Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
Add more filters










Publication year range
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.
Sci Data ; 9(1): 457, 2022 07 30.
Article in English | MEDLINE | ID: mdl-35907922

ABSTRACT

Neurons in the brainstem preBötzinger complex (preBötC) generate the rhythm and rudimentary motor pattern for inspiratory breathing movements. We performed whole-cell patch-clamp recordings from inspiratory neurons in the preBötC of neonatal mouse slices that retain breathing-related rhythmicity in vitro. We classified neurons based on their electrophysiological properties and genetic background, and then aspirated their cellular contents for single-cell RNA sequencing (scRNA-seq). This data set provides the raw nucleotide sequences (FASTQ files) and annotated files of nucleotide sequences mapped to the mouse genome (mm10 from Ensembl), which includes the fragment counts, gene lengths, and fragments per kilobase of transcript per million mapped reads (FPKM). These data reflect the transcriptomes of the neurons that generate the rhythm and pattern for inspiratory breathing movements.


Subject(s)
Neurons , Respiratory Center , Transcriptome , Animals , Animals, Newborn , Mice , Neurons/physiology , Patch-Clamp Techniques , Respiration , Respiratory Center/cytology , Respiratory Center/physiology , Single-Cell Analysis
3.
Neurosci Res ; 143: 20-30, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29803764

ABSTRACT

The ventral respiratory column (VRC) generates rhythmical respiration and is divided into four compartments: the Bötzinger complex (BC), pre-Bötzinger complex (PBC), rostral ventral respiratory group (rVRG), and caudal ventral respiratory group (cVRG). Serotonergic nerve fibers are densely distributed in the rostral to caudal VRC and serotonin would be one of the important modulators for the respiratory control in the VRC. In the present study, to elucidate detailed distribution of serotonergic neurons in raphe nuclei projecting to the various rostrocaudal levels of VRC, we performed combination of retrograde tracing technique by cholera toxin B subunit (CTB) with immunohistochemistry for tryptophan hydroxylase 2 (TPH2). The double-immunoreactive neurons with CTB and TPH2 were distributed in the both rostral and caudal raphe nuclei, i.e. dorsal raphe nucleus, raphe magnus nucleus, gigantocellular reticular nucleus alpha and ventral parts, lateral paragigantocellular nucleus, parapyramidal area, raphe obscurus nucleus, and raphe pallidus nucleus. The distributions of double-immunoreactive neurons were similar among injection groups of BC, PBC, anterior rVRG, and posterior rVRG/cVRG. In conclusion, serotonergic neurons in both rostral and caudal raphe nuclei projected throughout the VRC and these serotonergic projections may contribute to respiratory responses to various environmental and vital changes.


Subject(s)
Raphe Nuclei/anatomy & histology , Raphe Nuclei/cytology , Respiratory Center/anatomy & histology , Respiratory Center/cytology , Serotonergic Neurons/cytology , Animals , Cholera Toxin/metabolism , Male , Medulla Oblongata/anatomy & histology , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Neural Pathways , Neuroanatomical Tract-Tracing Techniques , Raphe Nuclei/metabolism , Rats , Rats, Wistar , Respiratory Center/metabolism , Serotonergic Neurons/metabolism , Serotonin/metabolism , Tryptophan Hydroxylase/metabolism
4.
Respir Physiol Neurobiol ; 260: 37-52, 2019 02.
Article in English | MEDLINE | ID: mdl-30502519

ABSTRACT

Glutamate is the predominant excitatory neurotransmitter in the ventral respiratory column; however, the contribution of glutamatergic excitation in the individual subregions to respiratory rhythm generation has not been fully delineated. In an adult, in vivo, decerebrate rabbit model during conditions of mild hyperoxic hypercapnia we blocked glutamatergic excitation using the receptor antagonists 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (NBQX) and d(-)-2-amino-5-phosphonopentanoic acid (AP5). Disfacilitation of the preBötzinger Complex caused a decrease in inspiratory and expiratory duration as well as peak phrenic amplitude and ultimately apnea. Disfacilitation of the Bötzinger Complex caused a decrease in inspiratory and expiratory duration; subsequent disfacilitation of the preBötzinger Complex resulted in complete loss of the respiratory pattern but maintained tonic inspiratory activity. We conclude that glutamatergic drive to the preBötzinger Complex is essential for respiratory rhythm generation. Glutamatergic drive to the Bötzinger Complex significantly affects inspiratory and expiratory phase duration. Bötzinger Complex neurons are responsible for maintaining the silent expiratory phase of the phrenic neurogram.


Subject(s)
Glutamic Acid/metabolism , Neurons/physiology , Respiration , Respiratory Center/cytology , Respiratory Center/physiology , Respiratory Mechanics/physiology , Analysis of Variance , Animals , Brain Mapping , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Microinjections , Neurons/drug effects , Periodicity , Phrenic Nerve , Rabbits , Respiration/drug effects , Respiratory Center/drug effects , Respiratory Mechanics/drug effects
5.
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
6.
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
7.
J Comp Neurol ; 526(8): 1389-1402, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29473167

ABSTRACT

The preBötzinger Complex (preBötC), a compact medullary region essential for generating normal breathing rhythm and pattern, is the kernel of the breathing central pattern generator (CPG). Excitatory preBötC neurons in rats project to major breathing-related brainstem regions. Here, we provide a brainstem connectivity map in mice for both excitatory and inhibitory preBötC neurons. Using a genetic strategy to label preBötC neurons, we confirmed extensive projections of preBötC excitatory neurons within the brainstem breathing CPG including the contralateral preBötC, Bötzinger Complex (BötC), ventral respiratory group, nucleus of the solitary tract, parahypoglossal nucleus, parafacial region (RTN/pFRG or alternatively, pFL /pFV ), parabrachial and Kölliker-Füse nuclei, as well as major projections to the midbrain periaqueductal gray. Interestingly, preBötC inhibitory projections paralleled the excitatory projections. Moreover, we examined overlapping projections in the pons in detail and found that they targeted the same neurons. We further explored the direct anatomical link between the preBötC and suprapontine brain regions that may govern emotion and other complex behaviors that can affect or be affected by breathing. Forebrain efferent projections were sparse and restricted to specific nuclei within the thalamus and hypothalamus, with processes rarely observed in cortex, basal ganglia, or other limbic regions, e.g., amygdala or hippocampus. We conclude that the preBötC sends direct, presumably inspiratory-modulated, excitatory and inhibitory projections in parallel to distinct targets throughout the brain that generate and modulate breathing pattern and/or coordinate breathing with other behaviors, physiology, cognition, or emotional state.


Subject(s)
Efferent Pathways/physiology , Neural Inhibition/physiology , Neurons/physiology , Respiratory Center/cytology , Animals , Choline O-Acetyltransferase/metabolism , Forkhead Transcription Factors/metabolism , Glycine Plasma Membrane Transport Proteins/genetics , Glycine Plasma Membrane Transport Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Phosphopyruvate Hydratase/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , Repressor Proteins/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Transduction, Genetic
8.
Respir Physiol Neurobiol ; 250: 14-18, 2018 04.
Article in English | MEDLINE | ID: mdl-29414420

ABSTRACT

Expression of the transcription factor FOXP2 is linked to brain circuits that control motor function and speech. Investigation of FOXP2 protein expression in respiratory areas of the ponto-medullary brainstem of adult rat revealed distinct rostro-caudal expression gradients. A high density of FOXP2 immunoreactive nuclei was observed within the rostral pontine Kölliker-Fuse nucleus, compared to low densities in caudal pontine and rostral medullary respiratory nuclei, including the: (i) noradrenergic A5 and parafacial respiratory groups; (ii) Bötzinger and pre-Bötzinger complex and; (iii) rostral ventral respiratory group. Moderate densities of FOXP2 immunoreactive nuclei were observed in the caudal ventral respiratory group and the nucleus retroambiguus, with significant density levels found in the caudal half of the dorsal respiratory group and the hypoglossal pre-motor area lateral around calamus scriptorius. FOXP2 immunoreactivity was absent in all cranial nerve motor nuclei. We conclude that FOXP2 expression in respiratory brainstem areas selectively delineates laryngeal and hypoglossal pre-motor neuron populations essential for the generation of sound and voice.


Subject(s)
Brain Stem/anatomy & histology , Brain Stem/metabolism , Forkhead Transcription Factors/metabolism , Motor Neurons/metabolism , Animals , Neural Pathways/metabolism , Neurons/metabolism , Rats , Respiration , Respiratory Center/cytology , Respiratory Center/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/metabolism
9.
Respir Physiol Neurobiol ; 256: 43-49, 2018 10.
Article in English | MEDLINE | ID: mdl-28822818

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating disease leading to progressive motor neuron degeneration and death by ventilatory failure. In a rat model of ALS (SOD1G93A), phrenic long-term facilitation (pLTF) following acute intermittent hypoxia (AIH) is enhanced greater than expected at disease end-stage but the mechanism is unknown. We suggest that one trigger for this enhancement is motor neuron death itself. Intrapleural injections of cholera toxin B fragment conjugated to saporin (CTB-SAP) selectively kill respiratory motor neurons and mimic motor neuron death observed in SOD1G93A rats. This CTB-SAP model allows us to study the impact of respiratory motor neuron death on breathing without many complications attendant to ALS. Here, we tested the hypothesis that phrenic motor neuron death is sufficient to enhance pLTF. pLTF was assessed in anesthetized, paralyzed and ventilated Sprague Dawley rats 7 and 28 days following bilateral intrapleural injections of: 1) CTB-SAP (25 µg), or 2) un-conjugated CTB and SAP (control). CTB-SAP enhanced pLTF at 7 (CTB-SAP: 162 ±â€¯18%, n = 8 vs. Control: 63 ±â€¯3%; n = 8; p < 0.05), but not 28 days post-injection (CTB-SAP: 64 ±â€¯10%, n = 10 vs. Control: 60 ±â€¯13; n = 8; p > 0.05). Thus, pLTF at 7 (not 28) days post-CTB-SAP closely resembles pLTF in end-stage ALS rats, suggesting that processes unique to the early period of motor neuron death enhance pLTF. This project increases our understanding of respiratory plasticity and its implications for breathing in motor neuron disease.


Subject(s)
Cell Death/drug effects , Cholera Toxin/toxicity , Motor Neurons/drug effects , Phrenic Nerve/drug effects , Poisons/toxicity , Respiratory Center/cytology , Ribosome Inactivating Proteins, Type 1/toxicity , Action Potentials/drug effects , Action Potentials/physiology , Animals , Blood Gas Analysis , Blood Pressure/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Hypoxia/physiopathology , Male , Phrenic Nerve/physiology , Rats , Rats, Sprague-Dawley , Saporins , Time Factors
10.
eNeuro ; 5(6)2018.
Article in English | MEDLINE | ID: mdl-30627640

ABSTRACT

A subset of neurons in the retrotrapezoid nucleus (RTN) function as respiratory chemoreceptors by regulating depth and frequency of breathing in response to changes in tissue CO2/H+. The activity of chemosensitive RTN neurons is also subject to modulation by CO2/H+-dependent purinergic signaling. However, mechanisms contributing to purinergic regulation of RTN chemoreceptors are not entirely clear. Recent evidence suggests adenosine inhibits RTN chemoreception in vivo by activation of A1 receptors. The goal of this study was to characterize effects of adenosine on chemosensitive RTN neurons and identify intrinsic and synaptic mechanisms underlying this response. Cell-attached recordings from RTN chemoreceptors in slices from rat or wild-type mouse pups (mixed sex) show that exposure to adenosine (1 µM) inhibits chemoreceptor activity by an A1 receptor-dependent mechanism. However, exposure to a selective A1 receptor antagonist (8-cyclopentyl-1,3-dipropylxanthine, DPCPX; 30 nM) alone did not potentiate CO2/H+-stimulated activity, suggesting activation of A1 receptors does not limit chemoreceptor activity under these reduced conditions. Whole-cell voltage-clamp from chemosensitive RTN neurons shows that exposure to adenosine activated an inward rectifying K+ conductance, and at the network level, adenosine preferentially decreased frequency of EPSCs but not IPSCs. These results show that adenosine activation of A1 receptors inhibits chemosensitive RTN neurons by direct activation of a G-protein-regulated inward-rectifier K+ (GIRK)-like conductance, and presynaptically, by suppression of excitatory synaptic input to chemoreceptors.


Subject(s)
Adenosine/metabolism , Chemoreceptor Cells/physiology , Receptors, Purinergic P1/metabolism , Respiratory Center/cytology , Signal Transduction/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Adenosine/pharmacology , Animals , Animals, Newborn , Barium/pharmacology , Carbon Dioxide/pharmacology , Chemoreceptor Cells/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Neurotransmitter Agents/pharmacology , Potassium Channel Blockers/pharmacology , Purinergic Agents/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P1/genetics , Signal Transduction/drug effects , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
Elife ; 5: e13403, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26974345

ABSTRACT

This study focuses on computational and theoretical investigations of neuronal activity arising in the pre-Bötzinger complex (pre-BötC), a medullary region generating the inspiratory phase of breathing in mammals. A progressive increase of neuronal excitability in medullary slices containing the pre-BötC produces mixed-mode oscillations (MMOs) characterized by large amplitude population bursts alternating with a series of small amplitude bursts. Using two different computational models, we demonstrate that MMOs emerge within a heterogeneous excitatory neural network because of progressive neuronal recruitment and synchronization. The MMO pattern depends on the distributed neuronal excitability, the density and weights of network interconnections, and the cellular properties underlying endogenous bursting. Critically, the latter should provide a reduction of spiking frequency within neuronal bursts with increasing burst frequency and a dependence of the after-burst recovery period on burst amplitude. Our study highlights a novel mechanism by which heterogeneity naturally leads to complex dynamics in rhythmic neuronal populations.


Subject(s)
Interneurons/physiology , Respiratory Center/cytology , Ventral Thalamic Nuclei/cytology , Action Potentials , Animals , Biological Clocks , Inhalation , Mammals , Models, Neurological , Models, Theoretical , Nerve Net
SELECTION OF CITATIONS
SEARCH DETAIL
...