (2.5 nmol; N = 4), were microinjected into the SRFN. Both of these drugs produced decreases in BP (-29 +/- 4 and -23 +/- 3 mmHg, respectively; P < 0.05) similar to that observed with KYN. No significant changes in HR were noted with CPP, 6 cyano-7-nitroquinoxaline-2,3-dione or gamma-G-glutamylamino-methylsulfonate. These data indicate that a non-NMDA EAA receptor plays the major role in control of cardiovascular function by the SRFN.


Subject(s)
Blood Pressure/physiology , Cardiovascular Physiological Phenomena , Medulla Oblongata/physiology , Medulla Oblongata/ultrastructure , Receptors, Amino Acid/physiology , Receptors, N-Methyl-D-Aspartate/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione , Animals , Blood Pressure/drug effects , Cats , Female , Glutamine/analogs & derivatives , Glutamine/pharmacology , Kynurenic Acid/pharmacology , Male , Microinjections , Neurons/drug effects , Neurons/ultrastructure , Piperazines/pharmacology , Quinoxalines/pharmacology , Receptors, Amino Acid/antagonists & inhibitors , Receptors, Amino Acid/classification , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
15.
Eur J Pharmacol ; 238(2-3): 223-33, 1993 Jul 20.
Article in English | MEDLINE | ID: mdl-8405094

ABSTRACT

The aim of our study was to determine the role of excitatory amino acids in controlling cardiorespiratory activity. For this purpose we administered an antagonist of both N-methyl-D-aspartate (NMDA) and non-NMDA receptors (kynurenic acid), and an antagonist of the NMDA receptor complex (dizocilpine, more commonly known as MK-801) i.v. to chloralose-anesthetized cats while monitoring tracheal air flow, tidal volume, respiratory rate, inspiratory and expiratory durations, end tidal CO2, arterial blood pressure and heart rate. Administration of kynurenic acid in doses of 350 and 500 mg/kg produced respiratory depression as reflected by decreases in respiratory minute volume and increases in end tidal CO2. Inspiratory duration was increased with both doses and apnea (occurring during expiration) was observed with the high dose. Apnea was preceded by an apneustic pattern of breathing. Both doses resulted in an increase in blood pressure and, with the high dose, a later decrease in blood pressure was noted. Dizocilpine in doses ranging from 0.03 to 1 mg/kg produced dose-related decreases in respiratory minute volume, and increases in end tidal CO2. In addition, dizocilpine produced increases in inspiratory duration, an apneustic pattern of breathing and apnea (occurring during inspiration). Effects on blood pressure were similar to those observed with kynurenic acid. It is concluded that blockade of excitatory amino acid receptors results in pronounced effects on cardiorespiratory activity.


Subject(s)
Blood Pressure/drug effects , Dizocilpine Maleate/pharmacology , Kynurenic Acid/pharmacology , Receptors, Amino Acid/antagonists & inhibitors , Respiration/drug effects , Animals , Carotid Sinus/physiology , Cats , Denervation , Dizocilpine Maleate/administration & dosage , Female , Heart Rate/drug effects , Injections, Intravenous , Kynurenic Acid/administration & dosage , Male , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Tidal Volume/drug effects , Trachea/drug effects , Vagus Nerve/physiology
16.
Eur J Pharmacol ; 227(2): 189-97, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1426028

ABSTRACT

Experiments were performed to investigate if protein kinase C is involved in the norepinephrine-induced alpha 2-adrenoceptor-mediated inhibition of the Ca2+ current in adult rat superior cervical ganglion neurons. Ca2+ currents were recorded from dispersed superior cervical ganglion neurons, acutely isolated from adult rats, using the whole-cell patch-clamp technique. Both norepinephrine and the protein kinase C activator, 1,2-dioctanoylglycerol (diC8) decreased the Ca2+ current induced by step depolarizations to +10 mV from a holding potential of -80 mV. In the presence of norepinephrine, the Ca2+ current rising phase was adequately fit by a double exponential with a second time constant much larger than control, whereas in the presence of diC8 the rising phase became mono-exponential and the current displayed a prominent decay. Control tail current activation curves were described by the sum of two Boltzmann functions. Both norepinephrine and diC8 reduced peak tail current amplitude. Norepinephrine preferentially reduced the component activated at more hyperpolarized potentials, while diC8 preferentially reduced the component activated at more depolarized potentials. Intracellular application of three protein kinase C inhibitors: protein kinase C pseudosubstrate (PKC-19-36) (2 microM), staurosporine (1 microM) and 1-(5-isoquinolinylsulonyl)-2-sulfonylpiperazine (H-7) (50 microM), failed to affect norepinephrine-induced Ca2+ current inhibition. In addition, these protein kinase C inhibitors did not decrease the Ca2+ current inhibition induced by diC8.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Calcium/metabolism , Ganglia, Sympathetic/drug effects , Neurons/drug effects , Norepinephrine/pharmacology , Protein Kinase C/metabolism , Animals , Diglycerides/pharmacology , Electric Conductivity , Enzyme Activation , Ganglia, Sympathetic/enzymology , Ganglia, Sympathetic/metabolism , Male , Neurons/enzymology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
17.
J Pharmacol Exp Ther ; 259(3): 1388-95, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1684823

ABSTRACT

The purpose of the present study was to identify sites(s) in the ventrolateral medulla where excitatory amino acids are involved in respiratory control. For this purpose, the respiratory effects produced by bilateral microinjection of excitatory amino acid antagonist drugs were examined while tidal volume (Vt), respiratory rate (f), arterial blood pressure and heart rate were monitored in chloralose-anesthetized cats. Microinjection of kynurenic acid (12.5 nmol) into a site approximately 3 mm rostral to obex, 4 mm lateral to midline and 1.5 mm below the ventral surface produced a decrease in Vt (-20 +/- 2 ml), an increase in f (+20 +/- 3 breaths/min) and a decrease in respiratory minute volume (-108 +/- 19 ml/min) (n = 8). These changes progressed to apnea in each animal tested. No significant changes in blood pressure or heart rate were observed. To determine the excitatory amino acid receptor subtype(s) involved, antagonists of n-methyl-D-aspartate (NMDA) (3-[(RS)-carboxypiperazin-4-yl]-propyl-1-phosphoric acid (CPP] and non-NMDA [6-cyano-7-nitroquinoxaline-2,3-dione (CNQX)] receptors were microinjected bilaterally into this site. In the case of CPP, three doses were studied (0.25 nmol, n = 4; 0.75 nmol, n = 3; 2.25 nmol, n = 2). All three doses produced similar decreases in Vt (-12 +/- 1, P less than .05; -10 +/- 1, P less than .05; and -16 +/- 5 ml, respectively) and increases in f (+14 +/- 2, P less than .05; +10 +/- 3, P less than .05; and +12 +/- 3 breaths/min, respectively). None of these animals exhibited apnea.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Amino Acids/physiology , Medulla Oblongata/physiology , Respiration/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione , Amino Acids/antagonists & inhibitors , Animals , Cats , Female , Glycine/physiology , Kynurenic Acid/administration & dosage , Kynurenic Acid/pharmacology , Male , Microinjections , Piperazines/pharmacology , Quinoxalines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Respiration/drug effects
18.
J Pharmacol Exp Ther ; 257(1): 511-9, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2020007

ABSTRACT

Cocaine was administered i.v. to decerebrate cats while monitoring cardiac preganglionic sympathetic nerve activity (SNA), arterial blood pressure (BP) and heart rate (HR). Cocaine, 4 mg/kg i.v., reduced SNA by 55 +/- 6%, but did not significantly affect BP or HR. Cocaine, in doses that were ineffective by the i.v. route, was administered into the vertebral artery and produced decreases in SNA, BP and HR in anesthetized cats. Administration of cocaine into the carotid artery was without effect. Topical administration of cocaine to the intermediate area of the ventrolateral medullary surface (25 micrograms/side) evoked hypotension and bradycardia. Nisoxetine, an inhibitor of norepinephrine uptake, applied bilaterally to the intermediate area (30 micrograms/side) exerted a similar hypotensive effect. Lidocaine administered in doses equivalent to those of cocaine had no significant effect on SNA when given i.v. or on BP when given into the vertebral artery. These results indicate that cocaine inhibits central sympathetic outflow and that the site of action appears to be in the hindbrain at a site that is reached by placement of the drug at the intermediate area of the ventrolateral medulla. The data also indicate that the mechanism of action of cocaine to inhibit sympathetic outflow may be unrelated to its local anesthetic action and may involve inhibition of catecholamine uptake in the ventrolateral medulla.


Subject(s)
Brain/drug effects , Cocaine/pharmacology , Sympathetic Nervous System/drug effects , Animals , Blood Pressure/drug effects , Cats , Cocaine/administration & dosage , Female , Heart Rate/drug effects , Injections, Intraventricular , Male , Respiration/drug effects
19.
J Pharmacol Exp Ther ; 248(2): 863-70, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2537419

ABSTRACT

Studies were carried out in chloralose-anesthetized cats while monitoring respiratory (tidal volume and respiratory rate) and cardiovascular (arterial pressure and heart rate) activity. Midazolam applied bilaterally to the intermediate area of the ventral surface of the medulla in doses of 0.75, 7.5 and 75 micrograms/side reduced tidal volume by -6 +/- 3, -10 +/- 1 and -11 +/- 1 ml, respectively. A dose of 250 micrograms/side produced apnea in each animal tested. Corresponding changes in arterial pressure were -35 +/- 9, -44 +/- 6, -43 +/- 9 and -64 +/- 17 mm Hg, respectively. Larger doses of chlordiazepoxide (e.g., 1000 micrograms/side) were required to produce similar effects. Intravenous administration of midazolam in doses of 1.5 to 150 micrograms had no significant effect on cardiorespiratory activity. However, larger doses of midazolam given i.v. produced cardiorespiratory depression that was similar to that observed with centrally applied drug. Pretreatment or treatment with centrally applied flumazenil or bicuculline counteracted the cardiorespiratory effects of centrally applied midazolam. Most importantly, ventral surface application of flumazenil counteracted the cardiorespiratory depressant effects of i.v. midazolam. Central administration of ethyl-beta-carboline-3-carboxylate produced cardiorespiratory effects opposite to those seen with midazolam, and these stimulatory effects were also counteracted by centrally applied flumazenil. These results indicate that alterations in cardiorespiratory activity can be produced by drugs interacting with gamma-aminobutyric acid/benzodiazepine receptors at the ventral surface of the medulla.


Subject(s)
Blood Pressure/drug effects , Heart Rate/drug effects , Medulla Oblongata/drug effects , Receptors, GABA-A/drug effects , Respiration/drug effects , Allosteric Regulation , Animals , Carbolines/pharmacology , Cats , Chlordiazepoxide/pharmacology , Dose-Response Relationship, Drug , Female , Male , Midazolam/pharmacology , Tachyphylaxis
SELECTION OF CITATIONS
SEARCH DETAIL
...