Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Neurosci ; 42(26): 5212-5228, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35610046

ABSTRACT

Activity in the dorsal vagal complex (DVC) is essential to gastric motility regulation. We and others have previously shown that this activity is greatly influenced by local GABAergic signaling, primarily because of somatostatin (SST)-expressing GABAergic neurons. To further understand the network dynamics associated with gastric motility control in the DVC, we focused on another neuron prominently distributed in this complex, neuropeptide-Y (NPY) neurons. However, the effect of these neurons on gastric motility remains unknown. Here, we investigate the anatomic and functional characteristics of the NPY neurons in the nucleus tractus solitarius (NTS) and their interactions with SST neurons using transgenic mice of both sexes. We sought to determine whether NPY neurons influence the activity of gastric-projecting neurons, synaptically interact with SST neurons, and affect end-organ function. Our results using combined neuroanatomy and optogenetic in vitro and in vivo show that NPY neurons are part of the gastric vagal circuit as they are trans-synaptically labeled by a viral tracer from the gastric antrum, are primarily excitatory as optogenetic activation of these neurons evoke EPSCs in gastric-antrum-projecting neurons, are functionally coupled to each other and reciprocally connected to SST neurons, whose stimulation has a potent inhibitory effect on the action potential firing of the NPY neurons, and affect gastric tone and motility as reflected by their robust optogenetic response in vivo. These findings indicate that interacting NPY and SST neurons are integral to the network that controls vagal transmission to the stomach.SIGNIFICANCE STATEMENT The brainstem neurons in the dorsal nuclear complex are essential for regulating vagus nerve activity that affects the stomach via tone and motility. Two distinct nonoverlapping populations of predominantly excitatory NPY neurons and predominantly inhibitory SST neurons form reciprocal connections with each other in the NTS and with premotor neurons in the dorsal motor nucleus of the vagus to control gastric mechanics. Light activation and inhibition of NTS NPY neurons increased and decreased gastric motility, respectively, whereas both activation and inhibition of NTS SST neurons enhanced gastric motility.


Subject(s)
Brain Stem , Stomach , Animals , Brain Stem/physiology , Female , GABAergic Neurons/physiology , Male , Mice , Neuropeptide Y/pharmacology , Rats , Rats, Sprague-Dawley , Solitary Nucleus/physiology , Stomach/innervation , Vagus Nerve/physiology
2.
Cell Mol Neurobiol ; 42(2): 333-360, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33813668

ABSTRACT

This review is on how current knowledge of brainstem control of gastric mechanical function unfolded over nearly four decades from the perspective of our research group. It describes data from a multitude of different types of studies involving retrograde neuronal tracing, microinjection of drugs, whole-cell recordings from rodent brain slices, receptive relaxation reflex, accommodation reflex, c-Fos experiments, immunohistochemical methods, electron microscopy, transgenic mice, optogenetics, and GABAergic signaling. Data obtained indicate the following: (1) nucleus tractus solitarius (NTS)-dorsal motor nucleus of the vagus (DMV) noradrenergic connection is required for reflex control of the fundus; (2) second-order nitrergic neurons in the NTS are also required for reflex control of the fundus; (3) a NTS GABAergic connection is required for reflex control of the antrum; (4) a single DMV efferent pathway is involved in brainstem control of gastric mechanical function under most experimental conditions excluding the accommodation reflex. Dual-vagal effectors controlling cholinergic and non-adrenergic and non-cholinergic (NANC) input to the stomach may be part of the circuitry of this reflex. (5) GABAergic signaling within the NTS via Sst-GABA interneurons determine the basal (resting) state of gastric tone and phasic contractions. (6) For the vagal-vagal reflex to become operational, an endogenous opioid in the NTS is released and the activity of Sst-GABA interneurons is suppressed. From the data, we suggest that the CNS has the capacity to provide region-specific control over the proximal (fundus) and distal (antrum) stomach through engaging phenotypically different efferent inputs to the DMV.


Subject(s)
Brain Stem , Stomach , Animals , Brain Stem/physiology , Mice , Patch-Clamp Techniques , Solitary Nucleus , Vagus Nerve/physiology
3.
J Physiol ; 594(10): 2661-79, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26959279

ABSTRACT

KEY POINTS: The dorsal motor nucleus of the vagus (DMV) in the brainstem consists primarily of vagal preganglionic neurons that innervate postganglionic neurons of the upper gastrointestinal tract. The activity of the vagal preganglionic neurons is predominantly regulated by GABAergic transmission in the DMV. The present findings indicate that the overwhelming GABAergic drive present at the DMV is primarily from somatostatin positive GABA (Sst-GABA) DMV neurons. Activation of both melanocortin and µ-opioid receptors at the DMV inhibits Sst-GABA DMV neurons. Sst-GABA DMV neurons may serve as integrative targets for modulating vagal output activity to the stomach. ABSTRACT: We have previously shown that local GABA signalling in the brainstem is an important determinant of vagally-mediated gastric activity. However, the neural identity of this GABA source is currently unknown. To determine this, we focused on the somatostatin positive GABA (Sst-GABA) interneuron in the dorsal motor nucleus of the vagus (DMV), a nucleus that is intimately involved in regulating gastric activity. Also of particular interest was the effect of melanocortin and µ-opioid agonists on neural activity of Sst-GABA DMV neurons because their in vivo administration in the DMV mimics GABA blockade in the nucleus. Experiments were conducted in brain slice preparation of transgenic adult Sst-IRES-Cre mice expressing tdTomato fluorescence, channelrhodopsin-2, archaerhodopsin or GCaMP3. Electrophysiological recordings were obtained from Sst-GABA DMV neurons or DiI labelled gastric-antrum projecting DMV neurons. Our results show that optogenetic stimulation of Sst-GABA neurons results in a robust inhibition of action potentials of labelled premotor DMV neurons to the gastric-antrum through an increase in inhibitory post-synaptic currents. The activity of the Sst-GABA neurons in the DMV is inhibited by both melanocortin and µ-opioid agonists. These agonists counteract the pronounced inhibitory effect of Sst-GABA neurons on vagal pre-motor neurons in the DMV that control gastric motility. These observations demonstrate that Sst-GABA neurons in the brainstem are crucial for regulating the activity of gastric output neurons in the DMV. Additionally, they suggest that these neurons serve as targets for converging CNS signals to regulate parasympathetic gastric function.


Subject(s)
GABAergic Neurons/physiology , Optogenetics/methods , Parasympathetic Fibers, Postganglionic/physiology , Pyloric Antrum/innervation , Pyloric Antrum/physiology , Somatostatin/physiology , Animals , Brain Stem/drug effects , Brain Stem/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , GABAergic Neurons/drug effects , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Transgenic , Organ Culture Techniques , Parasympathetic Fibers, Postganglionic/drug effects , Pyloric Antrum/drug effects , Stomach/drug effects , Stomach/innervation , Stomach/physiology , alpha-MSH/pharmacology
4.
J Neurosci ; 33(33): 13286-99, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23946387

ABSTRACT

Activation of melanocortin 4 receptors (MC4-Rs) in brain nuclei associated with food intake profoundly influences consummatory behavior. Of these nuclei, the dorsal motor vagal nucleus (DMV), which has a dense concentration of MC4-Rs, is an important regulator of gastric tone and motility. Hence, the present study sought to examine the role of MC4-Rs in this nucleus on these activities. Using an in vivo approach, MC4-R agonists, melanotan-II (MT-II) or α-melanocyte stimulating hormone (α-MSH), were unilaterally microinjected into the DMV of rats, and their effects were noted on gastric activity. MT-II decreased phasic contractions, whereas α-MSH increased their amplitude. Both effects were blocked by the MC4-R antagonist SHU9119 or by ipsilateral vagotomy. Microinjection of the agonists (MT-II and α-MSH) into the overlying nucleus of the solitary tract (NTS), an important component of "vago-vagal" gastric circuitry, decreased phasic contractions. In addition, α-MSH reduced gastric tone and mean arterial blood pressure. To study the underlying mechanisms of the effect of MC4-R stimulation on gastric activity, electrophysiological recordings were made from labeled DMV antrum neurons in rat pups and MC4-R(-/-) mice. Bath application of MT-II or α-MSH significantly reduced spontaneous action potentials (but not in MC4-R(-/-) mice). However, in low-calcium ACSF, MT-II decreased neuronal firing, whereas α-MSH increased it. These effects mirror those of our in vivo DMV studies. Altogether, our novel findings show that activation of MC4-Rs in the brainstem, particularly in the medial NTS by the endogenous peptide α-MSH, modulates gastric activity, which may have physiological relevance for food intake and gastric function.


Subject(s)
Brain Stem/metabolism , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction/physiology , Stomach/innervation , Animals , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Vagus Nerve/physiology , alpha-MSH/metabolism , alpha-MSH/pharmacology
5.
J Comp Neurol ; 531(15): 1562-1581, 2023 10.
Article in English | MEDLINE | ID: mdl-37507853

ABSTRACT

The pyloric sphincter receives parasympathetic vagal innervation from the dorsal motor nucleus of the vagus (DMV). However, little is known about its higher-order neurons and the nuclei that engage the DMV neurons controlling the pylorus. The purpose of the present study was twofold. First, to identify neuroanatomical connections between higher-order neurons and the DMV. This was carried out by using the transneuronal pseudorabies virus PRV-152 injected into rat pylorus torus and examining the brains of these animals for PRV labeling. Second, to identify the specific sites within the DMV that functionally control the motility and tone of the pyloric sphincter. For these studies, experiments were performed to assess the effect of DMV stimulation on pylorus activity in urethane-anesthetized male rats. A strain gauge force transducer was sutured onto the pyloric tonus to monitor tone and motility. L-glutamate (500 pmol/30 nL) was microinjected unilaterally into the rostral and caudal areas of the DMV. Data from the first study indicated that neurons labeled with PRV occurred in the DMV, hindbrain raphe nuclei, midbrain Edinger-Westphal nucleus, ventral tegmental area, lateral habenula, and arcuate nucleus. Data from the second study indicated that microinjected L-glutamate into the rostral DMV results in contraction of the pylorus blocked by intravenously administered atropine and ipsilateral vagotomy. L-glutamate injected into the caudal DMV relaxed the pylorus. This response was abolished by ipsilateral vagotomy but not by intravenously administered atropine or L-NG-nitroarginine methyl ester (L-NAME). These findings identify the anatomical and functional brain neurocircuitry involved in controlling the pyloric sphincter. Our results also show that site-specific stimulation of the DMV can differentially influence the activity of the pyloric sphincter by separate vagal nerve pathways.


Subject(s)
Glutamic Acid , Pylorus , Rats , Male , Animals , Pylorus/innervation , Vagus Nerve/physiology , Medulla Oblongata/physiology , Atropine/pharmacology
6.
J Neurophysiol ; 107(3): 1022-31, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22114164

ABSTRACT

Our laboratory previously reported that gastric activity is controlled by a robust GABA(A) receptor-mediated inhibition in the medial nucleus of the tractus solitarius (mNTS) (Herman et al. 2009), and that µ-opioid receptor activation inhibits gastric tone by suppression of this GABA signaling (Herman et al. 2010). These data raised two questions: 1) whether any of this inhibition was due to tonic GABA(A) receptor-mediated conductance in the mNTS; and 2) whether µ-opioid receptor activation suppressed both tonic and phasic GABA signaling. In whole cell recordings from rat mNTS neurons, application of three GABA(A) receptor antagonists (gabazine, bicuculline, and picrotoxin) produced a persistent reduction in holding current and decrease in population variance or root mean square (RMS) noise, suggesting a blockade of tonic GABA signaling. Application of gabazine at a lower concentration abolished phasic currents, but had no effect on tonic currents or RMS noise. Application of the δ-subunit preferring agonist gaboxadol (THIP) produced a dose-dependent persistent increase in holding current and RMS noise. Pretreatment with tetrodotoxin prevented the action of gabazine, but had no effect on the THIP-induced current. Membrane excitability was unaffected by the selective blockade of phasic inhibition, but was increased by blockade of both phasic and tonic currents. In contrast, activation of tonic currents decreased membrane excitability. Application of the µ-opioid receptor agonist DAMGO produced a persistent reduction in holding current that was not observed following pretreatment with a GABA(A) receptor antagonist and was not evident in mice lacking the δ-subunit. These data suggest that mNTS neurons possess a robust tonic inhibition that is mediated by GABA(A) receptors containing the δ-subunit, that determines membrane excitability, and that is partially regulated by µ-opioid receptors.


Subject(s)
Receptors, GABA-A/physiology , Receptors, Opioid, mu/physiology , Solitary Nucleus/physiology , Analgesics, Opioid/pharmacology , Animals , Bicuculline/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Isoxazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Patch-Clamp Techniques , Picrotoxin/pharmacology , Pyridazines/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Solitary Nucleus/drug effects , Tetrodotoxin/pharmacology
7.
Front Neurosci ; 16: 961042, 2022.
Article in English | MEDLINE | ID: mdl-35983226

ABSTRACT

Background and aim: Local GABAergic signaling in the dorsal vagal complex (DVC) is essential to control gastric function. While the inhibitory GABAA receptor action on motility in the DVC is well-documented, the role of the GABAB receptor on gastric function is less well-established. Microinjection of baclofen, a selective GABAB receptor agonist, in the dorsal motor nucleus of the vagus (DMV) increases gastric tone and motility, while the effect on motility in the nucleus tractus solitarius (NTS) needs to be investigated. Previous in vitro studies showed that GABAB receptors exert a local inhibitory effect in unidentified NTS neurons. Since the NTS and DMV nuclei have differential control of gastric motility, we compared GABAB receptor activation in the NTS to that reported in the DMV. We microinjected baclofen unilaterally in the NTS while monitoring intragastric pressure and compared its action to optogenetic activation of somatostatin (SST) neurons in transgenic sst-Cre::channelrhodopsin-2 (ChR2) mice. We also performed patch-clamp recordings from SST and DMV neurons in brainstem slices from these mice. Methods: In vivo drug injections and optogenetic stimulation were performed in fasted urethane/α-chloralose anesthetized male mice. Gastric tone and motility were monitored by an intragastric balloon inserted in the antrum and inflated with warm water to provide a baseline intragastric pressure (IGP). Coronal brainstem slices were obtained from the sst-Cre::ChR2 mice for interrogation with optogenetics and pharmacology using electrophysiology. Results: The unilateral microinjection of baclofen into the NTS caused a robust increase in gastric tone and motility that was not affected by ipsilateral vagotomy. Optogenetic activation of SST neurons that followed baclofen effectively suppresses the gastric motility in vivo. In brain slices, baclofen suppressed spontaneous and light-activated inhibitory postsynaptic currents in SST and gastrointestinal-projection DMV neurons and produced outward currents. Conclusion: Our results show that GABAB receptors in the NTS strongly increase gastric tone and motility. Optogenetic stimulation in vivo and in vitro suggests that these receptors activated by baclofen suppress the glutamatergic sensory vagal afferents in the NTS and also inhibit the interneurons and the inhibitory neurons that project to the DMV, which, in turn, increase motility via a cholinergic excitatory pathway to the stomach.

8.
Am J Physiol Gastrointest Liver Physiol ; 299(2): G494-506, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20489046

ABSTRACT

We examined the effects of altering mu-opioid receptor (MOR) activity in the medial subnucleus of the tractus solitarius (mNTS) on several gastric end points including intragastric pressure (IGP), fundus tone, and the receptive relaxation reflex (RRR). Microinjection of the MOR agonist [d-Ala(2),MePhe(4),Gly(ol)(5)]enkephalin (DAMGO; 1-10 fmol) into the mNTS produced dose-dependent decreases in IGP. Microinjection of the endogenous MOR agonists endomorphin-1 and endomorphin-2 (20 fmol) into the mNTS mimicked the effects of 10 fmol DAMGO. Microinjection of 1 and 100 pmol DAMGO into the mNTS produced a triphasic response consisting of an initial decrease, a transient increase, and a persistent decrease in IGP. The increase in IGP appeared to be due to diffusion to the dorsal motor nucleus of the vagus. The effects of 10 fmol DAMGO in the mNTS were blocked by vagotomy and by blockade of MORs, GABA(A) receptors, and ionotropic glutamate receptors in the mNTS. The RRR response was abolished by bilateral microinjection of the opioid receptor antagonist naltrexone into the mNTS and reduced by intravenous administration of naltrexone. Our data demonstrate that 1) activation of MORs in the mNTS with femtomole doses of agonist inhibits gastric motility, 2) the mechanism of MOR effects in the mNTS is through suppression of local GABA activity, and 3) blockade of MORs in the mNTS prevents the RRR response. These data suggest that opioids play an important role in mediating a vagovagal reflex through release of an endogenous opioid in the mNTS, which, in turn, inhibits ongoing local GABA activity and allows vagal sensory input to excite second-order mNTS neurons.


Subject(s)
Gastrointestinal Motility/physiology , Receptors, Opioid/metabolism , Solitary Nucleus/metabolism , Stomach/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Down-Regulation , Efferent Pathways/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , GABA Antagonists/administration & dosage , GABA-A Receptor Antagonists , Gastric Fundus/drug effects , Gastric Fundus/physiology , Gastrointestinal Motility/radiation effects , Male , Microinjections , Muscle Tonus/drug effects , Naltrexone/administration & dosage , Narcotic Antagonists/administration & dosage , Neurotransmitter Agents/administration & dosage , Oligopeptides/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Stomach/drug effects , Vagus Nerve/drug effects , Vagus Nerve/physiology
9.
Neuropharmacology ; 166: 107921, 2020 04.
Article in English | MEDLINE | ID: mdl-31881170

ABSTRACT

Desensitization of the nicotinic acetylcholine receptor (nAChR) containing the ß2 subunit is a potentially critical mechanism underlying the body weight (BW) reducing effects of nicotine. The purpose of this study was a) to determine the α subunit(s) that partners with the ß2 subunit to form the nAChR subtype that endogenously regulates energy balance and b) to probe the extent to which nAChR desensitization could be involved in the regulation of BW. We demonstrate that deletion of either the α4 or the ß2, but not the α5, subunit of the nAChR suppresses weight gain in a sex-dependent manner. Furthermore, chronic treatment with the ß2-selective nAChR competitive antagonist dihydro-ß-erythroidine (DHßE) in mice fed a high-fat diet suppresses weight gain. These results indicate that heteromeric α4ß2 nAChRs play a role as intrinsic regulators of energy balance and that desensitizing or inhibiting this nAChR is likely a relevant mechanism and thus could be a strategy for weight loss.


Subject(s)
Body Weight/physiology , Dihydro-beta-Erythroidine/administration & dosage , Receptors, Nicotinic/metabolism , Animals , Body Weight/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Diet, High-Fat/adverse effects , Female , Infusion Pumps , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Nicotinic/deficiency , Weight Gain/drug effects , Weight Gain/physiology
10.
Front Neurosci ; 13: 967, 2019.
Article in English | MEDLINE | ID: mdl-31572117

ABSTRACT

Central nervous system regulation of the gastric tone and motility is primarily mediated via preganglionic neurons of the dorsal motor nucleus of the vagus (DMV). This is thought to occur by simultaneous engagement of both independent excitatory and inhibitory pathways from the DMV and has been proposed to underlie the opposing effects seen on gastric tone and motility in a number of in vivo models. Contrary to this view, we have been unable to find any evidence for this "dual effector" pathway. Since this possibility is so fundamental to how the brain-gut axis may interact in light of both peripheral and central demands, we decided to explore it further in two separate animal models previously used in conjunction with GABAB signaling to report the existence of a "dual effector" pathway. Using anesthetized rats or ferrets, we microinjected baclofen (7.5 pmol; n = 6), a GABAB agonist into the DMV of rats or intravenously administered it (0.5 mg/kg; n = 4) in ferrets. In rats, unilateral microinjection of baclofen into the DMV caused a robust dose-dependent increase in gastric tone and motility that was abolished by ipsilateral vagotomy and counteracted by pretreatment with atropine (0.1 mg/kg; IV). Similarly, as microinjection in the rats, IV administration of baclofen (0.5 mg/kg) in the ferrets induced its characteristic excitatory effects on gastric tone and motility, which were blocked by either pre- or post-treatment with atropine (0.1 mg/kg; IV). Altogether, our data provide evidence that the gastric musculature (other than the gastric sphincters) is regulated by a "single effector" DMV pathway using acetylcholine.

11.
Front Neurosci ; 12: 104, 2018.
Article in English | MEDLINE | ID: mdl-29545738

ABSTRACT

Background/Objectives: We tested the hypothesis that abolishing vagal nerve activity will reverse the obesity phenotype of melanocortin 4 receptor knockout mice (Mc4r-/-). Subjects/Methods: In two separate studies, we examined the efficacy of bilateral subdiaphragmatic vagotomy (SDV) with pyloroplasty in the prevention and treatment of obesity in Mc4r-/- mice. Results: In the first study, SDV prevented >20% increase in body weight (BW) associated with this genotype. This was correlated with a transient reduction in overall food intake (FI) in the preventative arm of the study. Initially, SDV mice had reduced weekly FI; however, FI normalized to that of controls and baseline FI within the 8-week study period. In the second study, the severe obesity that is characteristic of the adult Mc4r-/- genotype was significantly improved by SDV with a magnitude of 30% loss in excess BW over a 4-week period. Consistent with the first preventative study, within the treatment arm, SDV mice also demonstrated a transient reduction in FI relative to control and baseline levels that normalized over subsequent weeks. In addition to the accompanying loss in weight, mice subjected to SDV showed a decrease in respiratory exchange ratio (RER), and an increase in locomotor activity (LA). Analysis of the white fat-pad deposits of these mice showed that they were significantly less than the control groups. Conclusions: Altogether, our data demonstrates that SDV both prevents gain in BW and causes weight loss in severely obese Mc4r-/- mice. Moreover, it suggests that an important aspect of weight reduction for this type of monogenic obesity involves loss of signaling in vagal motor neurons.

12.
Auton Neurosci ; 136(1-2): 31-42, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17572158

ABSTRACT

We reported pharmacological data suggesting that stimulation of the vago-vagal reflex activates noradrenergic neurons in the hindbrain that inhibit dorsal motor nucleus of the vagus (DMV) neurons projecting to the fundus, but not to the antrum [Ferreira Jr., M., Sahibzada, N., Shi, M., Panico, W., Neidringhaus, M., Wasserman, A., Kellar, K.J., Verbalis, J., Gillis, R.A., 2002. CNS site of action and brainstem circuitry responsible for the intravenous effects of nicotine on gastric tone. J. Neurosci. 22, 2764-2779.]. The purpose of this study was to use an ultrastructural approach to test the hypothesis that noradrenergic terminals form synapses with DMV fundus-projecting neurons, but not with DMV antrum-projecting neurons. A retrograde tracer, CTbeta-HRP, was injected into the gastric smooth muscle of either the fundus or the antrum of rats. Animals were re-anesthetized 48 h later and perfusion-fixed with acrolein and paraformaldehyde. Brainstems were processed histochemically for CTbeta-HRP, and immunocytochemically for either DbetaH or PNMT by dual-labeling electron microscopic methods. Most cell bodies and dendrites of neurons that were retrogradely labeled from the stomach occurred at the level of the area postrema. Examination of 482 synapses on 238 neurons that projected to the fundus revealed that 17.4+/-2.7% (n=4) of synaptic contacts were with DbetaH-IR terminals. Of 165 fundus-projecting neurons, 4.4+/-1.5% (n=4) formed synaptic contacts with PNMT-IR terminals. In contrast, the examination of 384 synapses on 223 antrum-projecting neurons revealed no synaptic contact with DbetaH-IR terminals. These data provide proof that norepinephrine containing nerve terminals synapse with DMV fundus-projecting neurons but not with DMV antrum-projecting neurons. These data also suggest that brainstem circuitry controlling the fundus differs from circuitry controlling the antrum.


Subject(s)
Gastric Fundus/innervation , Norepinephrine/metabolism , Rhombencephalon/ultrastructure , Vagus Nerve/ultrastructure , Visceral Afferents/ultrastructure , Animals , Area Postrema/physiology , Area Postrema/ultrastructure , Autonomic Pathways/physiology , Autonomic Pathways/ultrastructure , Cell Communication/physiology , Cholera Toxin , Dendrites/physiology , Dendrites/ultrastructure , Dopamine beta-Hydroxylase/analysis , Dopamine beta-Hydroxylase/metabolism , Gastric Fundus/physiology , Horseradish Peroxidase , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Phenylethanolamine N-Methyltransferase/analysis , Phenylethanolamine N-Methyltransferase/metabolism , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Rhombencephalon/physiology , Sympathetic Nervous System/physiology , Sympathetic Nervous System/ultrastructure , Synaptic Transmission/physiology , Vagus Nerve/physiology , Visceral Afferents/physiology
13.
Neuropharmacology ; 110(Pt A): 165-174, 2016 11.
Article in English | MEDLINE | ID: mdl-27444741

ABSTRACT

Nicotine's effect on food intake and body weight has been well documented; however, the relevant receptors underlying these effects have not been firmly established. The purpose of the present study was to: (1) identify the nicotinic acetylcholine receptor (nAChR) subtype involved in food intake and body weight; (2) establish whether food intake and body weight reduction produced by nicotinic drugs are due to activation or desensitization of nAChRs; and, (3) assess the role of the melanocortin system in nicotinic drug effects on food intake and body weight. To identify the nAChR, we tested the effect of sazetidine-A (SAZ-A), a relatively selective ligand of ß2-containing nAChRs, on food intake and body weight in obese mice. SAZ-A (3 mg/kg; SC) administered twice-daily significantly decreased food intake and body weight. To assess whether these effects involved desensitization, SAZ-A was administered to non-obese mice via osmotic pump, which, due to its slow sustained drug delivery method, causes prolonged desensitization. SAZ-A via osmotic pump delivery significantly decreased the gain in body weight and reduced food intake. In contrast, body weight was unaffected by SAZ-A in ß2(-/-) mice or in mice lacking the melanocortin 4 receptor (MC4R). These results indicate that ß2 containing nAChRs are essential to SAZ-A's inhibitory effect on body weight and food intake and engage the melanocortin system.


Subject(s)
Azetidines/administration & dosage , Body Weight/physiology , Eating/physiology , Pyridines/administration & dosage , Receptors, Nicotinic/physiology , Animals , Body Weight/drug effects , Eating/drug effects , Infusion Pumps , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Nicotine/administration & dosage
14.
J Neurosci ; 23(10): 4182-9, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12764106

ABSTRACT

Contusion spinal cord injury (SCI) at T8 produces respiratory abnormalities in conscious rats breathing room air and challenged with CO2. In seeking ways to improve respiration after SCI, we tested drugs that stimulate serotonin 1A (5-HT1A) receptors, based on our previous findings that these agents can counteract respiratory depression produced by morphine overdose. Respiratory function was measured with a head-out plethysmograph system in conscious rats. T8 SCI rats (n = 5) showed decreased tidal volume (Vt; 0.90 +/- 0.02-0.66 +/- 0.03 ml; p < 0.05) and increased respiratory rate (f;91 +/- 3.7-132 +/- 5.7 breaths/min; p < 0.05) with room air ventilation at 24 hr after injury. They also exhibited a diminished response to the respiratory stimulating effect of 7% CO2; minute ventilation increased to 250 +/- 17 ml/min before, but only to 162 +/- 15 ml/min at 24 hr after SCI (p < 0.05). Respiratory deficits during room air ventilation were also observed at 7 d after injury (n = 3). Treatment with the 5-HT1A receptor agonist 8-hydroxy-2-(di-n-propylmino)tetralin (8-OH-DPAT; 250 microg/kg, i.p.) at 24 hr (n = 5) or 7 d (n = 3) after injury normalized Vt, f, and the respiratory response to 7% CO2. Identical results were obtained with another 5-HT1A receptor agonist, buspirone (1.5 mg/kg, i.p.; n = 3). In contrast, intraperitoneal saline vehicle administration (n = 5) showed no beneficial effects on SCI-impaired respiration. Finally, pretreatment with a specific antagonist of 5-HT1A receptors, 4-iodo-N-[2-[4-(methoxyphenyl)-1-piperazinyl]ethyl]-N-2-pyridinyl-benzamide (3 mg/kg, i.p.; n = 3) given 20 min before 8-OH-DPAT, prevented 8-OH-DPAT from restoring respiration to normal. Our results demonstrate that drugs that stimulate 5-HT1A receptors counteract respiratory abnormalities in conscious rats after SCI.


Subject(s)
Receptors, Serotonin/metabolism , Respiratory System Abnormalities/drug therapy , Respiratory System Abnormalities/etiology , Serotonin Receptor Agonists/pharmacology , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/therapeutic use , Aminopyridines/pharmacology , Animals , Buspirone/administration & dosage , Buspirone/pharmacology , Buspirone/therapeutic use , Female , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT1 , Respiration/drug effects , Respiratory Function Tests/instrumentation , Respiratory Function Tests/methods , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/administration & dosage , Serotonin Receptor Agonists/therapeutic use , Wounds, Nonpenetrating/complications
15.
J Neurosci ; 22(7): 2764-79, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11923442

ABSTRACT

The purposes of our study were to determine (1) the effects of intravenous (i.v.) nicotine on gastric mechanical function of anesthetized rats, (2) the CNS site of action of nicotine to produce these effects, (3) the CNS nicotinic acetylcholine receptor (nAChR) subtype(s) responsible for mediating the i.v. effects of nicotine, and (4) the brainstem neurocircuitry engaged by i.v. nicotine for eliciting its gastric effects. This was accomplished by monitoring intragastric pressure (gastric tone) and contractility of the fundus and antrum while administering five doses of i.v. nicotine and microinjecting nicotine into specific brainstem nuclei. Additionally, c-Fos expression in the brainstem after i.v. nicotine and pharmacological agents were used as tools to identify the CNS site and circuitry and reveal the nAChR subtype(s) mediating the gastric effects of nicotine. Using these experimental approaches, we found the following. (1) When given intravenously in doses of 56.5, 113, 226, 452, and 904 nmol/kg, nicotine elicited only inhibitory effects on gastric mechanical function. The most sensitive area of the stomach to nicotine was the fundus, and this effect was mediated by the vagus nerve at doses of 56.5, 113, and 226 nmol/kg. (2) The CNS site of action and nAChR subtype responsible were glutamatergic vagal afferent nerve terminals in the medial subnucleus of the tractus solitarious (mNTS) and alpha4beta2, respectively. (3) The brainstem neurocircuitry that was involved appeared to consist of a mNTS noradrenergic pathway projecting to the dorsal motor nucleus of the vagus (DMV). This pathway seems to be activated via nitriergic interneurons engaged by vagally released glutamate in the mNTS and results in alpha2 adrenergic receptor-mediated inhibition of DMV neurons projecting to the fundus and controlling gastric tone.


Subject(s)
Central Nervous System/drug effects , Neural Pathways/physiology , Nicotine/pharmacology , Stomach/drug effects , Stomach/physiology , Animals , Blood Pressure/drug effects , Brain Stem/drug effects , Brain Stem/metabolism , Central Nervous System/physiology , Dose-Response Relationship, Drug , Gastrointestinal Motility/drug effects , Injections, Intravenous , Male , Manometry , Microinjections , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Pathways/drug effects , Nicotine/administration & dosage , Nicotinic Antagonists/pharmacology , Pressure , Protein Subunits , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/classification , Receptors, Nicotinic/metabolism , Stomach/innervation , Vagotomy , Vagus Nerve/drug effects , Vagus Nerve/physiology
16.
PLoS One ; 10(7): e0130255, 2015.
Article in English | MEDLINE | ID: mdl-26132172

ABSTRACT

For over a century, the behavior of the aorta and other large arteries has been described as passive elastic tubes in which no active contraction occurs in the smooth muscle wall. In response to pulsatile pressure changes, the vessels undergo a 'passive' elastic dilatation-contraction cycle, described as a "Windkessel" effect. However, Mangel and colleagues have presented evidence that is contrary to this view. They reported that in the rabbit, the aorta undergoes rhythmic 'active' (contraction) during the cardiac cycle; but these findings have been largely ignored. In the present study, we observed spontaneous contractions in synchrony with the heartbeat in another species (rat). In addition we demonstrate that aorta contractions are of neurogenic origin. Electrical stimulation of the aorta evoked contractions that occur at a rate that is in the range of the animal's heartbeat and are suppressed by tetrodotoxin and the alpha-adrenergic receptor blocker, phentolamine. Altogether, these findings indicate that aortic contractions are under neural control from the heart.


Subject(s)
Aorta/physiology , Biological Clocks , Muscle Contraction , Animals , Electric Stimulation , Male , Rats , Rats, Sprague-Dawley , Vasoconstriction
17.
Auton Neurosci ; 160(1-2): 21-6, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21112817

ABSTRACT

We reported pharmacological data suggesting that stimulation of a vago-vagal reflex activates GABAergic neurons in the hindbrain that inhibit dorsal motor nucleus of the vagus (DMV) neurons projecting to the antrum, but not to the fundus (Ferreira et al., 2002). The purpose of this study was to use an ultrastructural approach to test the hypothesis that GABAergic terminals form synapses with DMV antrum-projecting neurons, but not with DMV fundus-projecting neurons. A retrograde tracer, CTB-HRP, was injected into the gastric smooth muscle of either the fundus or the antrum of anesthetized rats. Animals were re-anesthetized 48 h later and perfusion-fixed with acrolein and paraformaldehyde. Brainstems were processed histochemically for CTB-HRP, and immunocytochemically for glutamic acid decarboxylase isoenzyme 67 immunoreactivity (GAD67-IR) by dual-labeling electron microscopic methods. Most cell bodies and dendrites of neurons that were retrogradely labeled from the stomach occurred at the level of the area postrema. Examination of 214 synapses on 195 neurons that projected to the antrum revealed that 23.0+/-3.6% (n = 4) of synaptic contacts were with GAD67-IR terminals. The examination of 220 synapses on 203 fundus-projecting neurons revealed that only 7.9+/-3.1% (n = 4) of synaptic contacts were with GAD67-IR terminals. The difference between GAD67-IR synaptic contacts with antrum- and fundus-projecting neurons was statistically significant (p<0.05). These data suggest that brainstem circuitry controlling the antrum involves GABAergic transmission.


Subject(s)
Efferent Pathways/metabolism , Pyloric Antrum/innervation , Vagus Nerve/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Efferent Pathways/ultrastructure , Glutamate Decarboxylase/metabolism , Immunohistochemistry , Microscopy, Electron, Transmission , Pyloric Antrum/metabolism , Rats , Vagus Nerve/ultrastructure
18.
Am J Physiol Gastrointest Liver Physiol ; 296(1): G101-11, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19008339

ABSTRACT

It has been proposed that there is an "apparent monosynaptic" connection between gastric vagal afferent nerve terminals and inhibitory projection neurons in the nucleus tractus solitarius (NTS) and that two efferent parallel pathways from the dorsal motor nucleus of the vagus (DMV) influence peripheral organs associated with these reflexes (6). The purpose of our study was to verify the validity of these views as they relate to basal control of gastric motility. To test the validity of a direct connection of vagal afferent terminals (known to release l-glutamate) directly impacting second-order projection neurons, we evaluated the effect of GABA(A) receptor blockade in the area of the medial subnucleus of the tractus solitarius (mNTS) on gastric motility. Microinjection of bicuculline methiodide into the mNTS produced robust decreases in gastric motility (-1.6 +/- 0.2 mmHg, P < 0.05, n = 23), which were prevented by cervical vagotomy and by pretreatment with kynurenic acid microinjected into the mNTS. Kynurenic acid per se had no effect on gastric motility. However, after GABA(A) receptor blockade in the mNTS, kynurenic acid produced a robust increase in gastric motility. To test for the contribution of two parallel efferent DMV pathways, we assessed the effect of either intravenous atropine methylbromide or N(G)-nitro-l-arginine methyl ester on baseline motility and on decreases in gastric motility induced by GABA(A) receptor blockade in the mNTS. Only atropine methylbromide altered baseline motility and prevented the effects of GABA(A) receptor blockade on gastric motility. Our data demonstrate the presence of intra-NTS GABAergic signaling between the vagal afferent nerve terminals and inhibitory projection neurons in the NTS and that the cholinergic-cholinergic excitatory pathway comprises the functionally relevant efferent arm of the vagovagal circuit.


Subject(s)
Cholinergic Fibers/metabolism , Gastrointestinal Motility , Neural Inhibition , Reflex , Solitary Nucleus/metabolism , Stomach/innervation , Vagus Nerve/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Atropine Derivatives/pharmacology , Bicuculline/administration & dosage , Bicuculline/analogs & derivatives , Cholinergic Fibers/drug effects , Excitatory Amino Acid Antagonists/administration & dosage , GABA Antagonists/administration & dosage , GABA-A Receptor Antagonists , Gastrointestinal Motility/drug effects , Kynurenic Acid/administration & dosage , Male , Microinjections , Models, Animal , Muscarinic Antagonists/pharmacology , Neural Inhibition/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Reflex/drug effects , Reproducibility of Results , Solitary Nucleus/drug effects , Vagotomy , Vagus Nerve/drug effects
19.
Auton Neurosci ; 144(1-2): 50-60, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18986853

ABSTRACT

The lower esophageal sphincter (LES) and the crural diaphragm (CD) surrounding the esophagogastric junction are key components of the gastroesophageal reflex mechanism, which engages the vago-vagal brainstem circuitry. Although both components work in conjunction to prevent gastroesophageal reflux, little is known about the brain area(s) where this integration takes place. The aims of this study were to: (1) trace the brainstem circuitry associated with the CD and the LES, and (2) determine possible sites of convergence. Experiments were done in adult male ferrets. Under isoflurane anesthesia, recombinant strains of the transneuronal pseudorabies virus (PRV-151 or PRV-Bablu) or the monosynaptic retrograde tracer cholera toxin beta-subunit (CTb) were injected into either the CD or the LES. Following a survival period of 5-7 days, animals were euthanized, perfused and their brains removed for dual-labeling immunofluorescence processing. In animals injected with recombinants of PRV into the CD and the LES, distinct labeling was found in various brainstem nuclei including: area postrema, DMV, nucleus tractus solitarius (NTS), medial reticular formation (MRF) and nucleus ambiguous (NA). Double-labeled cells were only evident in the DMV, NTS and MRF. Injections of CTb into the CD or the LES resulted in retrograde labeling only in the DMV. These findings demonstrate the presence of a direct projection from the DMV to the CD. They further suggest that the neuronal connections responsible for CD or LES function are contained in circuitries that, though largely independent, may converge at the level of DMV, NTS and MRF.


Subject(s)
Brain Stem/anatomy & histology , Diaphragm/innervation , Esophageal Sphincter, Lower/innervation , Ferrets/anatomy & histology , Vagus Nerve/anatomy & histology , Visceral Afferents/anatomy & histology , Animals , Area Postrema/anatomy & histology , Area Postrema/physiology , Brain Mapping , Brain Stem/physiology , Cholera Toxin , Diaphragm/physiology , Esophageal Sphincter, Lower/physiology , Esophagogastric Junction/innervation , Esophagogastric Junction/physiology , Ferrets/physiology , Gastroesophageal Reflux/physiopathology , Herpesvirus 1, Suid , Male , Medulla Oblongata/anatomy & histology , Medulla Oblongata/physiology , Motor Neurons/cytology , Motor Neurons/physiology , Reticular Formation/anatomy & histology , Reticular Formation/physiology , Solitary Nucleus/anatomy & histology , Solitary Nucleus/physiology , Species Specificity , Staining and Labeling , Vagus Nerve/physiology , Visceral Afferents/physiology
20.
Am J Physiol Regul Integr Comp Physiol ; 294(3): R720-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18199591

ABSTRACT

Quantitative analysis of innervation to dorsal motor nucleus of the vagus (DMV) fundus-projecting neurons indicates that approximately 17% of input neurons are noradrenergic. To determine whether this small percentage of neurons innervating DMV output to the stomach is physiologically relevant, we evaluated the role of norepinephrine at the DMV in mediating a vagovagal reflex controlling the fundus. A strain gauge was sutured onto the fundus of isoflurane-anesthetized rats to monitor changes in tone evoked by esophageal distension (ED). ED produced a decrease in fundus tone of 0.31 +/- 0.02 g (P < 0.05), which could be reproduced after a 30-min interval between distensions. Bilateral cervical vagotomy and/or pretreatment with intravenous atropine methylbromide prevented the reflex-induced fundus relaxation. In contrast, intravenous N(G)-nitro-L-arginine methyl ester had no effect. Bilateral microinjection of alpha2-adrenoreceptor antagonists (yohimbine and RS-79948) into the DMV also prevented the response. Before microinjection of alpha2-adrenoreceptor antagonists, ED decreased fundus tone by 0.33 +/- 0.05 g (P < 0.05). After antagonist microinjection, ED decreased fundus tone by only 0.05 +/- 0.06 g (P > 0.05). Bilateral microinjection of prazosin into the DMV had no effect on the response. Microinjection of norepinephrine into the DMV mimicked the effect of ED and was also prevented by prior microinjection of an alpha2-adrenoreceptor antagonist. Our results indicate that noradrenergic innervation of DMV fundus-projecting neurons is physiologically important and suggest that norepinephrine released at the DMV acts on alpha2-adrenoreceptors to inhibit activity in a cholinergic-cholinergic excitatory pathway to the fundus.


Subject(s)
Motor Neurons/physiology , Muscle Tonus/physiology , Norepinephrine/physiology , Parasympathetic Nervous System/physiology , Reflex/physiology , Synaptic Transmission/physiology , Vagus Nerve/physiology , Adrenergic Antagonists/pharmacology , Adrenergic alpha-1 Receptor Agonists , Animals , Atropine/administration & dosage , Atropine/pharmacology , Dilatation , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Esophagus/physiology , Gastric Fundus , Male , Microinjections , Muscarinic Antagonists/administration & dosage , Muscarinic Antagonists/pharmacology , Muscle Relaxation/drug effects , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Norepinephrine/antagonists & inhibitors , Norepinephrine/pharmacology , Parasympathetic Nervous System/drug effects , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL