Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-28762595

ABSTRACT

BACKGROUND: Previous studies suggest an increased inhibition of dorsal motor nucleus of the vagus (DMV) neurons following exposure to a perinatal high fat diet (PNHFD); the underlying neural mechanisms, however, remain unknown. This study assessed the effects of PNHFD on inhibitory synaptic inputs to DMV neurons and the vagally dependent control of gastric tone and motility. METHODS: Whole-cell patch clamp recordings were made from DMV neurons in thin brainstem slices from Sprague-Dawley rats fed either a control diet or HFD (14 or 60% kcal from fat, respectively) from embryonic day 13 onwards; gastric tone and motility were recorded in in vivo anesthetized rats. KEY RESULTS: The non-selective GABAA antagonist, BIC (10 µmol L-1 ), induced comparable inward currents in PNHFD and control DMV neurons, but a larger current in PNHFD neurons at higher concentrations (50 µmol L-1 ). Differences were not apparent in neuronal responses to the phasic GABAA antagonist, gabazine (GBZ), the extrasynaptic GABAA agonist, THIP, the GABA transport blocker, nipecotic acid, or the gliotoxin, fluoroacetate, suggesting that PNHFD altered inhibitory transmission but not GABAA receptor density or function, GABA uptake or glial modulation of synaptic strength. Similarly, the increase in gastric motility and tone following brainstem microinjection of low doses of BIC (1-10 pmoles) and GBZ (0.01-0.1 pmoles) were unchanged in PNHFD rats while higher doses of BIC (25 pmoles) induced a significantly larger increase in gastric tone compared to control. CONCLUSIONS AND INFERENCES: These studies suggest that exposure to PNHFD increases the tonic inhibition of DMV neurons, possibly contributing to dysregulated vagal control of gastric functions.


Subject(s)
Brain Stem/physiology , Diet, High-Fat , Gastrointestinal Motility , Neural Inhibition , Neurons/physiology , Vagus Nerve/physiology , Animals , Bicuculline/administration & dosage , GABA Agonists/administration & dosage , GABA Antagonists , Isoxazoles/administration & dosage , Male , Neurons/drug effects , Pyridazines/administration & dosage , Rats, Sprague-Dawley , Vagus Nerve/drug effects
3.
Neurogastroenterol Motil ; 26(6): 832-40, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24720579

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is an acute neonatal inflammatory disease which may lead to intestinal necrosis, multisystem failure, and death. Currently, NEC is diagnosed by a combination of laboratory and radiographic tests conducted a posteriori i.e., when NEC is already clinically significant. Given the acute onset and rapid progression of NEC, a non-invasive biomarker that allows early detection of patients at risk is required as a matter of urgency. We evaluated whether the high frequency (HF) component of heart rate variability (HRV), a measure of vagal efferent tonic cholinergic activity may be used as a predictive biomarker for NEC-risk before the onset of clinical disease. METHODS: In this prospective study, stable preterm (gestational age 28-35 weeks) infants had HRV power spectra analyzed from surface electrocardiogram waveforms taken at rest on day 5-8 of life. We used regression modeling to determine the utility of HF-HRV in predicting NEC. KEY RESULTS: HF-HRV power was 21.5 ± 2.7 and 3.9 ± 0.81 ms(2) in infants that remained healthy and those that later developed stage 2+ NEC, respectively (p < 0.001). Nine of 70 enrolled infants developed NEC. The ROC discriminated a HF-HRV value of 4.68 ms(2) predictive for developing NEC with a sensitivity and specificity of 89% and 87%, and positive and negative predictive value of 50% and 98%, respectively. With predictive regression modeling, the risk (odds ratio) of developing NEC was 10 per every one SD decrease in HF-HRV. CONCLUSIONS & INFERENCES: Our preliminary data indicate that HF-HRV may serve as a potential, non-invasive predictive biomarker of NEC-risk in NICU infants.


Subject(s)
Enterocolitis, Necrotizing/diagnosis , Infant, Newborn, Diseases/diagnosis , Vagus Nerve/physiopathology , Biomarkers , Electrocardiography , Female , Gestational Age , Heart Rate/physiology , Humans , Infant, Newborn , Infant, Premature , Male , Risk
4.
Neurogastroenterol Motil ; 26(2): 272-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24261332

ABSTRACT

BACKGROUND: Vagally dependent gastric reflexes are mediated through vagal afferent fibers synapsing upon neurons of the nucleus tractus solitarius (NTS) which, in turn modulate the preganglionic parasympathetic dorsal motor nucleus of the vagus (DMV) neurons within the medullary dorsal vagal complex (DVC). The expression and transport of ghrelin receptors has been documented for the afferent vagus nerve, and functional studies have confirmed that vagal pathways are integral to ghrelin-induced stimulation of gastric motility. However, the central actions of ghrelin within the DVC have not been explored fully. METHODS: We assessed the responses to ghrelin in fasted rats using: (i) in vivo measurements of gastric tone and motility following IVth ventricle application or unilateral microinjection of ghrelin into the DVC and (ii) whole cell recordings from gastric-projecting neurons of the DMV. KEY RESULTS: (i) IVth ventricle application or unilateral microinjection of ghrelin into the DVC-elicited contractions of the gastric corpus via excitation of a vagal cholinergic efferent pathway and (ii) ghrelin facilitates excitatory, but not inhibitory, presynaptic transmission to DMV neurons. CONCLUSIONS & INFERENCES: Our data indicate that ghrelin acts centrally by activating excitatory synaptic inputs onto DMV neurons, resulting in increased cholinergic drive by way of vagal motor innervation to the stomach.


Subject(s)
Gastrointestinal Motility/drug effects , Ghrelin/pharmacology , Medulla Oblongata/drug effects , Vagus Nerve/drug effects , Animals , Gastrointestinal Motility/physiology , Male , Medulla Oblongata/physiology , Neurons/drug effects , Neurons/physiology , Rats , Rats, Long-Evans , Synaptic Potentials/drug effects , Vagus Nerve/physiology
5.
J Physiol ; 591(6): 1563-80, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23297311

ABSTRACT

Perivagal application of capsaicin (1% solution) is considered to cause a selective degeneration of vagal afferent C fibres and has been used extensively to examine the site of action of many gastrointestinal (GI) neuropeptides. The actions of both capsaicin and GI neuropeptides may not be restricted to vagal afferent fibres, however, as other non-sensory neurones have displayed sensitivity to capsaicin and brainstem microinjections of these neuropeptides induce GI effects similar to those obtained upon systemic application. The aim of the present study was to test the hypothesis that perivagal capsaicin induces degeneration of vagal efferents controlling GI functions. Experiments were conducted 7-14 days after 30 min unilateral perivagal application of 0.1-1% capsaicin. Immunohistochemical analyses demonstrated that, as following vagotomy, capsaicin induced dendritic degeneration, decreased choline acetyltransferase but increased nitric oxide synthase immunoreactivity in rat dorsal motor nucleus of the vagus (DMV) neurones. Electrophysiological recordings showed a decreased DMV input resistance and excitability due, in part, to the expression of a large conductance calcium-dependent potassium current and the opening of a transient outward potassium window current at resting potential. Furthermore, the number of DMV neurones excited by thyrotrophin-releasing hormone and the gastric motility response to DMV microinjections of TRH were decreased significantly. Our data indicate that perivagal application of capsaicin induced DMV neuronal degeneration and decreased vagal motor responses. Treatment with perivagal capsaicin cannot therefore be considered selective for vagal afferent C fibres and, consequently, care is needed when using perivagal capsaicin to assess the mechanism of action of GI neuropeptides.


Subject(s)
Capsaicin/pharmacology , Vagus Nerve/drug effects , Action Potentials/drug effects , Animals , Brain Stem/metabolism , Brain Stem/physiology , Calcium/metabolism , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Dendrites/physiology , Gastrointestinal Motility/drug effects , Gene Expression/drug effects , Motor Neurons/physiology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Thyrotropin-Releasing Hormone/pharmacology , Vagotomy , Vagus Nerve/metabolism , Vagus Nerve/physiology
6.
Neurogastroenterol Motil ; 24(10): e476-88, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22845622

ABSTRACT

BACKGROUND: Intestinal glucose induces gastric relaxation via vagally mediated sensory-motor reflexes. Glucose can alter the activity of gastrointestinal (GI) vagal afferent (sensory) neurons directly, via closure of ATP-sensitive potassium channels, and indirectly, via the release of 5-hydroxytryptamine (5-HT) from mucosal enteroendocrine cells. We hypothesized that glucose may also be able to modulate the ability of GI vagal afferent neurons to respond to the released 5-HT, via regulation of neuronal 5-HT(3) receptors. METHODS: Whole-cell patch clamp recordings were made from acutely dissociated GI-projecting vagal afferent neurons exposed to equiosmolar Krebs' solution containing different concentrations of d-glucose (1.25-20 mmol L(-1)) and the response to picospritz application of 5-HT assessed. The distribution of 5-HT(3) receptors in neurons exposed to different glucose concentrations was also assessed immunohistochemically. KEY RESULTS: Increasing or decreasing extracellular d-glucose concentration increased or decreased, respectively, the 5-HT-induced inward current and the proportion of 5-HT(3) receptors associated with the neuronal membrane. These responses were blocked by the Golgi-disrupting agent Brefeldin-A (5 µmol L(-1)) suggesting involvement of a protein-trafficking pathway. Furthermore, l-glucose did not mimic the response of d-glucose implying that metabolic events downstream of neuronal glucose uptake are required to observe the modulation of 5-HT(3) receptor mediated responses. CONCLUSIONS & INFERENCES: These results suggest that, in addition to inducing the release of 5-HT from enterochromaffin cells, glucose may also increase the ability of GI vagal sensory neurons to respond to the released 5-HT, providing a means by which the vagal afferent signal can be amplified or prolonged.


Subject(s)
Glucose/metabolism , Neurons, Afferent/metabolism , Protein Transport/physiology , Receptors, Serotonin, 5-HT3/metabolism , Vagus Nerve/metabolism , Animals , Female , Immunohistochemistry , Male , Microscopy, Confocal , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Visceral Afferents/metabolism
7.
Neurogastroenterol Motil ; 24(3): e136-46, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22188490

ABSTRACT

BACKGROUND: Oxytocin (OXT) has been implicated in reproduction and social interactions and in the control of digestion and blood pressure. OXT-immunoreactive axons occur in the dorsal vagal complex (DVC; nucleus tractus solitarius, NTS, dorsal motor nucleus of the vagus, DMV, and area postrema, AP), which contains neurons that regulate autonomic homeostasis. The aim of the present work is to provide a systematic investigation of the OXT-immunoreactive innervation of dorsal motor nucleus of the vagus (DMV) neurons involved in the control of gastrointestinal (GI) function. METHODS: We studied DMV neurons identified by (i) prior injection of retrograde tracers in the stomach, ileum, or cervical vagus or (ii) induction of c-fos expression by glucoprivation with 2-deoxyglucose. Another subgroup of DMV neurons was identified electrophysiologically by stimulation of the cervical vagus and then juxtacellularly labeled with biotinamide. We used two- or three-color immunoperoxidase labeling for studies at the light microscopic level. KEY RESULTS: Close appositions from OXT-immunoreactive varicosities were found on the cell bodies, dendrites, and axons of DMV neurons that projected to the GI tract and that responded to 2-deoxyglucose and juxtacellularly labeled DMV neurons. Double staining for OXT and choline acetyltransferase revealed that OXT innervation was heavier in the caudal and lateral DMV than in other regions. OXT-immunoreactive varicosities also closely apposed a small subset of tyrosine hydroxylase-immunoreactive NTS and DMV neurons. CONCLUSIONS & INFERENCES: Our results provide the first anatomical evidence for direct OXT-immunoreactive innervation of GI-related neurons in the DMV.


Subject(s)
Area Postrema/cytology , Gastrointestinal Tract/innervation , Neurons/metabolism , Oxytocin/metabolism , Solitary Nucleus/cytology , Vagus Nerve/cytology , Animals , Area Postrema/metabolism , Deoxyglucose/metabolism , Female , Humans , Immunohistochemistry , Male , Neurons/cytology , Rats , Rats, Sprague-Dawley , Solitary Nucleus/metabolism , Vagus Nerve/metabolism
8.
Neurogastroenterol Motil ; 23(2): e69-79, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20950355

ABSTRACT

BACKGROUND: We have shown recently that our model of experimental high-thoracic spinal cord injury (T3-SCI) mirrors the gastrointestinal clinical presentation of neurotrauma patients, whereby T3-SCI animals show diminished gastric emptying and dysmotility. In this study we used cholecystokinin as a model peptide to test the hypothesis that the T3-SCI induced gastroparesis is due, in part, to an impaired vagally-mediated response to gastrointestinal peptides. METHODS: We measured the responses to sulfated cholecystokinin (CCK-8s) in control and T3-SCI (3 or 21 days after injury) rats utilizing: (i) c-fos expression in the nucleus tractus solitarius (NTS) following peripherally administered CCK-8s; (ii) in vivo gastric tone and motility following unilateral microinjection of CCK-8s into the dorsal vagal complex (DVC); and (iii) whole cell recordings of glutamatergic synaptic inputs to NTS neurons. KEY RESULTS: Our results show that: (i) medullary c-fos expression in response to peripheral CCK-8s was significantly lower in T3-SCI rats 3 days after the injury, but recovered to control values at 3 weeks post-SCI, (ii) Unilateral microinjection of CCK-8s in the DVC induced a profound gastric relaxation in control animals, but did not induce any response in T3-SCI rats at both 3 and 21 days after SCI, (iii) Perfusion with CCK-8s increased glutamatergic currents in 55% of NTS neurons from control rats, but failed to induce any response in NTS neurons from T3-SCI rats. CONCLUSIONS & INFERENCES: Our data indicate alterations of vagal responses to CCK-8s in T3-SCI rats that may reflect a generalized impairment of gastric vagal neurocircuitry, leading to a reduction of gastric functions after SCI.


Subject(s)
Cholecystokinin/pharmacology , Peptide Fragments/pharmacology , Spinal Cord Injuries/physiopathology , Stomach/drug effects , Stomach/physiopathology , Vagus Nerve/physiopathology , Animals , Cholecystokinin/administration & dosage , Excitatory Postsynaptic Potentials/physiology , Gastroparesis/etiology , Gastroparesis/physiopathology , Injections, Intraperitoneal , Male , Models, Animal , Patch-Clamp Techniques , Peptide Fragments/administration & dosage , Rats , Rats, Wistar , Solitary Nucleus/physiopathology , Spinal Cord Injuries/complications , Thoracic Vertebrae/injuries
9.
Neurogastroenterol Motil ; 22(11): 1154-63, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20804520

ABSTRACT

BACKGROUND: Sensory information from the viscera, including the gastrointestinal (GI) tract, is transmitted through the afferent vagus via a glutamatergic synapse to neurons of the nucleus tractus solitarius (NTS), which integrate this sensory information to regulate autonomic functions and homeostasis. The integrated response is conveyed to, amongst other nuclei, the preganglionic neurons of the dorsal motor nucleus of the vagus (DMV) using mainly GABA, glutamate and catecholamines as neurotransmitters. Despite being modulated by almost all the neurotransmitters tested so far, the glutamatergic synapse between NTS and DMV does not appear to be tonically active in the control of gastric motility and tone. Conversely, tonic inhibitory GABAergic neurotransmission from the NTS to the DMV appears critical in setting gastric tone and motility, yet, under basal conditions, this synapse appears resistant to modulation. PURPOSE: Here, we review the available evidence suggesting that vagal efferent output to the GI tract is regulated, perhaps even controlled, in an 'on-demand' and efficient manner in response to ever-changing homeostatic conditions. The focus of this review is on the plasticity induced by variations in the levels of second messengers in the brainstem neurons that form vago-vagal reflex circuits. Emphasis is placed upon the modulation of GABAergic transmission to DMV neurons and the modulation of afferent input from the GI tract by neurohormones/neurotransmitters and macronutrients. Derangement of this 'on-demand' organization of brainstem vagal circuits may be one of the factors underlying the pathophysiological changes observed in functional dyspepsia or hyperglycemic gastroparesis.


Subject(s)
Brain Stem/physiology , Neural Pathways/physiology , Neuronal Plasticity/physiology , Stomach/innervation , Stomach/physiology , Vagus Nerve/physiology , Animals , Cyclic AMP/physiology , Dyspepsia/physiopathology , Gastrointestinal Tract/innervation , Humans , Neurons, Afferent/physiology , Solitary Nucleus/physiology , Synapses/physiology , gamma-Aminobutyric Acid/physiology
10.
Neurogastroenterol Motil ; 22(4): 361-5, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20377791

ABSTRACT

Protease-activated receptors (PARs) are members of a subfamily of G-protein-coupled receptors that regulate diverse cell functions in response to proteolytic cleavage of an anchored peptide domain that acts as a 'tethered' receptor-activating ligand. PAR-1 and PAR-2 in particular are present throughout the gastrointestinal (GI) tract and play prominent roles in the regulation of GI epithelial function, motility, inflammation and nociception. In a recent article in Neurogastroenterology and Motility, Wang et al. demonstrate, for the first time, that PAR-1 and PAR-2 are present on preganglionic parasympathetic neurons within the rat brainstem. As in other cellular systems, proteases such as thrombin and trypsin activate PAR-1 and PAR-2 on neurons of the dorsal motor nucleus of the vagus (DMV), leading to an increase in intracellular calcium levels via signal transduction mechanisms involving activation of phospholipase C and inositol triphosphate (IP3). The authors also report that the level of PAR-1 and PAR-2 transcripts in DMV tissue is increased following experimental colitis, suggesting that inflammatory conditions may modulate neuronal behavior or induce plasticity within central vagal neurocircuits. It seems reasonable to hypothesize, therefore, that the activity and behavior of vagal efferent motoneurons may be modulated directly by local and/or systemic proteases released during inflammation. This, in turn, may contribute to the increased incidence of functional GI disorders, including gastric dysmotility, delayed emptying and gastritis observed in patients with inflammatory bowel diseases.


Subject(s)
Brain Stem/physiology , Gastrointestinal Tract/physiology , Receptors, Proteinase-Activated/physiology , Animals , Autonomic Fibers, Preganglionic/physiology , Gastrointestinal Motility/physiology , Neurons/physiology , Rats
11.
Neurogastroenterol Motil ; 21(12): 1309-e126, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19622099

ABSTRACT

Pancreatic polypeptides such as neuropeptide Y (NPY) and peptide YY (PYY) exert profound, vagally mediated effects on gastrointestinal (GI) motility. Vagal efferent outflow to the GI tract is determined principally by tonic GABAergic synaptic inputs onto dorsal motor nucleus of the vagus (DMV) neurons, yet neither peptide modulates GABAergic transmission. We showed recently that opioid peptides appear similarly ineffective because of the low resting cAMP levels. Using whole cell recordings from identified DMV neurons, we aimed to correlate the influence of brainstem cAMP levels with the ability of pancreatic polypeptides to modulate GABAergic synaptic transmission. Neither NPY, PYY, nor the Y1 or Y2 receptor selective agonists [Leu,Pro]NPY or NPY(3-36) respectively, inhibited evoked inhibitory postsynaptic current (eIPSC) amplitude unless cAMP levels were elevated by forskolin or 8-bromo-cAMP, by exposure to adenylate cyclase-coupled modulators such as cholecystokinin octapeptide (sulfated) (CCK-8s) or thyrotropin releasing hormone (TRH), or by vagal deafferentation. The inhibition of eIPSC amplitude by [Leu,Pro]NPY or NPY(3-36) was stable for approximately 30 min following the initial increase in cAMP levels. Thereafter, the inhibition declined gradually until the agonists were again ineffective after 60 min. Analysis of spontaneous and miniature currents revealed that such inhibitory effects were due to actions at presynaptic Y1 and Y2 receptors. These results suggest that, similar to opioid peptides, the effects of pancreatic polypeptides on GABAergic transmission depend upon the levels of cAMP within gastric inhibitory vagal circuits.


Subject(s)
Brain Stem/physiology , Cyclic AMP/metabolism , Cyclic AMP/physiology , Neuropeptide Y/pharmacology , Peptide YY/pharmacology , Synaptic Transmission/drug effects , Vagus Nerve/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Denervation , Electric Stimulation , Electrophysiology , Nerve Net/drug effects , Neurons, Afferent/drug effects , Rats , Receptors, Neuropeptide Y/drug effects , Stomach/innervation , Synaptic Potentials/drug effects
12.
Am J Physiol Regul Integr Comp Physiol ; 292(3): R1092-100, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17122331

ABSTRACT

We have shown recently that cholecystokinin octapeptide (CCK-8s) increases glutamate release from nerve terminals onto neurons of the nucleus tractus solitarius pars centralis (cNTS). The effects of CCK on gastrointestinal-related functions have, however, been attributed almost exclusively to its paracrine action on vagal afferent fibers. Because it has been reported that systemic or perivagal capsaicin pretreatment abolishes the effects of CCK, the aim of the present work was to investigate the response of cNTS neurons to CCK-8s in vagally deafferented rats. In surgically deafferented rats, intraperitoneal administration of 1 or 3 mug/kg CCK-8s increased c-Fos expression in cNTS neurons (139 and 251% of control, respectively), suggesting that CCK-8s' effects are partially independent of vagal afferent fibers. Using whole cell patch-clamp techniques in thin brain stem slices, we observed that CCK-8s increased the frequency of spontaneous and miniature excitatory postsynaptic currents in 43% of the cNTS neurons via a presynaptic mechanism. In slices from deafferented rats, the percentage of cNTS neurons receiving glutamatergic inputs responding to CCK-8s decreased by approximately 50%, further suggesting that central terminals of vagal afferent fibers are not the sole site for the action of CCK-8s in the brain stem. Taken together, our data suggest that the sites of action of CCK-8s include the brain stem, and in cNTS, the actions of CCK-8s are not restricted to vagal central terminals but that nonvagal synapses are also involved.


Subject(s)
Neurons/drug effects , Sincalide/pharmacology , Solitary Nucleus/drug effects , Vagus Nerve/surgery , Analgesics, Non-Narcotic/pharmacology , Animals , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/physiology , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Female , Injections, Intraperitoneal , Male , Neurons/metabolism , Patch-Clamp Techniques/methods , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Sincalide/administration & dosage , Solitary Nucleus/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Vagus Nerve/drug effects
13.
Neuroscience ; 135(2): 611-25, 2005.
Article in English | MEDLINE | ID: mdl-16111819

ABSTRACT

Neural signaling by melanin-concentrating hormone and its receptor (SLC-1) has been implicated in the control of energy balance, but due to the wide distribution of melanin-concentrating hormone-containing fibers throughout the neuraxis, its critical sites of action for a particular effect have not been identified. The present study aimed to anatomically and functionally characterize melanin-concentrating hormone innervation of the rat caudal brainstem, as this brain area plays an important role in the neural control of ingestive behavior and autonomic outflow. Using retrograde tracing we demonstrate that a significant proportion (5-15%) of primarily perifornical and far-lateral hypothalamic melanin-concentrating hormone neurons projects to the dorsal vagal complex. In the caudal brainstem, melanin-concentrating hormone-ir axon profiles are distributed densely in most areas including the nucleus of the solitary tract, dorsal motor nucleus of the vagus, and sympathetic premotor areas in the ventral medulla. Close anatomical appositions can be demonstrated between melanin-concentrating hormone-ir axon profiles and tyrosine hydroxylase, GABA, GLP-1, NOS-expressing, and nucleus of the solitary tract neurons activated by gastric nutrient infusion. In medulla slice preparations, bath application of melanin-concentrating hormone inhibited in a concentration-dependent manner the amplitude of excitatory postsynaptic currents evoked by solitary tract stimulation via a pre-synaptic mechanism. Fourth ventricular administration of melanin-concentrating hormone (10 microg) in freely moving rats decreased core body temperature but did not change locomotor activity and food and water intake. We conclude that the rich hypothalamo-medullary melanin-concentrating hormone projections in the rat are mainly inhibitory to nucleus of the solitary tract neurons, but are not involved in the control of food intake. Projections to ventral medullary sites may play a role in the inhibitory effect of melanin-concentrating hormone on energy expenditure.


Subject(s)
Brain Stem/cytology , Energy Metabolism , Feeding Behavior/physiology , Hypothalamic Hormones/metabolism , Melanins/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism , Animals , Autonomic Nervous System/physiology , Axons/metabolism , Behavior, Animal , Body Temperature/drug effects , Body Temperature/physiology , Brain Stem/metabolism , Cell Count/methods , Cholera Toxin/metabolism , Dose-Response Relationship, Drug , Eating/drug effects , Eating/physiology , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Feeding Behavior/drug effects , Glucagon-Like Peptide 1/metabolism , Hypothalamic Hormones/pharmacology , Immunohistochemistry/methods , In Vitro Techniques , Male , Melanins/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Motor Activity/drug effects , Motor Activity/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Inhibition/radiation effects , Neurons/cytology , Neurons/parasitology , Nitric Oxide Synthase/metabolism , Patch-Clamp Techniques/methods , Pituitary Hormones/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/metabolism
14.
J Physiol ; 536(Pt 1): 141-52, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11579164

ABSTRACT

1. To examine the effects of glucose on the central components of the vago-vagal reflex control of gastric function, we performed both in vivo and in vitro experiments on neurones in the medial nucleus of the tractus solitarius (mNTS) and in the dorsal motor nucleus of the vagus (DMV). 2. In the in vivo anaesthetized rat preparation, unilateral microinjection of D-glucose (10 or 50 mM (60 nl)(-1)) in mNTS produced inhibition of gastric motility and an increase in intragastric pressure. D-glucose had no effect in the DMV. 3. In the in vitro rat brainstem slice preparation, whole-cell recordings of DMV neurones showed that increasing the glucose concentration of the perfusion solution from 5 mM to 15 or 30 mM produced outward currents of 35 +/- 5 pA (n = 7) and 51 +/- 10 pA (n = 11), respectively. These were blocked by tetrodotoxin and picrotoxin, indicating that glucose was acting indirectly to cause the release of GABA. Decreasing the glucose concentration of the perfusing solution by one-half produced an inward current of 36 +/- 5 pA (n = 7). 4. Stimulation of the NTS evoked inhibitory postsynaptic currents (IPSCs) in DMV neurones. The amplitude of the evoked IPSCs was positively correlated with glucose concentration. Perfusion with the ATP-sensitive K(+) (K(ATP)) channel opener diazoxide mimicked the effect of reduced glucose, while perfusion with the K(ATP) channel blocker glibenclamide mimicked the effects of increased glucose. 5. Our data indicate that glucose had no direct excitatory effect on DMV neurones, but DMV neurones appear to be affected by an action of glucose on cell bodies of mNTS neurones via effects on an ATP-sensitive potassium channel.


Subject(s)
Gastric Emptying/drug effects , Glucose/pharmacology , Solitary Nucleus/physiology , Vagus Nerve/physiology , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Extracellular Space/metabolism , Gastric Emptying/physiology , Male , Microinjections , Neurons/drug effects , Neurons/physiology , Organ Culture Techniques , Patch-Clamp Techniques , Pressure , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Rats , Rats, Sprague-Dawley , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Stomach/innervation , Stomach/physiology , Vagotomy , Vagus Nerve/cytology , Vagus Nerve/surgery
15.
Am J Physiol Gastrointest Liver Physiol ; 280(6): G1106-13, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352803

ABSTRACT

Using whole cell patch-clamp recordings, we investigated the effects of the GABA(B) receptor agonist baclofen in thin slices of rat brain stem containing identified gastric- or intestinal-projecting dorsal motor nucleus of the vagus (DMV) neurons. Perfusion with baclofen (0.1-100 microM) induced a concentration-dependent outward current (EC(50), 3 microM) in 54% of DMV neurons with no apparent differences between gastric- and intestinal-projecting neurons. The outward current was attenuated by pretreatment with the selective GABA(B) antagonists saclofen and 2-hydroxysaclofen, but not by the synaptic blocker TTX, indicating a direct effect at GABA(B) receptors on DMV neurons. Using the selective ion channel blockers barium, nifedipine, and apamin, we showed that the outward current was due to effects on potassium and calcium currents as well as calcium-dependent potassium currents. The calcium-mediated components of the outward current were more prominent in intestinal-projecting neurons than in gastric-projecting neurons. These data indicate that although baclofen inhibits both intestinal- and gastric-projecting neurons in the rat DMV, its mechanism of action differs among the neuronal subpopulations.


Subject(s)
Baclofen/pharmacology , Brain Stem/drug effects , Brain Stem/physiology , GABA Agonists/pharmacology , Vagus Nerve/physiology , Animals , Brain Stem/cytology , Electric Conductivity , Electrophysiology , Female , In Vitro Techniques , Ion Channels/drug effects , Ion Channels/physiology , Male , Neurons/drug effects , Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/drug effects
16.
J Physiol ; 531(Pt 2): 425-35, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11230515

ABSTRACT

It is well recognized that brainstem microinjections of 5-hydroxytryptamine (serotonin, 5-HT) and thyrotropin-releasing hormone (TRH) act synergistically to stimulate gastric function in vivo. Previous in vitro experiments have shown that this synergism does not occur at the level of the dorsal motor nucleus of the vagus (DMV) motoneurone. In order to determine the mechanism of this action, whole cell patch clamp recordings were made from identified gastric-projecting rat DMV neurones to investigate the effects of 5-HT and TRH on GABAergic inhibitory postsynaptic currents (IPSCs) evoked by stimulation of the nucleus of the tractus solitarius (NTS). 5-HT (30 microM) decreased IPSC amplitude by 26 +/- 2.5% in approximately 43% of DMV neurones. In the remaining neurones in which 5-HT had no effect on IPSC amplitude, exposure to TRH (1 microM) uncovered the ability of subsequent applications of 5-HT to decrease IPSC amplitude by 28 +/- 3%. Such TRH-induced 5-HT responses were prevented by the 5-HT1A antagonist NAN-190 (1 microM) and mimicked by the 5-HT1A agonist 8-OH-DPAT (1 microM). Increasing cAMP levels using the phosphodiesterase inhibitor isobutylmethylxanthine (IBMX; 10 microM), the non-hydrolysable cAMP analogue 8-bromo-cAMP (1 mM), or the adenylate cyclase activator forskolin (10 microM), like TRH, uncovered the ability of 5-HT to decrease evoked IPSC amplitude (17 +/- 2.2 %, 28.5 +/- 5.3 % and 30 +/- 4.8%, respectively), in neurones previously unresponsive to 5-HT. Conversely, the adenylate cyclase inhibitor, dideoxyadenosine (10 microM) and the protein kinase A inhibitor, Rp-cAMP (10 microM), blocked the ability of TRH to uncover the presynaptic inhibitory actions of 5-HT. These results suggest that activation of presynaptic TRH receptors initiates an intracellular signalling cascade that raises the levels of cAMP sufficient to uncover previously silent 5-HT1A receptors on presynaptic nerve terminals within the dorsal vagal complex.


Subject(s)
Presynaptic Terminals/metabolism , Receptors, Serotonin/metabolism , Second Messenger Systems/physiology , Solitary Nucleus/physiology , Thyrotropin-Releasing Hormone/pharmacology , Vagus Nerve/physiology , Action Potentials/physiology , Animals , Cyclic AMP/metabolism , Female , Male , Neural Inhibition/physiology , Rats , Rats, Sprague-Dawley , Receptors, Serotonin/drug effects , Receptors, Serotonin, 5-HT1 , Serotonin/pharmacology , Synaptic Transmission/physiology
17.
Am J Physiol Gastrointest Liver Physiol ; 280(3): G361-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171618

ABSTRACT

Nitric oxide synthase-immunoreactive (NOS-IR) neurons in the rat caudal dorsal motor nucleus of the vagus (DMV) project selectively to the gastric fundus and may be involved in vagal reflexes controlling gastric distension. This study aimed to identify the gastric projections of tyrosine hydroxylase-immunoreactive (TH-IR) DMV neurons, whether such neurons colocalize NOS-IR, and if they are activated after esophageal distension. Gastric-projecting neurons were identified after injection of retrograde tracers into the muscle wall of the gastric fundus, corpus, or antrum/pylorus before removal and processing of the brain stems for TH- and NOS-IR. A significantly higher proportion of corpus- compared with fundus- and antrum/pylorus-projecting neurons were TH-IR (14% compared with 4% and 2%, respectively, P < 0.05). Colocalization of NOS- and TH-IR was never observed in gastric-projecting neurons. In rats tested for c-Fos activation after intermittent esophageal balloon distension, no colocalization with TH-IR was observed in DMV neurons. These findings suggest that TH-IR neurons in the caudal DMV project mainly to the gastric corpus, constitute a subpopulation distinct from that of nitrergic vagal neurons, and are not activated on esophageal distension.


Subject(s)
Brain Stem/cytology , Neurons/cytology , Stomach/innervation , Vagus Nerve/cytology , Animals , Brain Stem/metabolism , Catecholamines , Esophagus/innervation , Esophagus/physiology , Female , Gastric Fundus/innervation , Male , Neurons/classification , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Pyloric Antrum/innervation , Pylorus/innervation , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism , Vagus Nerve/metabolism
18.
Neurogastroenterol Motil ; 12(1): 33-41, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10652114

ABSTRACT

Intracellular recordings were made from myenteric neurones of the guinea-pig descending colon. Neuropeptide Y (NPY) and related pancreatic polypeptides were applied by superfusion and the effects upon the amplitude of fast excitatory synaptic potentials (ESPs) and the ratio of paired fast ESPs evoked by stimulation of internodal fibre tracts were noted. NPY produced a concentration-dependent inhibition in fast ESP amplitude in the majority of neurones (17/21) with a calculated IC50 value of 7 nM; in some neurones this inhibition was mediated via the local release of noradrenaline. Peptide YY (PYY) (eight out of 11 neurones; IC50 = 1 nM), NPY(3-36) (three out of three neurones) and [Leu31, Pro34]NPY (four out of five neurones) also decreased the amplitude of fast ESPs. The effects of two or more pancreatic polypeptides or analogues on fast synaptic transmission were compared directly in six neurones; the apparent relative potency of agonists suggested the involvement of Y2-receptors and at least one other Y-receptor type. In the absence of any direct postsynaptic effects of pancreatic polypeptides on the active or passive properties of myenteric neurones, or on their sensitivity to ionophoretically applied acetylcholine, inhibition of fast ganglionic transmission was presumed to be presynaptic in origin. It is concluded that, in addition to their previously described depressant actions on neuro-effector transmission to colonic smooth muscle, pancreatic polypeptides can exert powerful inhibitory effects on myenteric neurones of the descending colon.


Subject(s)
Colon/innervation , Ganglia, Sympathetic/drug effects , Myenteric Plexus/drug effects , Neural Inhibition/drug effects , Neuropeptide Y/pharmacology , Peptide Fragments/pharmacology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Clonidine/pharmacology , Colon/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Ganglia, Sympathetic/physiology , Guinea Pigs , Idazoxan/pharmacology , Male , Motor Neurons/drug effects , Motor Neurons/physiology , Myenteric Plexus/physiology , Neuropeptide Y/analogs & derivatives , Norepinephrine/pharmacology , Pancreatic Polypeptide/pharmacology , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Propranolol/pharmacology
19.
Br J Pharmacol ; 128(6): 1307-15, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10578146

ABSTRACT

1 Whole cell patch clamp techniques were used on thin brainstem slices to investigate the effects of 5-hydroxytryptamine (5-HT) on gastrointestinal-projecting dorsal motor nucleus of the vagus (DMV) neurones. Neurones were identified as projecting to the stomach (n=122) or intestine (n=84) if they contained the fluorescent tracer Dil after it had been applied to the gastric fundus, corpus or antrum/pylorus or to the duodenum or caecum. 2 A higher proportion of intestinal neurones (69%) than gastric neurones (47%) responded to 5-HT with a concentration-dependent inward current which was antagonized fully by the 5-HT2A receptor antagonist ketanserin (1 microM). 3 Stimulation of the nucleus tractus solitarius (NTS) induced inhibitory synaptic currents that were reduced in amplitude by application of the 5-HT1A receptor agonist 8-OHDPAT (1 microM) or the 5-HT1A/1B receptor agonist TFMPP (1 microM) in 61% and 52% of gastric- and intestinal-projecting neurones, respectively. 5-HT also significantly reduced the frequency but not the amplitude of spontaneous inhibitory currents. 4 These data show that 5-HT excites directly a larger proportion of intestinal projecting neurones than gastric-projecting neurones, as well as inhibiting synaptic transmission from the NTS to the DMV. These data imply that the response to DMV neurones to 5-HT may be determined and classified by their specific projections.


Subject(s)
Motor Neurons/drug effects , Serotonin/pharmacology , Solitary Nucleus/drug effects , Vagus Nerve/drug effects , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Cesium/pharmacology , Chlorides/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Female , In Vitro Techniques , Indoles/pharmacology , Intestines/innervation , Ketanserin/pharmacology , Ligands , Male , Membrane Potentials/drug effects , Motor Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1B , Receptors, Serotonin/drug effects , Receptors, Serotonin, 5-HT1 , Receptors, Serotonin, 5-HT3 , Serotonin/analogs & derivatives , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Stomach/innervation , Synaptic Transmission/drug effects , Tropisetron , Vagus Nerve/physiology
20.
J Comp Neurol ; 410(4): 515-30, 1999 Aug 09.
Article in English | MEDLINE | ID: mdl-10398045

ABSTRACT

Region-specific patterns of nerves with immunoreactivity to neuropeptide Y (NPY) have been described previously in the submucous plexus of guinea pig large intestine. Because these may have functional significance, the possibility of similar, characteristic variations of NPY-like immunoreactivity (NPY-ir) in the myenteric plexus was explored. Regional differences were found in the occurrence and pattern of distribution of NPY-ir in the myenteric plexus of the guinea pig large intestine. NPY-ir was present rarely within neuron somata in any region of the large intestine, and NPY-ir nerve fibers were present only within the distal large intestine, increasing progressively in density from the distal spiral to the rectum. Lesion of the colonic nerves, but not the hypogastric, intermesenteric, or lumbar splanchnic nerves, resulted in a loss of NPY-ir in the distal spiral and transverse colon but not in the descending colon or rectum. Ring myotomies in the descending colon resulted in a loss of NPY-ir proximal to the lesion. Dual-labeling immunohistochemical studies revealed that the NPY-ir nerve fibers rarely contained immunoreactivity for tyrosine hydroxylase (TH). Extrinsic nerve lesions resulted in an unequivocal reduction in NPY-ir in intraganglionic fibers of the submucosal plexuses of the transverse colon and a partial loss in the distal spiral and descending colon: the rectum was unaffected; in only a minority of guinea pigs, however, was any decrease in the NPY-ir innervation of submucosal blood vessels detected. The principal projections of NPY-ir nerves were from and through the inferior mesenteric ganglion; however, NPY-ir was not colocalized with TH-ir. It is proposed that nonnoradrenergic, NPY-containing neurons located in the inferior mesenteric ganglion project through the colonic nerves and that these proximally directed fibers innervate the transverse colon and the distal spiral. Nonnoradrenergic, NPY-ir neurons lying in the pelvic ganglia or sacral sympathetic chain may make an important contribution to the innervation of the myenteric plexus of the rectum and the descending colon.


Subject(s)
Guinea Pigs/anatomy & histology , Intestine, Large/innervation , Neuropeptide Y/metabolism , Sympathetic Nervous System/anatomy & histology , Sympathetic Nervous System/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Denervation , Immunohistochemistry , Male , Myenteric Plexus/metabolism , Submucous Plexus/metabolism , Tissue Distribution/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...