Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
J Neurosci ; 44(25)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38729762

ABSTRACT

Inhibitory neurons embedded within mammalian neural circuits shape breathing, walking, and other rhythmic motor behaviors. At the core of the neural circuit controlling breathing is the preBötzinger Complex (preBötC), where GABAergic (GAD1/2+) and glycinergic (GlyT2+) neurons are functionally and anatomically intercalated among glutamatergic Dbx1-derived (Dbx1+) neurons that generate rhythmic inspiratory drive. The roles of these preBötC inhibitory neurons in breathing remain unclear. We first characterized the spatial distribution of molecularly defined preBötC inhibitory subpopulations in male and female neonatal double reporter mice expressing either tdTomato or EGFP in GlyT2+, GAD1+, or GAD2+ neurons. We found that the majority of preBötC inhibitory neurons expressed both GlyT2 and GAD2 while a much smaller subpopulation also expressed GAD1. To determine the functional role of these subpopulations, we used holographic photostimulation, a patterned illumination technique, in rhythmically active medullary slices from neonatal Dbx1tdTomato;GlyT2EGFP and Dbx1tdTomato;GAD1EGFP double reporter mice of either sex. Stimulation of 4 or 8 preBötC GlyT2+ neurons during endogenous rhythm prolonged the interburst interval in a phase-dependent manner and increased the latency to burst initiation when bursts were evoked by stimulation of Dbx1+ neurons. In contrast, stimulation of 4 or 8 preBötC GAD1+ neurons did not affect interburst interval or latency to burst initiation. Instead, photoactivation of GAD1+ neurons during the inspiratory burst prolonged endogenous and evoked burst duration and decreased evoked burst amplitude. We conclude that GlyT2+/GAD2+ neurons modulate breathing rhythm by delaying burst initiation while a smaller GAD1+ subpopulation shapes inspiratory patterning by altering burst duration and amplitude.


Subject(s)
Inhalation , Animals , Mice , Female , Male , Inhalation/physiology , Neural Inhibition/physiology , Medulla Oblongata/physiology , Medulla Oblongata/cytology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Mice, Transgenic , Glycine Plasma Membrane Transport Proteins/genetics , Glycine Plasma Membrane Transport Proteins/metabolism , Respiratory Center/physiology , Respiratory Center/cytology , Neurons/physiology , Periodicity , Animals, Newborn
2.
Nature ; 536(7614): 76-80, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27462817

ABSTRACT

Breathing must be tightly coordinated with other behaviours such as vocalization, swallowing, and coughing. These behaviours occur after inspiration, during a respiratory phase termed postinspiration. Failure to coordinate postinspiration with inspiration can result in aspiration pneumonia, the leading cause of death in Alzheimer's disease, Parkinson's disease, dementia, and other neurodegenerative diseases. Here we describe an excitatory network that generates the neuronal correlate of postinspiratory activity in mice. Glutamatergic-cholinergic neurons form the basis of this network, and GABA (γ-aminobutyric acid)-mediated inhibition establishes the timing and coordination relative to inspiration. We refer to this network as the postinspiratory complex (PiCo). The PiCo has autonomous rhythm-generating properties and is necessary and sufficient for postinspiratory activity in vivo.The PiCo also shows distinct responses to neuromodulators when compared to other excitatory brainstem networks. On the basis of the discovery of the PiCo, we propose that each of the three phases of breathing is generated by a distinct excitatory network: the pre-Bötzinger complex, which has been linked to inspiration; the PiCo, as described here for the neuronal control of postinspiration; and the lateral parafacial region (pF(L)), which has been associated with active expiration, a respiratory phase that is recruited during high metabolic demand.


Subject(s)
Neural Pathways/physiology , Respiration , Respiratory Center/physiology , Animals , Cholinergic Neurons/metabolism , Female , Glutamine/metabolism , Male , Mice , Neural Inhibition/physiology , Neural Pathways/cytology , Respiratory Center/anatomy & histology , Respiratory Center/cytology , Synapses/metabolism , Time Factors , gamma-Aminobutyric Acid/metabolism
3.
Nature ; 530(7590): 293-297, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26855425

ABSTRACT

Sighs are long, deep breaths expressing sadness, relief or exhaustion. Sighs also occur spontaneously every few minutes to reinflate alveoli, and sighing increases under hypoxia, stress, and certain psychiatric conditions. Here we use molecular, genetic, and pharmacologic approaches to identify a peptidergic sigh control circuit in murine brain. Small neural subpopulations in a key breathing control centre, the retrotrapezoid nucleus/parafacial respiratory group (RTN/pFRG), express bombesin-like neuropeptide genes neuromedin B (Nmb) or gastrin-releasing peptide (Grp). These project to the preBötzinger Complex (preBötC), the respiratory rhythm generator, which expresses NMB and GRP receptors in overlapping subsets of ~200 neurons. Introducing either neuropeptide into preBötC or onto preBötC slices, induced sighing or in vitro sigh activity, whereas elimination or inhibition of either receptor reduced basal sighing, and inhibition of both abolished it. Ablating receptor-expressing neurons eliminated basal and hypoxia-induced sighing, but left breathing otherwise intact initially. We propose that these overlapping peptidergic pathways comprise the core of a sigh control circuit that integrates physiological and perhaps emotional input to transform normal breaths into sighs.


Subject(s)
Gastrin-Releasing Peptide/metabolism , Neurokinin B/analogs & derivatives , Neurons/physiology , Receptors, Bombesin/metabolism , Respiration , Signal Transduction/physiology , Animals , Bombesin/pharmacology , Emotions/physiology , Female , Gastrin-Releasing Peptide/deficiency , Gastrin-Releasing Peptide/genetics , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Neurokinin B/deficiency , Neurokinin B/genetics , Neurokinin B/metabolism , Neurokinin B/pharmacology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Respiration/drug effects , Respiratory Center/cytology , Respiratory Center/drug effects , Respiratory Center/physiology , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins , Signal Transduction/drug effects
4.
Proc Natl Acad Sci U S A ; 114(48): 12815-12820, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133427

ABSTRACT

Central neural networks operate continuously throughout life to control respiration, yet mechanisms regulating ventilatory frequency are poorly understood. Inspiration is generated by the pre-Bötzinger complex of the ventrolateral medulla, where it is thought that excitation increases inspiratory frequency and inhibition causes apnea. To test this model, we used an in vitro optogenetic approach to stimulate select populations of hindbrain neurons and characterize how they modulate frequency. Unexpectedly, we found that inhibition was required for increases in frequency caused by stimulation of Phox2b-lineage, putative CO2-chemosensitive neurons. As a mechanistic explanation for inhibition-dependent increases in frequency, we found that phasic stimulation of inhibitory neurons can increase inspiratory frequency via postinhibitory rebound. We present evidence that Phox2b-mediated increases in frequency are caused by rebound excitation following an inhibitory synaptic volley relayed by expiration. Thus, although it is widely thought that inhibition between inspiration and expiration simply prevents activity in the antagonistic phase, we instead propose a model whereby inhibitory coupling via postinhibitory rebound excitation actually generates fast modes of inspiration.


Subject(s)
Carbon Dioxide/pharmacology , Exhalation/drug effects , Inhalation/drug effects , Neurons/drug effects , Respiratory Center/drug effects , Respiratory Rate/drug effects , Animals , Carbon Dioxide/metabolism , Exhalation/physiology , Female , Hypoglossal Nerve/drug effects , Inhalation/physiology , Male , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Medulla Oblongata/physiology , Mice , Neurons/cytology , Neurons/physiology , Optogenetics/methods , Phrenic Nerve/drug effects , Picrotoxin/pharmacology , Prazosin/pharmacology , Propranolol/pharmacology , Respiratory Center/cytology , Respiratory Center/physiology , Respiratory Rate/physiology , Spinal Nerve Roots/drug effects , Strychnine/pharmacology , Substance P/pharmacology
5.
PLoS Comput Biol ; 14(4): e1006148, 2018 04.
Article in English | MEDLINE | ID: mdl-29698394

ABSTRACT

The circuit organization within the mammalian brainstem respiratory network, specifically within and between the pre-Bötzinger (pre-BötC) and Bötzinger (BötC) complexes, and the roles of these circuits in respiratory pattern generation are continuously debated. We address these issues with a combination of optogenetic experiments and modeling studies. We used transgenic mice expressing channelrhodopsin-2 under the VGAT-promoter to investigate perturbations of respiratory circuit activity by site-specific photostimulation of inhibitory neurons within the pre-BötC or BötC. The stimulation effects were dependent on the intensity and phase of the photostimulation. Specifically: (1) Low intensity (≤ 1.0 mW) pulses delivered to the pre-BötC during inspiration did not terminate activity, whereas stronger stimulations (≥ 2.0 mW) terminated inspiration. (2) When the pre-BötC stimulation ended in or was applied during expiration, rebound activation of inspiration occurred after a fixed latency. (3) Relatively weak sustained stimulation (20 Hz, 0.5-2.0 mW) of pre-BötC inhibitory neurons increased respiratory frequency, while a further increase of stimulus intensity (> 3.0 mW) reduced frequency and finally (≥ 5.0 mW) terminated respiratory oscillations. (4) Single pulses (0.2-5.0 s) applied to the BötC inhibited rhythmic activity for the duration of the stimulation. (5) Sustained stimulation (20 Hz, 0.5-3.0 mW) of the BötC reduced respiratory frequency and finally led to apnea. We have revised our computational model of pre-BötC and BötC microcircuits by incorporating an additional population of post-inspiratory inhibitory neurons in the pre-BötC that interacts with other neurons in the network. This model was able to reproduce the above experimental findings as well as previously published results of optogenetic activation of pre-BötC or BötC neurons obtained by other laboratories. The proposed organization of pre-BötC and BötC circuits leads to testable predictions about their specific roles in respiratory pattern generation and provides important insights into key circuit interactions operating within brainstem respiratory networks.


Subject(s)
Models, Neurological , Respiratory Center/physiology , Animals , Central Pattern Generators/physiology , Computational Biology , Computer Simulation , Connectome , Electrophysiological Phenomena , Mice , Mice, Transgenic , Optogenetics , Photic Stimulation , Respiratory Center/cytology , Respiratory Physiological Phenomena , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
6.
Pflugers Arch ; 470(11): 1659-1672, 2018 11.
Article in English | MEDLINE | ID: mdl-30054719

ABSTRACT

Optogenetic stimulation of the adrenergic C1 neurons produces cardiorespiratory activation, and selective depletion of these cells attenuates breathing responses induced by hypoxia. The preBötzinger complex (preBötC) is a group of neurons located in the intermediate aspect of the ventrolateral medulla, critical for respiratory rhythmogenesis, and is modulated by glutamate and catecholamines. Our hypothesis is that selective activation of C1 neurons leads to breathing responses by excitatory connections with the preBötC neurons. Anatomical connection between C1 cells and preBötC was evaluated using retrograde (Cholera Toxin b; preBötC) and anterograde (LVV-PRSx8-ChR2-eYFP; C1 region) tracers. LVV-PRSx8-ChR2-eYFP (viral vector that expresses channelrhodopsin-2 (ChR2) under the control of the catecholaminergic neuron-preferring promoter (PRSx8) was also injected into the C1 region of male Wistar rats for the functional experiments. Anatomical results demonstrated that preBötC neurons receive projections from C1 cells, and these projections express tyrosine hydroxylase and vesicular glutamate transporter 2. Functional connection between C1 cells and preBötC was evaluated by photostimulation of ChR2-transduced C1 neurons before and after unilateral injection of the ionotropic glutamate antagonist, kynurenic acid (kyn), or cocktail of adrenergic antagonists in the preBötC. Kyn injection into preBötC blocked the increase in DiaEMG frequency without changing the MAP increase elicited by photostimulation of C1 neurons, while the injection of adrenergic antagonists into the preBötC did not change DiaEMG frequency and MAP increase induced by photostimulation of C1 cells. Our results suggest that the increase in breathing produced by photostimulation of C1 neurons can be caused by a direct glutamatergic activation of preBötC neurons.


Subject(s)
Adrenergic Neurons/physiology , Respiration , Respiratory Center/physiology , Adrenergic Antagonists/pharmacology , Adrenergic Neurons/drug effects , Adrenergic Neurons/metabolism , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Excitatory Amino Acid Antagonists/pharmacology , Kynurenic Acid/pharmacology , Male , Optogenetics , Rats , Rats, Wistar , Respiratory Center/cytology , Respiratory Center/metabolism , Tyrosine 3-Monooxygenase/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
7.
J Neurosci ; 36(27): 7223-33, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27383596

ABSTRACT

UNLABELLED: Breathing in mammals depends on rhythms that originate from the preBötzinger complex (preBötC) of the ventral medulla and a network of brainstem and spinal premotor neurons. The rhythm-generating core of the preBötC, as well as some premotor circuits, consist of interneurons derived from Dbx1-expressing precursors (Dbx1 neurons), but the structure and function of these networks remain incompletely understood. We previously developed a cell-specific detection and laser ablation system to interrogate respiratory network structure and function in a slice model of breathing that retains the preBötC, the respiratory-related hypoglossal (XII) motor nucleus and XII premotor circuits. In spontaneously rhythmic slices, cumulative ablation of Dbx1 preBötC neurons decreased XII motor output by ∼50% after ∼15 cell deletions, and then decelerated and terminated rhythmic function altogether as the tally increased to ∼85 neurons. In contrast, cumulatively deleting Dbx1 XII premotor neurons decreased motor output monotonically but did not affect frequency nor stop XII output regardless of the ablation tally. Here, we couple an existing preBötC model with a premotor population in several topological configurations to investigate which one may replicate the laser ablation experiments best. If the XII premotor population is a "small-world" network (rich in local connections with sparse long-range connections among constituent premotor neurons) and connected with the preBötC such that the total number of incoming synapses remains fixed, then the in silico system successfully replicates the in vitro laser ablation experiments. This study proposes a feasible configuration for circuits consisting of Dbx1-derived interneurons that generate inspiratory rhythm and motor pattern. SIGNIFICANCE STATEMENT: To produce a breathing-related motor pattern, a brainstem core oscillator circuit projects to a population of premotor interneurons, but the assemblage of this network remains incompletely understood. Here we applied network modeling and numerical simulation to discover respiratory circuit configurations that successfully replicate photonic cell ablation experiments targeting either the core oscillator or premotor network, respectively. If premotor neurons are interconnected in a so-called "small-world" network with a fixed number of incoming synapses balanced between premotor and rhythmogenic neurons, then our simulations match their experimental benchmarks. These results provide a framework of experimentally testable predictions regarding the rudimentary structure and function of respiratory rhythm- and pattern-generating circuits in the brainstem of mammals.


Subject(s)
Motor Neurons/physiology , Nerve Net/physiology , Periodicity , Respiration , Respiratory Center/cytology , Spinal Cord/cytology , Action Potentials/physiology , Animals , Homeodomain Proteins/metabolism , Interneurons/physiology , Models, Neurological , Patch-Clamp Techniques , Respiratory Center/physiology , Reticular Formation/cytology
8.
J Physiol ; 595(23): 7063-7079, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28734063

ABSTRACT

KEY POINTS: The paratrigeminal respiratory group (pTRG) is responsible for the respiratory pattern generation in the lamprey. The role of ATP and astrocytes, known to control respiratory activity in mammals, was investigated in the lamprey respiratory network. ATP microinjected into the pTRG induces a biphasic response consisting of marked increases in respiratory frequency mediated by P2X receptors followed by a decrease in the respiratory motor output due to the ATP metabolite adenosine. We provide evidence that astrocytes are involved in the genesis of the normal respiratory pattern, ATP-induced responses and acidification-induced increases of the respiratory activity. The function of astrocytes in rhythmic networks appears to be phylogenetically conserved. ABSTRACT: The role of ATP and astrocytes in respiratory rhythm modulation has been recently investigated in neonatal rodents. However, no information on the role of ATP and astrocytes within the respiratory network of the lamprey is available, particularly within the paratrigeminal respiratory group (pTRG), the proposed respiratory central pattern generator. To address these issues, the present study was carried out on isolated brainstems of the adult lamprey. Bath application of ATP caused marked increases in respiratory frequency followed by decreases in the respiratory motor output, mediated by the ATP metabolite adenosine at the level of the pTRG. Bath applications and microinjections of agonists and antagonists of purinergic receptors showed that ATP increased respiratory activity through an action on pTRG P2X receptors. To disclose the respiratory role of astrocytes, we used bath application of the gliotoxin aminoadipic acid, which dramatically depressed the respiratory motor output that, however, promptly recovered following glutamine application. Furthermore, the excitatory responses to ATP-γ-S (a non-hydrolysable ATP analogue), but not to substance P, microinjected into the pTRG, were abolished. Finally, we also demonstrated that acidification-induced increases in respiratory activity were ATP-independent, but mediated by the astrocytes' glutamate-glutamine cycle. The results show for the first time that ATP and especially astrocytes strongly contribute to the modulation of the lamprey respiratory pattern. Their role in the modulation or maintenance of rhythmic neuronal activities appears to be phylogenetically conserved.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Central Pattern Generators/metabolism , Respiratory Center/metabolism , Animals , Astrocytes/physiology , Central Pattern Generators/cytology , Central Pattern Generators/physiology , Lampreys , Receptors, Purinergic P2X/metabolism , Respiratory Center/cytology , Respiratory Center/physiology
9.
Proc Natl Acad Sci U S A ; 111(17): 6449-54, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733934

ABSTRACT

Previously we defined neuronal subclasses within the mouse peripheral nervous system using an experimental strategy called "constellation pharmacology." Here we demonstrate the broad applicability of constellation pharmacology by extending it to the CNS and specifically to the ventral respiratory column (VRC) of mouse brainstem, a region containing the neuronal network controlling respiratory rhythm. Analysis of dissociated cells from this locus revealed three major cell classes, each encompassing multiple subclasses. We broadly analyzed the combinations (constellations) of receptors and ion channels expressed within VRC cell classes and subclasses. These were strikingly different from the constellations of receptors and ion channels found in subclasses of peripheral neurons from mouse dorsal root ganglia. Within the VRC cell population, a subset of dissociated neurons responded to substance P, putatively corresponding to inspiratory pre-Bötzinger complex (preBötC) neurons. Using constellation pharmacology, we found that these substance P-responsive neurons also responded to histamine, and about half responded to bradykinin. Electrophysiological studies conducted in brainstem slices confirmed that preBötC neurons responsive to substance P exhibited similar responsiveness to bradykinin and histamine. The results demonstrate the predictive utility of constellation pharmacology for defining modulatory inputs into specific neuronal subclasses within central neuronal networks.


Subject(s)
Central Nervous System/cytology , Neurons/physiology , Animals , Bradykinin/pharmacology , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/physiology , Calcium/metabolism , Cells, Cultured , Cluster Analysis , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Histamine/pharmacology , Imaging, Three-Dimensional , Male , Mice , Mice, Inbred C57BL , Nerve Net/cytology , Nerve Net/drug effects , Nerve Net/physiology , Neurons/drug effects , Receptors, Cholinergic/metabolism , Receptors, Glutamate/metabolism , Respiratory Center/cytology , Substance P/pharmacology
10.
J Neurophysiol ; 116(3): 1024-35, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27306669

ABSTRACT

Chemosensitive neurons in the retrotrapezoid nucleus (RTN) regulate breathing in response to CO2/H(+) changes and serve as an integration center for other autonomic centers, including brain stem noradrenergic neurons. Norepinephrine (NE) contributes to respiratory control and chemoreception, and, since disruption of NE signaling may contribute to several breathing disorders, we sought to characterize effects of NE on RTN chemoreception. All neurons included in this study responded similarly to CO2/H(+) but showed differential sensitivity to NE; we found that NE activated (79%), inhibited (7%), or had no effect on activity (14%) of RTN chemoreceptors. The excitatory effect of NE on RTN chemoreceptors was dose dependent, retained in the presence of neurotransmitter receptor blockers, and could be mimicked and blocked by pharmacological manipulation of α1-adrenergic receptors (ARs). In addition, NE-activation was blunted by XE991 (KCNQ channel blocker), and partially occluded the firing response to serotonin, suggesting involvement of KCNQ channels. However, in whole cell voltage clamp, activation of α1-ARs decreased outward current and conductance by what appears to be a mixed effect on multiple channels. The inhibitory effect of NE on RTN chemoreceptors was blunted by an α2-AR antagonist. A third group of RTN chemoreceptors was insensitive to NE. We also found that chemosensitive RTN astrocytes do not respond to NE with a change in voltage or by releasing ATP to enhance activity of chemosensitive neurons. These results indicate NE modulates subsets of RTN chemoreceptors by mechanisms involving α1- and α2-ARs.


Subject(s)
Action Potentials/drug effects , Chemoreceptor Cells/drug effects , Chemoreceptor Cells/physiology , Norepinephrine/metabolism , Receptors, Adrenergic/metabolism , Respiratory Center/cytology , Adrenergic Agents/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Astrocytes/drug effects , Carbon Dioxide/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Neurotransmitter Agents/pharmacology , Norepinephrine/pharmacology , Patch-Clamp Techniques , Rats , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
11.
J Neurophysiol ; 116(3): 1036-48, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27306670

ABSTRACT

Norepinephrine (NE) is a potent modulator of breathing that can increase/decrease respiratory activity by α1-/α2-adrenergic receptor (AR) activation, respectively. The retrotrapezoid nucleus (RTN) is known to contribute to central chemoreception, inspiration, and active expiration. Here we investigate the sources of catecholaminergic inputs to the RTN and identify respiratory effects produced by activation of ARs in this region. By injecting the retrograde tracer Fluoro-Gold into the RTN, we identified back-labeled catecholaminergic neurons in the A7 region. In urethane-anesthetized, vagotomized, and artificially ventilated male Wistar rats unilateral injection of NE or moxonidine (α2-AR agonist) blunted diaphragm muscle activity (DiaEMG) frequency and amplitude, without changing abdominal muscle activity. Those inhibitory effects were reduced by preapplication of yohimbine (α2-AR antagonist) into the RTN. Conversely, unilateral RTN injection of phenylephrine (α1-AR agonist) increased DiaEMG amplitude and frequency and facilitated active expiration. This response was blocked by prior RTN injection of prazosin (α1-AR antagonist). Interestingly, RTN injection of propranolol (ß-AR antagonist) had no effect on respiratory inhibition elicited by applications of NE into the RTN; however, the combined blockade of α2- and ß-ARs (coapplication of propranolol and yohimbine) revealed an α1-AR-dependent excitatory response to NE that resulted in increase in DiaEMG frequency and facilitation of active expiration. However, blockade of α1-, α2-, or ß-ARs in the RTN had minimal effect on baseline respiratory activity, on central or peripheral chemoreflexes. These results suggest that NE signaling can modulate RTN chemoreceptor function; however, endogenous NE signaling does not contribute to baseline breathing or the ventilatory response to central or peripheral chemoreceptor activity in urethane-anesthetized rats.


Subject(s)
Anesthesia , Chemoreceptor Cells/physiology , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Respiration , Respiratory Center/cytology , Action Potentials/drug effects , Adrenergic Agents/pharmacology , Animals , Chemoreceptor Cells/drug effects , Diaphragm/physiology , Enzyme Inhibitors , Male , Norepinephrine/pharmacology , Potassium Cyanide/pharmacology , Rats , Rats, Wistar , Respiration/drug effects , Respiratory Center/diagnostic imaging , Stilbamidines/metabolism , Vagotomy
12.
J Comput Neurosci ; 41(3): 245-268, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27491968

ABSTRACT

Experimental results in rodent medullary slices containing the pre-Bötzinger complex (pre-BötC) have identified multiple bursting mechanisms based on persistent sodium current (I NaP) and intracellular Ca2+. The classic two-timescale approach to the analysis of pre-BötC bursting treats the inactivation of I NaP, the calcium concentration, as well as the Ca2+-dependent inactivation of IP 3 as slow variables and considers other evolving quantities as fast variables. Based on its time course, however, it appears that a novel mixed bursting (MB) solution, observed both in recordings and in model pre-BötC neurons, involves at least three timescales. In this work, we consider a single-compartment model of a pre-BötC inspiratory neuron that can exhibit both I NaP and Ca2+ oscillations and has the ability to produce MB solutions. We use methods of dynamical systems theory, such as phase plane analysis, fast-slow decomposition, and bifurcation analysis, to better understand the mechanisms underlying the MB solution pattern. Rather surprisingly, we discover that a third timescale is not actually required to generate mixed bursting solutions. Through our analysis of timescales, we also elucidate how the pre-BötC neuron model can be tuned to improve the robustness of the MB solution.


Subject(s)
Action Potentials/physiology , Models, Neurological , Neurons/physiology , Nonlinear Dynamics , Respiratory Center/cytology , Animals , Computer Simulation , In Vitro Techniques , Rodentia , Time Factors
13.
Anesth Analg ; 122(5): 1586-93, 2016 May.
Article in English | MEDLINE | ID: mdl-26962714

ABSTRACT

BACKGROUND: Lidocaine is widely used in the clinical setting as a local anesthetic and antiarrhythmic drug. Although it has been suggested that lidocaine exerts inhibitory effects on the central and peripheral neurons, there are no reports on its effects on central respiratory activity in vertebrates. In this study, we examined the effects of lidocaine on respiratory rhythm generation and nociceptive response in brainstem-spinal cord preparations from the newborn rats. METHODS: Preparations were isolated from Wistar rats (postnatal day 0-3) and superfused with artificial cerebrospinal fluid equilibrated with 95% O2 and 5% CO2, pH 7.4, at 25°C to 26°C. We examined the effects of lidocaine on the fourth cervical ventral root (C4)-inspiratory activity and on the preinspiratory and inspiratory neurons in the rostral medulla. We also examined the effects on the C4/C5 reflex responses induced by ipsilateral C7/C8 dorsal root stimulation, which are thought to be related to the nociceptive response. RESULTS: The application of low doses of lidocaine (10-20 µM) resulted in a slight increase of the C4 burst rate, whereas high doses of lidocaine (100-400 µM) decreased the burst rate in a dose-dependent manner, eventually resulting in the complete cessation of respiratory rhythm. High doses of lidocaine decreased the burst duration and negative slope conductance of preinspiratory neurons, suggesting that lidocaine blocked persistent Na+ current. After the burst generation of the respiratory neurons ceased, depolarizing current stimulation continued to induce action potentials; however, the induction of the spike train was depressed because of strong adaptation. A low dose of lidocaine (20 µM) depressed C4/C5 spinal reflex responses. CONCLUSIONS: Our findings indicate that lidocaine depressed nociception-related responses at lower concentrations than those that induced respiratory depression. Our report provides the basic neuronal mechanisms to support the clinical use of lidocaine, which shows antinociceptive effects with minimal side effects on breathing.


Subject(s)
Anesthetics, Local/pharmacology , Lidocaine/pharmacology , Nerve Fibers, Unmyelinated/drug effects , Nociception/drug effects , Respiratory Center/drug effects , Respiratory Rate/drug effects , Sodium Channel Blockers/pharmacology , Spinal Cord/drug effects , Animals , Animals, Newborn , Dose-Response Relationship, Drug , In Vitro Techniques , Membrane Potentials , Nerve Fibers, Unmyelinated/metabolism , Rats, Wistar , Reflex/drug effects , Respiratory Center/cytology , Respiratory Center/metabolism , Sodium/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Time Factors
14.
Adv Exp Med Biol ; 949: 109-145, 2016.
Article in English | MEDLINE | ID: mdl-27714687

ABSTRACT

Astrocytes perform various homeostatic functions in the nervous system beyond that of a supportive or metabolic role for neurons. A growing body of evidence indicates that astrocytes are crucial for central respiratory chemoreception. This review presents a classical overview of respiratory central chemoreception and the new evidence for astrocytes as brainstem sensors in the respiratory response to hypercapnia. We review properties of astrocytes for chemosensory function and for modulation of the respiratory network. We propose that astrocytes not only mediate between CO2/H+ levels and motor responses, but they also allow for two emergent functions: (1) Amplifying the responses of intrinsic chemosensitive neurons through feedforward signaling via gliotransmitters and; (2) Recruiting non-intrinsically chemosensitive cells thanks to volume spreading of signals (calcium waves and gliotransmitters) to regions distant from the CO2/H+ sensitive domains. Thus, astrocytes may both increase the intensity of the neuron responses at the chemosensitive sites and recruit of a greater number of respiratory neurons to participate in the response to hypercapnia.


Subject(s)
Astrocytes/physiology , Carbon Dioxide/metabolism , Chemoreceptor Cells/physiology , Hypercapnia/metabolism , Neurons/physiology , Respiratory Center/physiology , Amino Acids/metabolism , Animals , Astrocytes/cytology , Calcium Signaling , Chemoreceptor Cells/cytology , Humans , Hypercapnia/physiopathology , Locus Coeruleus/cytology , Locus Coeruleus/physiology , Midbrain Raphe Nuclei/cytology , Midbrain Raphe Nuclei/physiology , Neurons/cytology , Neurotransmitter Agents/metabolism , Protons , Respiratory Center/cytology , Serotonin/metabolism , Synaptic Transmission
15.
J Neurophysiol ; 114(3): 1792-805, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26203106

ABSTRACT

The retrotrapezoid/parafacial respiratory group (RTN/pFRG) located ventral to the facial nucleus plays a key role in regulating breathing, especially enhanced expiratory activity during hypercapnic conditions. To clarify the roles of the RTN/pFRG region in evoking coughing, during which reflexive enhanced expiration is produced, and in swallowing, during which the expiratory activity is consistently halted, we recorded extracellular activity from RTN/pFRG neurons during these fictive behaviors in decerebrate, paralyzed, and artificially ventilated guinea pigs. The activity of the majority of recorded respiratory neurons was changed in synchrony with coughing and swallowing. To further evaluate the contribution of RTN/pFRG neurons to these nonrespiratory behaviors, the motor output patterns during breathing, coughing, and swallowing were compared before and after brain stem transection at the caudal margin of RTN/pFRG region. In addition, the effects of transection at its rostral margin were also investigated to evaluate pontine contribution to these behaviors. During respiration, transection at the rostral margin attenuated the postinspiratory activity of the recurrent laryngeal nerve. Meanwhile, the late expiratory activity of the abdominal nerve was abolished after caudal transection. The caudal transection also decreased the amplitude of the coughing-related abdominal nerve discharge but did not abolish the activity. Swallowing could be elicited even after the caudal end transection. These findings raise the prospect that the RTN/pFRG contributes to expiratory regulation during normal respiration, although this region is not an essential element of the neuronal networks involved in coughing and swallowing.


Subject(s)
Cough/physiopathology , Deglutition , Exhalation , Respiratory Center/physiology , Animals , Guinea Pigs , Male , Neurons/physiology , Respiratory Center/cytology
16.
Article in English | MEDLINE | ID: mdl-25634606

ABSTRACT

While once viewed as mere housekeepers, providing structural and metabolic support for neurons, it is now clear that neuroglia do much more. Phylogenetically, they have undergone enormous proliferation and diversification as central nervous systems grew in their complexity. In addition, they: i) are morphologically and functionally diverse; ii) play numerous, vital roles in maintaining CNS homeostasis; iii) are key players in brain development and responses to injury; and, iv) via gliotransmission, are likely participants in information processing. In this review, we discuss the diverse roles of neuroglia in maintaining homeostasis in the CNS, their evolutionary origins, the different types of neuroglia and their functional significance for respiratory control, and finally consider evidence that they contribute to the processing of chemosensory information in the respiratory network and the homeostatic control of blood gases.


Subject(s)
Neuroglia/physiology , Respiratory Center/physiology , Animals , Astrocytes/physiology , Biological Evolution , Central Nervous System/cytology , Central Nervous System/physiology , Ependymoglial Cells/physiology , Homeostasis , Humans , Microglia/physiology , Models, Neurological , Oligodendroglia/physiology , Respiratory Center/cytology , Respiratory Physiological Phenomena , Rett Syndrome/physiopathology
17.
J Neurosci ; 33(21): 9104-12, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23699521

ABSTRACT

A brainstem region, the paratrigeminal respiratory group (pTRG), has been suggested to play a crucial role in the respiratory rhythm generation in lampreys. However, a detailed characterization of the pTRG region is lacking. The present study performed on isolated brainstem preparations of adult lampreys provides a more precise localization of the pTRG region with regard to both connectivity and neurochemical markers. pTRG neurons projecting to the vagal motoneuronal pool were identified in a restricted area of the rostral rhombencephalon at the level of the isthmic Müller cell I1 close to sulcus limitans of His. Unilateral microinjections of lidocaine, muscimol, or glutamate antagonists into the pTRG inhibited completely the bilateral respiratory activity. In contrast, microinjections of glutamate agonists enhanced the respiratory activity, suggesting that this region is critical for the respiratory pattern generation. The retrogradely labeled pTRG neurons are glutamatergic and surrounded by terminals with intense substance P immunoreactivity. Cholinergic neurons were seen close to, and intermingled with, pTRG neurons. In addition, α-bungarotoxin binding sites (indicating nicotinic receptors) were found throughout the pTRG area and particularly on the soma of these neurons. During apnea, induced by blockade of ionotropic glutamate receptors within the same region, microinjections of 1 µm substance P or 1 mm nicotine into the pTRG restored rhythmic respiratory activity. The results emphasize the close similarities between the pTRG and the mammalian pre-Bötzinger complex as a crucial site for respiratory rhythmogenesis. We conclude that some basic features of the excitatory neurons proposed to generate respiratory rhythms are conserved throughout evolution.


Subject(s)
Neural Pathways/physiology , Neurons/physiology , Respiratory Center/cytology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Female , Functional Laterality , In Vitro Techniques , Lampreys , Male , Neural Pathways/anatomy & histology , Neurons/drug effects , Respiratory Center/metabolism , Substance P/metabolism , Substance P/pharmacology , Vagus Nerve/physiology
18.
J Neurosci ; 33(13): 5454-65, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23536061

ABSTRACT

Postsynaptic inhibition is a key element of neural circuits underlying behavior, with 20-50% of all mammalian (nongranule) neurons considered inhibitory. For rhythmic movements in mammals, e.g., walking, swimming, suckling, chewing, and breathing, inhibition is often hypothesized to play an essential rhythmogenic role. Here we study the role of fast synaptic inhibitory neurotransmission in the generation of breathing pattern by blocking GABA(A) and glycine receptors in the preBötzinger complex (preBötC), a site essential for generation of normal breathing pattern, and in the neighboring Bötzinger complex (BötC). The breathing rhythm continued following this blockade, but the lung inflation-induced Breuer-Hering inspiratory inhibitory reflex was suppressed. The antagonists were efficacious, as this blockade abolished the profound effects of the exogenously applied GABA(A) receptor agonist muscimol or glycine, either of which under control conditions stopped breathing in vagus-intact or vagotomized, anesthetized, spontaneously breathing adult rats. In vagotomized rats, GABA(A)ergic and glycinergic antagonists had little, if any, effect on rhythm. The effect in vagus-intact rats was to slow the rhythm to a pace equivalent to that seen after suppression of the aforementioned Breuer-Hering inflation reflex. We conclude that postsynaptic inhibition within the preBötC and BötC is not essential for generation of normal respiratory rhythm in intact mammals. We suggest the primary role of inhibition is in shaping the pattern of respiratory motor output, assuring its stability, and in mediating reflex or volitional apnea, but not in the generation of rhythm per se.


Subject(s)
Neural Inhibition/physiology , Respiration , Respiratory Center/cytology , Respiratory Center/physiology , Animals , Bicuculline/pharmacology , Choline O-Acetyltransferase/metabolism , Diaphragm/drug effects , Diaphragm/physiopathology , Electromyography , Functional Laterality , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Glycine/pharmacology , Glycine Agents/pharmacology , Indoles , Laryngeal Nerves/physiology , Male , Microinjections , Muscimol/pharmacology , Neural Inhibition/drug effects , Rats , Rats, Sprague-Dawley , Reflex/drug effects , Respiration/drug effects , Respiratory Center/drug effects , Respiratory Center/injuries , Strychnine/pharmacology , Vagotomy , Vagus Nerve/physiology
19.
J Comput Neurosci ; 34(2): 345-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23053862

ABSTRACT

Activity of neurons in the pre-Bötzinger complex (pre-BötC) within the mammalian brainstem drives the inspiratory phase of the respiratory rhythm. Experimental results have suggested that multiple bursting mechanisms based on a calcium-activated nonspecific cationic (CAN) current, a persistent sodium (NaP) current, and calcium dynamics may be incorporated within the pre-BötC. Previous modeling works have incorporated representations of some or all of these mechanisms. In this study, we consider a single-compartment model of a pre-BötC inspiratory neuron that encompasses particular aspects of all of these features. We present a novel mathematical analysis of the interaction of the corresponding rhythmic mechanisms arising in the model, including square-wave bursting and autonomous calcium oscillations, which requires treatment of a system of differential equations incorporating three slow variables.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Models, Neurological , Neurons/physiology , Respiratory Center/cytology , Animals , Humans , Mathematics
20.
Exp Physiol ; 98(2): 415-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22962286

ABSTRACT

In the present study, we used atomic force microscopy (AFM) to examine the ligand-binding properties of α7-containing nicotinic acetylcholine receptors (nAChRs) expressed on neurons from the ventral respiratory group. We also determined the effect of acute and prolonged exposure to nicotine on the binding probability of nAChRs. Neurons from neonatal (postnatal day 5-10) and juvenile rats (3-4 weeks old) were cultured. Internalization of Alexa Fluor 488-conjugated substance P was used to identify respiratory neurons that expressed the neurokinin-1 receptor (NK1-R), a recognized marker of ventral respiratory group neurons. To assess functional changes in nAChRs, AFM probes conjugated with anti-α7 subunit nAChR antibody were used to interact cyclically with the surface of the soma of NK1-R-positive neurons. Measurements were made of the frequency of antibody adhesion to the α7 receptor subunit and of the detachment forces between the membrane-attached receptor and the AFM probe tip. Addition of α-bungarotoxin (a specific antagonist of α7 subunit-containing nAChRs) to the cell bath produced a 69% reduction in binding to the α7 subunit (P < 0.05, n = 10), supporting specificity of binding. Acute exposure to nicotine (1 µM added to culture media) produced an 80% reduction in nAChR antibody binding to the α7 subunit (P < 0.05, n = 9). Prolonged incubation (72 h) of the cell culture in nicotine significantly reduced α7 binding in a concentration-dependent manner. Collectively, these findings demonstrate that AFM is a sensitive tool for assessment of functional changes in nAChRs expressed on the surface of living NK1-R-expressing medullary neurons. Moreover, these data demonstrate that nicotine exposure decreases the binding probability of α7 subunit-containing nAChRs.


Subject(s)
Microscopy, Atomic Force , Neurons/metabolism , Receptors, Neurokinin-1/metabolism , Receptors, Nicotinic/metabolism , Respiratory Center/metabolism , Age Factors , Animals , Animals, Newborn , Bungarotoxins/metabolism , Bungarotoxins/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Fluorescent Dyes/metabolism , Ligands , Male , Neurons/drug effects , Nicotine/metabolism , Nicotine/pharmacology , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/metabolism , Nicotinic Antagonists/pharmacology , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Respiratory Center/cytology , Respiratory Center/drug effects , Substance P/analogs & derivatives , Substance P/metabolism , Time Factors , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL