Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 180(3): 255-263, 2023 02.
Article in English | MEDLINE | ID: mdl-36529953

ABSTRACT

Scientists who plan to publish in the British Journal of Pharmacology (BJP) should read this article before undertaking studies utilising anaesthetics in mammalian animals. This editorial identifies certain gaps in the reporting of details on the use of anaesthetics in animal research studies published in the BJP. The editorial also provides guidance, based upon current best practices, for performing in vivo experiments that require anaesthesia. In addition, mechanisms of action and physiological impact of specific anaesthetic agents are discussed. Our goal is to identify best practices and to provide guidance on the information required for manuscripts submitted to the BJP that involve the use of anaesthetic agents in studies with experimental animals.


Subject(s)
Anesthesia , Anesthetics , Animal Experimentation , Animals , Anesthetics/pharmacology , Mammals
2.
J Neurosci ; 40(15): 3052-3062, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32132265

ABSTRACT

Maintenance of cardiorespiratory homeostasis depends on autonomic reflexes controlled by neuronal circuits of the brainstem. The neurophysiology and neuroanatomy of these reflex pathways are well understood, however, the mechanisms and functional significance of autonomic circuit modulation by glial cells remain largely unknown. In the experiments conducted in male laboratory rats we show that astrocytes of the nucleus of the solitary tract (NTS), the brain area that receives and integrates sensory information from the heart and blood vessels, respond to incoming afferent inputs with [Ca2+]i elevations. Astroglial [Ca2+]i responses are triggered by transmitters released by vagal afferents, glutamate acting at AMPA receptors and 5-HT acting at 5-HT2A receptors. In conscious freely behaving animals blockade of Ca2+-dependent vesicular release mechanisms in NTS astrocytes by virally driven expression of a dominant-negative SNARE protein (dnSNARE) increased baroreflex sensitivity by 70% (p < 0.001). This effect of compromised astroglial function was specific to the NTS as expression of dnSNARE in astrocytes of the ventrolateral brainstem had no effect. ATP is considered the principle gliotransmitter and is released by vesicular mechanisms blocked by dnSNARE expression. Consistent with this hypothesis, in anesthetized rats, pharmacological activation of P2Y1 purinoceptors in the NTS decreased baroreflex gain by 40% (p = 0.031), whereas blockade of P2Y1 receptors increased baroreflex gain by 57% (p = 0.018). These results suggest that glutamate and 5-HT, released by NTS afferent terminals, trigger Ca2+-dependent astroglial release of ATP to modulate baroreflex sensitivity via P2Y1 receptors. These data add to the growing body of evidence supporting an active role of astrocytes in brain information processing.SIGNIFICANCE STATEMENT Cardiorespiratory reflexes maintain autonomic balance and ensure cardiovascular health. Impaired baroreflex may contribute to the development of cardiovascular disease and serves as a robust predictor of cardiovascular and all-cause mortality. The data obtained in this study suggest that astrocytes are integral components of the brainstem mechanisms that process afferent information and modulate baroreflex sensitivity via the release of ATP. Any condition associated with higher levels of "ambient" ATP in the NTS would be expected to decrease baroreflex gain by the mechanism described here. As ATP is the primary signaling molecule of glial cells (astrocytes, microglia), responding to metabolic stress and inflammatory stimuli, our study suggests a plausible mechanism of how the central component of the baroreflex is affected in pathological conditions.


Subject(s)
Astrocytes/physiology , Baroreflex/physiology , Solitary Nucleus/physiology , Adenosine Triphosphate/physiology , Animals , Calcium Signaling/physiology , Male , Neurons, Afferent/metabolism , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/physiology , Purinergic P2Y Receptor Agonists/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/drug effects , Receptors, AMPA/drug effects , Receptors, Purinergic P2Y1/drug effects , SNARE Proteins/physiology , Serotonin/pharmacology , Vagus Nerve Stimulation
3.
Exp Physiol ; 102(4): 389-396, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28120502

ABSTRACT

NEW FINDINGS: What is the central question of this study? Arterial hypertension is associated with impaired neurovascular coupling in the somatosensory cortex. Abnormalities in activity-dependent oxygen consumption in brainstem regions involved in the control of cardiovascular reflexes have not been explored previously. What is the main finding and its importance? Using fast-cyclic voltammetry, we found that changes in local tissue PO2 in the nucleus tractus solitarii induced by electrical stimulation of the vagus nerve are significantly impaired in spontaneously hypertensive rats. This is consistent with previous observations showing that brainstem hypoxia plays an important role in the pathogenesis of arterial hypertension. The effects of arterial hypertension on cerebral blood flow remain poorly understood. Haemodynamic responses within the somatosensory cortex have been shown to be impaired in the spontaneously hypertensive rat (SHR) model. However, it is unknown whether arterial hypertension affects oxygen homeostasis in vital brainstem areas that control cardiovascular reflexes. In this study, we assessed vagus nerve stimulation-induced changes in local tissue PO2 (PtO2) in the caudal nucleus tractus solitarii (cNTS) of SHRs and normotensive Wistar rats. Measurements of PtO2 were performed using a novel application of fast-cyclic voltammetry, which allows higher temporal resolution of O2 changes than traditional optical fluorescence techniques. Electrical stimulation of the central cut end of the vagus nerve (ESVN) caused profound reductions in arterial blood pressure along with biphasic changes in PtO2 in the cNTS, characterized by a rapid decrease in PtO2 ('initial dip') followed by a post-stimulus overshoot above baseline. The initial dip was found to be significantly smaller in SHRs compared with normotensive Wistar rats even after ganglionic blockade. The post-ESVN overshoot was similar in both groups but was reduced in Wistar rats after ganglionic blockade. In conclusion, neural activity-dependent changes in tissue oxygen in brainstem cardiovascular autonomic centres are significantly impaired in animals with arterial hypertension.


Subject(s)
Homeostasis/physiology , Hypertension/metabolism , Oxygen/metabolism , Solitary Nucleus/metabolism , Animals , Baroreflex/physiology , Blood Pressure/physiology , Brain Stem/metabolism , Brain Stem/physiopathology , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Heart Rate/physiology , Hypertension/physiopathology , Hypoxia/metabolism , Hypoxia/physiopathology , Rats , Rats, Inbred SHR , Rats, Wistar , Solitary Nucleus/physiopathology
4.
Exp Physiol ; 101(5): 588-98, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26876733

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does genetic ablation of neurokinin-1 receptors alter arterial blood pressure? What is the main finding and its importance? NK1R(-/-) mice have increased mean arterial blood pressure, but without a concomitant change in vascular reactivity. This finding suggests that neurokinin-1 receptors play a role in the neural regulation of blood pressure. Mice with functional ablation of the neurokinin-1 receptor gene, Tacr1, (NK1R(-/-) ) express behavioural abnormalities equivalent to those seen in attention deficit hyperactivity disorder (ADHD). An established model of ADHD is the spontaneously hypertensive rat, which exhibits high blood pressure owing to increased central sympathetic drive. In light of the evidence that the neurokinin-1 receptor (NK1R) also influences cardiovascular haemodynamics, we have investigated whether NK1R(-/-) mice exhibit raised blood pressure. Cardiovascular parameters were recorded for 24 h in conscious mice using radiotelemetry. Vascular function was assessed in mesenteric resistance arteries by wire myography. The NK1R(-/-) mice exhibited a higher blood pressure than wild-type animals throughout the 24 h period. Heart rate and locomotor activity in NK1R(-/-) mice were higher than in wild-type mice during the night period (active phase), consistent with an ADHD-like phenotype, but not during the day. Mesenteric and renal arteries from NK1R(-/-) mice exhibited normal vascular function; the responses to vasoconstrictors (U46619 and phenylephrine) and the endothelium-dependent vasodilator, acetylcholine, were not altered in these animals, suggesting that the NK1R does not regulate vascular tone. Analysis of heart rate variability revealed a higher low-frequency to high-frequency ratio in NK1R(-/-) mice, indicative of increased cardiac sympathetic activity. We propose that the raised blood pressure in NK1R(-/-) mice could be due to a neural mechanism rather than a change in vascular reactivity. Further studies are required to understand this mechanism and to establish whether a subgroup of ADHD patients with polymorphism of the equivalent (TACR1) gene are affected in a similar way.


Subject(s)
Arterial Pressure/physiology , Arteries/metabolism , Arteries/physiopathology , Behavior, Animal/physiology , Receptors, Neurokinin-1/metabolism , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Animals , Attention Deficit Disorder with Hyperactivity/metabolism , Attention Deficit Disorder with Hyperactivity/physiopathology , Autonomic Nervous System Diseases/metabolism , Autonomic Nervous System Diseases/physiopathology , Disease Models, Animal , Heart Rate/physiology , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Phenotype , Reaction Time/physiology
5.
PLoS One ; 11(2): e0150108, 2016.
Article in English | MEDLINE | ID: mdl-26918777

ABSTRACT

Signalling pathways underlying the phenomenon of remote ischaemic preconditioning (RPc) cardioprotection are not completely understood. The existing evidence agrees that intact sensory innervation of the remote tissue/organ is required for the release into the systemic circulation of preconditioning factor(s) capable of protecting a transplanted or isolated heart. However, the source and molecular identities of these factors remain unknown. Since the efficacy of RPc cardioprotection is critically dependent upon vagal activity and muscarinic mechanisms, we hypothesized that the humoral RPc factor is produced by the internal organ(s), which receive rich parasympathetic innervation. In a rat model of myocardial ischaemia/reperfusion injury we determined the efficacy of limb RPc in establishing cardioprotection after denervation of various visceral organs by sectioning celiac, hepatic, anterior and posterior gastric branches of the vagus nerve. Electrical stimulation was applied to individually sectioned branches to determine whether enhanced vagal input to a particular target area is sufficient to establish cardioprotection. It was found that RPc cardioprotection is abolished in conditions of either total subdiaphragmatic vagotomy, gastric vagotomy or sectioning of the posterior gastric branch. The efficacy of RPc cardioprotection was preserved when hepatic, celiac or anterior gastric vagal branches were cut. In the absence of remote ischaemia/reperfusion, electrical stimulation of the posterior gastric branch reduced infarct size, mimicking the effect of RPc. These data suggest that the circulating factor (or factors) of RPc are produced and released into the systemic circulation by the visceral organ(s) innervated by the posterior gastric branch of the vagus nerve.


Subject(s)
Biological Factors/metabolism , Hindlimb/blood supply , Ischemia/blood , Ischemic Preconditioning , Myocardial Reperfusion Injury/prevention & control , Parasympathetic Nervous System/physiology , Vagus Nerve/physiology , Animals , Autonomic Denervation , Biological Factors/blood , Celiac Plexus/physiology , Constriction , Electric Stimulation , Ischemia/physiopathology , Ischemic Preconditioning/methods , Male , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Organ Specificity , Rats , Rats, Sprague-Dawley , Stomach/innervation , Vagotomy , Viscera/innervation
6.
J Physiol ; 593(7): 1715-29, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25694117

ABSTRACT

The nucleus tractus solitarii (NTS) integrates inputs from cardiovascular afferents and thus is crucial for cardiovascular homeostasis. These afferents primarily release glutamate, although 5-HT has also been shown to play a role in their actions. Using fast-cyclic voltammetry, an increase in 5-HT concentrations (range 12-50 nm) could be detected in the NTS in anaesthetized rats in response to electrical stimulation of the vagus and activation of cardiopulmonary, chemo- and baroreceptor reflexes. This 5-HT signal was not potentiated by the serotonin transporter (SERT) or the noradrenaline transporter (NET) inhibitors citalopram and desipramine (1 mg kg(-1) ). However, decynium-22 (600 µg kg(-1) ), an organic cation 3 transporter (OCT3)/plasma membrane monoamine transporter (PMAT) inhibitor, increased the 5-HT signal by 111 ± 21% from 29 ± 10 nm. The effectiveness of these inhibitors was tested against the removal time of 5-HT and noradrenaline applied by microinjection to the NTS. Citalopram and decynium-22 attenuated the removal of 5-HT but not noradrenaline, whereas desipramine had the reverse action. The OCT3 inhibitor corticosterone (10 mg kg(-1) ) had no effect. Blockade of glutamate receptors with topical kynurenate (10-50 nm) reduced the vagally evoked 5-HT signal by 50%, indicating that this release was from at least two sources. It is concluded that vagally evoked 5-HT release is under the regulation of the high-capacity, low-affinity transporter PMAT, not the low-capacity, high-affinity transporter SERT. This is the first demonstration that PMAT may be playing a physiological role in the regulation of 5-HT transmission and this could indicate that 5-HT is acting, in part, as a volume transmitter within the NTS.


Subject(s)
Equilibrative Nucleoside Transport Proteins/physiology , Serotonin Plasma Membrane Transport Proteins/physiology , Serotonin/physiology , Solitary Nucleus/physiology , Animals , Blood Pressure/drug effects , Citalopram/pharmacology , Desipramine/pharmacology , Electric Stimulation , Equilibrative Nucleoside Transport Proteins/antagonists & inhibitors , Heart Rate/drug effects , Kynurenic Acid/pharmacology , Male , Norepinephrine/pharmacology , Quinolines/pharmacology , Rats, Sprague-Dawley , Selective Serotonin Reuptake Inhibitors/pharmacology , Solitary Nucleus/drug effects , Vagus Nerve/physiology
7.
J Pharmacol Exp Ther ; 349(2): 288-96, 2014 May.
Article in English | MEDLINE | ID: mdl-24618127

ABSTRACT

The effect of blockade of either 5-hydroxytryptamine (5-HT)/serotonin transporter (SERT) with citalopram or the organic cation transporter 3 (OCT3)/plasma membrane monoamine transporter (PMAT) with decynium-22 (D-22) on spontaneous and evoked release of 5-HT in the nucleus tractus solitarius (NTS) was investigated in rat brainstem slices treated with gabazine. 5-HT release was measured indirectly by changes in the frequency and amplitude of glutamatergic miniature excitatory postsynaptic currents (mEPSCs) [in the presence of tetrodotoxin (TTX)] and evoked EPSCs. Blockade of 5-HT3 receptors with granisetron reduced, whereas the 5-HT3 agonist phenylbiguanide increased, the frequency of mEPSCs. 5-HT decreased mEPSC frequency at low concentrations and increased frequency at high concentrations. This inhibition was blocked by the 5-HT1A antagonist N-[2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl]-N-2-pyridinylcyclohexanecarboxamide (WAY-100635), which was ineffective on its own, whereas the excitation was reversed by granisetron. The addition of citalopram or D-22 caused inhibition, which was prevented by 5-HT1A blockade. Thus, in the NTS, the spontaneous release of 5-HT is able to activate 5-HT3 receptors, but not 5-HT1A receptors, as the release in their vicinity is removed by uptake. The ineffectiveness of corticosterone suggests that the low-affinity, high-capacity transporter is PMAT, not OCT3. For evoked 5-HT release, only D-22 caused an increase in the amplitude of EPSCs, with a decrease in the paired pulse ratio, and increased the number of spontaneous EPSCs after 20-Hz stimulation. Thus, for the evoked release of 5-HT, the low-affinity, high-capacity transporter PMAT, but not 5-HT transporter (5-HTT)/SERT, is important in the regulation of changes in 5-HT extracellular concentration.


Subject(s)
Glutamic Acid/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin/metabolism , Solitary Nucleus/metabolism , Vagus Nerve/physiology , Animals , Citalopram/pharmacology , Excitatory Postsynaptic Potentials , In Vitro Techniques , Male , Miniature Postsynaptic Potentials , Neurons/physiology , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Plasma Membrane Neurotransmitter Transport Proteins/antagonists & inhibitors , Pyridazines/pharmacology , Quinolines/pharmacology , Rats, Sprague-Dawley , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Solitary Nucleus/drug effects , Synaptic Transmission
8.
Eur J Pharmacol ; 682(1-3): 142-52, 2012 May 05.
Article in English | MEDLINE | ID: mdl-22374259

ABSTRACT

Central and peripheral 5-hydroxytryptamine (5-HT) receptors play a critical role in the regulation of micturition. Bolus doses of 5-HT(2A/2C) receptor agonists have been shown to activate the external urethral sphincter (EUS) and to inhibit micturition. This study was designed to determine the contribution of these two 5-HT receptor subtypes to activation of the EUS and inhibition of micturition utilising pharmacokinetic knowledge to better control drug exposure. Recordings of urethral and bladder pressure, EUS-Electromyogram (EMG), the micturition reflex induced by bladder filling, blood pressure and heart rate were made in anaesthetized female rats. The effects of intravenous (i.v.) infusions of the 5-HT(2) receptor agonist (2S)-1-(6-chloro-5-fluoroindol-1-yl)propan-2-amine fumarate (Ro 60-0175) in the absence or presence of the selective 5-HT(2C) receptor antagonist 6-chloro-5-methyl-N-[6-(2-methylpyridin-3-yl)oxypyridin-3-yl]-2,3-dihydroindole-1-carboxamide dihydrochloride (SB 242084) or 5-HT(2A) receptor antagonist (R)-(2,3-dimethoxyphenyl)-[1-[2-(4-fluorophenyl)ethyl]piperidin-4-yl]methanol (MDL-100,907) were studied on these variables. Continuous infusion of increasing concentrations of Ro 60-0175 only evoked EUS-EMG activity at the highest concentration, which was blocked by co-infusion of MDL-100,907 but not SB 242084. Urethral pressure was unaffected by any drug infusion. Ro 60-0175 at the lowest concentration inhibited the micturition reflex but as the concentration increased this was reversed to facilitation. SB 242084 blocked the inhibition while MDL-100,907 blocked the excitation. Activation of 5-HT(2A) not 5-HT(2C) receptors evoked EUS-EMG activity. In conclusion, 5-HT(2A) receptor activation facilitated the micturition reflex and evoked EUS-EMG while 5-HT(2C) receptor activation only inhibited the micturition reflex.


Subject(s)
Muscle, Skeletal/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacokinetics , Urethra/metabolism , Urination/drug effects , Anesthesia , Animals , Blood Pressure/drug effects , Drug Interactions , Electromyography , Female , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Rats , Serotonin 5-HT2 Receptor Antagonists/administration & dosage , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Urethra/drug effects , Urethra/physiology
9.
Eur J Pharmacol ; 668(1-2): 177-83, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21763305

ABSTRACT

Centrally applied nicotine causes changes in blood pressure and vasopressin release. The involvement of different neuronal nicotinic receptor subtypes in these actions was investigated in anaesthetized rats. Nicotine administered i.c.v. caused a dose-related increase in blood pressure and renal sympathoinhibition, while i.c. administration also caused a bradycardia. In the presence of the V(1A) receptor antagonist (i.v.), nicotine (i.c) now caused a depressor response along with sympathoinhibition and bradycardia. Nicotine (0.3 µmol/kg; i.c.v.) in the presence of the α4ß2 receptor antagonist, dihydro-ß-erythroidine, (i.c.v.) evoked renal sympathoexcitation, while the α7 receptor antagonist, methyllycaconitine, delayed the expected sympathoinhibition. Both receptor antagonists blocked the pressor response. Dihydro-ß-erythroidine (i.c., 10µmol/kg) alone caused a transient pressor response and increased renal nerve activity. Methyllycaconitine (i.c., 0.1 µmol/kg) alone caused a slow fall in blood pressure and renal nerve activity, while the higher doses caused a pressor response and increased renal nerve activity. It was concluded that for nicotine to release vasopressin, activation of both α4ß2 and α7 receptors is required. The ability of nicotine to cause sympathoinhibition is mediated by ß4*-containing receptors, possibly α3ß4 receptors, and that activation of these receptors can override the sympathoexcitatory action of α4ß2 and α7 receptors. The ability of dihydro-ß-erythroidine and high doses of methyllycaconitine i.c. to cause sympathoexcitation and a pressor response is due to receptor antagonists blocking these sympathoinhibitory ß4*-containing receptors, which receive a tonic cholinergic input. As the low dose of methyllycaconitine caused sympathoinhibition, this indicates that sympathoexcitatory α7 receptors also receive a tonic input.


Subject(s)
Anesthesia , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Nicotine/pharmacology , Receptors, Nicotinic/metabolism , Aconitine/administration & dosage , Aconitine/analogs & derivatives , Aconitine/pharmacology , Animals , Antidiuretic Hormone Receptor Antagonists , Dihydro-beta-Erythroidine/administration & dosage , Dihydro-beta-Erythroidine/pharmacology , Male , Nicotine/administration & dosage , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor
10.
Auton Neurosci ; 152(1-2): 75-83, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19926533

ABSTRACT

GABA receptors in the nucleus tractus solitarius (NTS) are known to play an important role in the mediation/modulation of various cardiovascular/respiratory functions. Vagal afferent activation of these neurones usually evokes an initial excitation followed by a long lasting inhibition. The present study examines the role of GABA(B) as well as GABA(A) receptors in the mediation of this inhibition in anaesthetized rats, using CGP 35348 and bicuculline, respective antagonists at these receptors, applied topically or by ionophoresis. Bicuculline delayed the onset and reduced the duration of this inhibition. The duration of this inhibition was further and significantly decreased when CGP 35348 was administered along with bicuculline but the delay in onset was unaffected. CGP 35348 application alone, had no effect on the vagal afferent-evoked inhibition. From intracellular recordings the early and late components of this inhibition were found to have reversal potentials close to E(Cl) and E(K), respectively. Therefore it is concluded that this inhibition is mediated by both GABA(A) and GABA(B) receptors, although GABA(B) mediated inhibition can only be unmasked when GABA(A) receptors are blocked.


Subject(s)
Neural Inhibition/physiology , Neurons/physiology , Receptors, GABA-A/physiology , Receptors, GABA-B/physiology , Solitary Nucleus/physiology , Vagus Nerve/physiology , Administration, Cutaneous , Afferent Pathways/drug effects , Anesthesia , Animals , Bicuculline/administration & dosage , Bicuculline/pharmacology , GABA Antagonists/administration & dosage , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , GABA-B Receptor Antagonists , Iontophoresis , Male , Neural Inhibition/drug effects , Neurons/drug effects , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Synaptic Transmission/physiology , Time Factors , Vagus Nerve/drug effects
11.
Br J Pharmacol ; 158(5): 1387-94, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19785653

ABSTRACT

BACKGROUND AND PURPOSE: Central 5-hydroxytryptamine (5-HT)-containing pathways utilizing 5-HT(7) receptors are known to be critical for the mediation of cardiovascular reflexes. The nucleus tractus solitarius (NTS) is a site involved in the integration of cardiovascular afferent information. The present experiments examined the involvement of the 5-HT(7) receptor in the processing of cardiovascular reflexes in the NTS. EXPERIMENTAL APPROACH: In anaesthetized rats extracellular recordings were made from 104 NTS neurones that were excited by electrical stimulation of the vagus nerve and/or activation of cardiopulmonary afferents. Drugs were applied ionophoretically in the vicinity of these neurones. KEY RESULTS: The non-selective 5-HT(7) receptor agonist 5-carboxamidotryptamine maleate (5-CT) applied to 78 neurones increased the firing rate in 18 by 59% and decreased it in 38 neurones by 47%. Similarly, the 5-HT(1A) agonist 8-OH-DPAT applied to 20 neurones had an excitatory (8), inhibitory (7) or no effect (5) on the 20 neurones tested. In the presence of the 5-HT(7) antagonist SB 258719 the 5-CT excitation was attenuated. Furthermore, the excitatory response of NTS neurones evoked by electrical stimulation of the vagus nerve or activation of cardiopulmonary afferents with intra atrial phenylbiguanide was attenuated by SB 258719. The inhibitory action of 5-CT was unaffected by SB 258719 and the 5-HT(1A) antagonist WAY-100635. WAY-100635 failed to have any effect on 5-CT and vagal afferent-evoked excitations. CONCLUSIONS AND IMPLICATIONS: Vagal afferent-evoked excitation of NTS neurones can be blocked by SB 258719, a selective 5-HT(7) antagonist. This observation further supports the involvement of 5-HT neurotransmission in NTS afferent processing.


Subject(s)
Receptors, Serotonin/physiology , Solitary Nucleus/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Action Potentials/drug effects , Afferent Pathways , Anesthesia , Animals , Electric Stimulation , Heart/innervation , Lung/innervation , Male , Neurons/drug effects , Neurons/physiology , Piperazines/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/analogs & derivatives , Serotonin/pharmacology , Serotonin 5-HT1 Receptor Agonists , Serotonin 5-HT1 Receptor Antagonists , Serotonin Antagonists/pharmacology , Sulfonamides/pharmacology , Synaptic Transmission , Vagus Nerve/physiology
12.
Trends Pharmacol Sci ; 29(9): 472-81, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19086344

ABSTRACT

All 5-hydroxytryptamine (5-HT; serotonin) receptors, except the 5-HT6 type, have been shown to be involved in cardiovascular regulation. In the periphery, 5-HT is stored in platelets, but the physiological role of 5-HT in the regulation of vascular tone (as opposed to its role in coagulation) remains unclear. However, central 5-HT1A, 5-HT3 and 5-HT7 receptors do play a physiological part in the regulation of cardiovascular reflexes, controlling changes in parasympathetic (vagal) drive to the heart. These reflexes also affect activity in the sympathetic nervous system, which itself can be inhibited by central 5-HT(1A) receptors to cause falls in blood pressure and excited by 5-HT2 receptors to cause rises in blood pressure. The physiological role of these receptors in the central regulation of the sympathetic nervous system is unclear, although 5-HT2 receptors could be involved in the development of deoxycorticosterone-acetate-salt hypertension, which is probably related to their role in the control of vasopressin release.


Subject(s)
Cardiovascular Physiological Phenomena/drug effects , Serotonin/physiology , Animals , Humans , Receptors, Serotonin/drug effects , Serotonin Antagonists/pharmacology , Serotonin Antagonists/therapeutic use , Serotonin Receptor Agonists/pharmacology , Serotonin Receptor Agonists/therapeutic use
13.
Exp Physiol ; 92(6): 1023-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17644702

ABSTRACT

Cardiac vagal preganglionic neurones (CVPNs) show respiratory modulation in the nucleus ambiguus but not in the dorsal vagal nucleus. Both types of neurones can be activated by pulmonary C fibre afferents. Another brainstem area that has been identified as containing CVPNs is the intermediate zone between the dorsal vagal nucleus and nucleus ambiguus. Experiments were carried out in alpha-chloralose-anaesthetized cats to determine the physiological properties of these CVPNs and their responses to pulmonary C fibre afferent activation. Seven CVPN axons in the right cardiac vagal branches were identified and found to be localized in the intermediate zone with a conduction velocity of between 1.2 and 1.6 m s(-1), in the C fibre range. These seven CVPNs [either showing spontaneous activity (n = 1) or having activity induced by dl-homocysteic acid applied ionophoretically (n = 3)] were neither respiratory modulated nor did they receive a baroreceptor input, thus being similar to those found in the dorsal vagal nucleus. Right atrial injections of phenylbiguanide excited all four CVPNs tested. In conclusion, CVPNs located in the intermediate zone have similar properties to those in the dorsal vagal nucleus but not the nucleus ambiguus.


Subject(s)
Heart Conduction System/cytology , Heart/innervation , Medulla Oblongata/cytology , Vagus Nerve/cytology , Action Potentials/drug effects , Action Potentials/physiology , Anesthetics, Intravenous , Animals , Autonomic Fibers, Preganglionic/physiology , Biguanides/pharmacology , Cats , Chloralose , Female , Heart/physiology , Heart Conduction System/physiology , Male , Medulla Oblongata/physiology , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Respiratory Physiological Phenomena , Serotonin Receptor Agonists/pharmacology , Vagus Nerve/drug effects , Vagus Nerve/physiology
14.
Brain Res ; 1144: 82-90, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17320834

ABSTRACT

This study evaluated the role of 5-HT7 receptors within the central nervous system in modulating cardiovascular responses to the activation of chemo-, baro- and cardiopulmonary reflexes and in the regulation of mean arterial pressure and heart rate, using intracisternal (i.c.) application of the selective 5-HT7 receptor antagonist SB-269970 in awake rats. Experiments were performed on male Wistar rats (300-320 g). At 4 days before the experiment, rats were anesthetized and placed in a stereotaxic frame implantation of a guide cannula in the direction of the cisterna magna to be used for microinjection of saline or SB-269970 (100 microg/kg). On the day before the experiments a femoral artery and vein were cannulated to record arterial pressure and heart rate and to inject drugs to activate cardiovascular reflexes, respectively. The chemo-, baro- and cardiopulmonary reflexes were activated in different experimental groups before and after i.c. injection of saline or SB-269970. The antagonism of 5-HT7 receptors reduced: (a) the pressor (50+/-4 vs. 19+/-9 mm Hg) and bradycardic (-247+/-13 vs. -69+/-27 bpm) responses to chemoreflex activation; (b) the fall in MAP (-54+/-4 vs. -20+/-6 mm Hg) and the bradycardia (-294+/-12 vs. -98+/-34 bpm) in response to cardiopulmonary reflex activation; and (c) the gain of the baroreflex (-2.3+/-0.1 to -0.9+/-0.2 bpm/mm Hg). Intracisternal application of SB-269970 increased significantly baseline MAP in those rats previously submitted to the activation of a cardiovascular reflex but in naïve rats produced no changes in the baseline MAP were observed. The fact that cardiovascular responses to all reflexes tested were attenuated by the antagonism of 5-HT7 receptors suggests that brainstem 5-HT7 receptors brainstem facilitate the processing of the autonomic responses to cardiovascular reflex activation and that a 5-HT-containing pathway to the brainstem provides a normalizing input during challenges produced by cardiovascular reflex activation which seems to be mediated by 5-HT7 receptors.


Subject(s)
Baroreflex/physiology , Blood Pressure/physiology , Heart Rate/physiology , Receptors, Serotonin/physiology , Wakefulness , Animals , Baroreflex/drug effects , Blood Pressure/drug effects , Drug Administration Routes , Heart Rate/drug effects , Male , Microinjections/methods , Phenols/pharmacology , Rats , Rats, Wistar , Serotonin/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Sulfonamides/pharmacology , Time Factors
15.
Br J Pharmacol ; 147 Suppl 2: S120-31, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16465176

ABSTRACT

At present the most investigated 5-HT receptor that has been shown to play a role in the control of micturition is the 5-HT(1A) receptor followed by 5-HT(7), 5-HT(2) and 5-HT(3) receptors. Most experiments focus on the control these receptors have on the parasympathetic outflow to the bladder and the somatic outflow to the external urethral sphincter (EUS) in the rat. Furthermore, 5-HT(1A) and 5-HT(7) receptors have been identified as having an excitatory physiological role in the control of bladder function. 5-HT(1A) receptors act, at least in the rat, at both a spinal (probably a heteroreceptor) and supraspinal (probably an autoreceptor) level, while 5-HT(7) receptors only act at a supraspinal level. Additionally, in the rat, 5-HT administered at a spinal or supraspinal site has an excitatory action, although earlier experiments have shown that activating 5-HT-containing brain areas causes inhibition of the bladder. Recent experiments have also indicated that blockade of the 5-HT(1A) receptor pathway shows rapid tolerance. However, no data exist for the development of tolerance for the 5-HT(7) receptor pathway. Neither receptor seems to play a role in the control of the urethra. Regarding 5-HT(2) receptors, activation of this receptor subtype inhibits micturition, and this inhibitory action may occur at a spinal, supraspinal or both levels. Although no physiological role for 5-HT(2C) receptors can yet be identified, 5-HT(2C) receptors have been implicated in the proposed supraspinal tonically active 5-HT(1A) autoreceptor (negative feedback) pathway. This proposition reconciles the data that central 5-HT-containing pathways are inhibitory to micturition, while 5-HT(1A) receptors, although inhibitory to adenylyl cyclase, have an excitatory function. This is because activation of 5-HT(1A) autoreceptors reduces the release of 5-HT thus reducing the activation of the 5-HT(2C) receptors, which are inhibitory in the control of micturition (disinhibition). Furthermore, 5-HT(2A) receptors in the rat and 5-HT(2C) receptors in the guinea pig cause activation of the EUS. In this respect, 5-ht(5A) receptors have also been identified in Onuf's nucleus, the site of somatic motoneurones controlling this sphincter. In the cat there is very little evidence to indicate that 5-HT receptors are involved in micturition except under pathological conditions in which activation of 5-HT(1A) receptors causes inhibition of micturition. Interestingly, under such conditions 5-HT(1A) receptors cause excitation of the EUS. Nevertheless, spinal 5HT(3) receptors have been implicated in the physiological control of micturition in the cat, but not yet in the rat. Overall, the data support the view that 5-HT receptors are important in the control of micturition. However, many more studies are required to fully understand these roles and why there are such species differences.


Subject(s)
Central Nervous System/metabolism , Receptors, Serotonin/metabolism , Urinary Bladder/metabolism , Urination , Animals , Autonomic Nervous System/metabolism , Cats , Central Nervous System/drug effects , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Rats , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin/drug effects , Receptors, Serotonin, 5-HT2/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Urinary Bladder/innervation , Urination/drug effects
16.
Brain Res ; 1054(1): 61-72, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16054601

ABSTRACT

Antagonism of central 5-HT1A and 5-HT7 receptors inhibits reflex-evoked vagal bradycardias indicating that 5-HT is released during these reflexes. The present experiments examined the effect of 5-HT depletion with para-chlorophenylalanine (p-CPA) on the cardiac vagal baroreflex and cardiopulmonary reflex in awake and anesthetized rats. Immunocytochemistry and neurochemical detection showed that p-CPA depleted the brainstem of 5-HT, but not of norepinephrine or dopamine. Depletion of 5-HT was associated with an increase in mean arterial pressure (MAP) in awake rats. This difference was abolished by anesthesia, which reduced MAP in both groups of animals. The baroreflex gain, whether calculated from the rise in pressure induced by phenylephrine or the fall in pressure evoked by sodium nitroprusside, was significantly attenuated in depleted rats compared to controls. This attenuation of the baroreflex gain was unaffected by subsequent anesthesia. 5-HT depletion also attenuated the cardiopulmonary reflex vagal bradycardias but this only reached statistical significance when the rats were anesthetized. The data support the view that 5-HT is released in the reflex activation of the cardiac vagal pathway.


Subject(s)
Anesthesia , Baroreflex/physiology , Cardiovascular System/physiopathology , Serotonin/deficiency , Vagus Nerve/physiopathology , Wakefulness/physiology , Adrenergic alpha-Agonists/pharmacology , Animals , Baroreflex/drug effects , Biguanides/pharmacology , Biogenic Monoamines/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Brain Chemistry/drug effects , Cardiovascular System/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Fenclonine/pharmacology , Heart Rate/drug effects , Heart Rate/physiology , Immunohistochemistry/methods , Male , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Vagus Nerve/drug effects , Vasodilator Agents/pharmacology , Wakefulness/drug effects
17.
Eur J Pharmacol ; 518(2-3): 152-7, 2005 Aug 22.
Article in English | MEDLINE | ID: mdl-16014306

ABSTRACT

Central 5-HT2A receptors have been implicated in central volume control by activating a central angiotensinergic pathway to cause the release of vasopressin. Interestingly, to induce DOCA-salt hypertension in rats vasopressin release is required. Thus the present experiments were carried out to determine whether continuous blockade of these receptors over 20 days, with the non-selective 5-HT2 receptor antagonist mianserin would prevent the development of deoxycorticosterone acetate (DOCA)-salt hypertension. Mianserin, given i.c.v. 90 or 60 microg twice daily for 20 days prevented the development of hypertension in conscious rats receiving DOCA-salt but did not affect blood pressure in rats on salt alone. Further, the dose of 30 microg given i.c.v. twice daily had no effect nor did the vehicle, polyethylene glycol (PEG), on the development of the hypertension. Mianserin 90 microg twice daily i.c.v. was also shown to prevent the increase in fluid intake, urinary flow and sodium excretion caused by DOCA-salt treatment. These data indicate that this action of mianserin is not due to an intrinsic hypotensive action but an action which involves interference with the mechanism by which DOCA-salt treatment causes hypertension. Thus the data overall support the view that to induce hypertension with DOCA-salt a central 5-HT-containing pathway needs to be activated, which then activates 5-HT2 receptors to cause the release of vasopressin which has previously been shown to be responsible for the initiation of DOCA-salt treatment hypertension.


Subject(s)
Hypertension/prevention & control , Mianserin/pharmacology , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/pharmacology , Animals , Blood Pressure/drug effects , Desoxycorticosterone/administration & dosage , Desoxycorticosterone/toxicity , Dose-Response Relationship, Drug , Drinking/drug effects , Heart Rate/drug effects , Hypertension/chemically induced , Hypertension/physiopathology , Injections, Intraventricular , Male , Mianserin/administration & dosage , Rats , Rats, Wistar , Receptors, Serotonin, 5-HT2/physiology , Serotonin Antagonists/administration & dosage , Sodium/urine , Time Factors , Urodynamics/drug effects , Weight Gain/drug effects
18.
J Physiol ; 566(Pt 3): 939-53, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15905216

ABSTRACT

Brainstem 5-hydroxytryptamine (5-HT, serotonin)-containing neurones modulate cardiovascular reflex responses but the differing roles of the many 5-HT receptors have not been thoroughly investigated. The present experiments on anaesthetized rats investigated the role of 5-HT3 receptors in modulating vagal afferent evoked activity of nucleus tractus solitarius (NTS) neurones. Recordings were made from 301 NTS neurones receiving an input at long (> 20 ms) minimum onset latency from stimulation of the vagus nerve. These included 140 neurones excited by activating non-myelinated cardiopulmonary afferents by right atrial injection of phenylbiguanide (PBG). Ionophoretic application of PBG, a highly selective 5-HT3 receptor agonist, significantly increased activity (from 2.4 +/- 0.4 to 5.5 +/- 0.8 spikes s(-1)) in 96 of 106 neurones tested and in all 17 neurones tested the increase in activity (3.4 +/- 1.1 to 7.0 +/- 1.9 spikes s(-1)) was significantly attenuated (3.0 +/- 0.9 to 3.8 +/- 1.1 spikes s(-1)) by the selective 5-HT3 receptor antagonist granisetron. Ionophoretic application of PBG potentiated responses to vagus nerve and cardiopulmonary afferent stimulation, and granisetron significantly attenuated this cardiopulmonary input (20.2 +/- 5.7 to 10.6 +/- 4.1 spikes burst(-1)) in 9 of 10 neurones tested. Ionophoretic application of AMPA and NMDA also excited NTS neurones and these excitations could be selectively antagonized by the non-NMDA and NMDA receptor antagonists DNQX and AP-5, respectively. At these selective currents, DNQX and AP-5 also attenuated PBG- and cardiopulmonary input-evoked increases in NTS activity. These data are consistent with the hypothesis that vagal inputs, including non-myelinated cardiopulmonary inputs to the NTS, utilize a 5-HT-containing pathway which activates 5-HT3 receptors. This excitatory response to 5-HT3 receptor activation may be partly a direct postsynaptic action but part may also be due to facilitation of the release of glutamate which in turn acts on either non-NMDA or NMDA receptors to evoke excitation.


Subject(s)
Action Potentials/physiology , Neurons/physiology , Receptors, Serotonin, 5-HT3/metabolism , Reflex/physiology , Solitary Nucleus/physiology , Vagus Nerve/physiology , Anesthesia , Animals , Feedback/physiology , Male , Rats , Rats, Sprague-Dawley
19.
J Physiol ; 563(Pt 1): 319-31, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15611034

ABSTRACT

5-Hydroxytryptamine (5-HT; serotonin)-containing neurones contribute to reflex activation of parasympathetic outflow in a number of species, but the 5-HT receptors mediating these effects have yet to be fully determined. The present experiments demonstrate that central 5-HT7 receptors are involved in the vagal bradycardia evoked during the cardiopulmonary reflex, baroreflexes and the chemoreflex, as well as other autonomic changes caused by these reflexes. The experiments examined the effects of the selective 5-HT7 receptor antagonists SB-269970 and SB-656104 on these reflexes. For the cardiopulmonary reflex, when compared to time-matched vehicle control experiments, intracisternal application of SB-269970 (30-300 microg kg(-1), i.c.) dose-dependently attenuated the evoked bradycardia. At the highest dose, SB-269970 also attenuated the reflex hypotension and sympathoinhibition. The structurally different 5-HT7 receptor antagonist SB-656104 (100 microg kg(-1), i.c.) similarly attenuated the reflex bradycardia and hypotension. SB-269970 (100 microg kg(-1), i.c.) also attenuated the bradycardias evoked by electrical stimulation of aortic nerve afferents and the baroreflex evoked by the pressor response to phenylephrine (3-25 microg kg(-1), i.v.). The gain of the baroreflex was also significantly attenuated (0.15 +/- 0.06 versus 0.34 +/- 0.06 ms mmHg(-1)). Finally, SB-269970 (100 microg kg(-1), i.c.) significantly attenuated both the bradycardia and sympathoexcitation evoked by the chemoreflex. These data indicate that central 5-HT7 receptors play an important facilitatory role in the reflex activation of vagal outflow to the heart.


Subject(s)
Brain/physiology , Heart/innervation , Heart/physiology , Neurons/physiology , Receptors, Serotonin/metabolism , Reflex/physiology , Vagus Nerve/physiology , Anesthesia , Animals , Baroreflex/physiology , Bradycardia/physiopathology , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...