Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 452
Filtrar
1.
J Neurophysiol ; 132(1): 108-129, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38748514

RESUMEN

µ-Opioid receptors (MORs) are responsible for mediating both the analgesic and respiratory effects of opioid drugs. By binding to MORs in brainstem regions involved in controlling breathing, opioids produce respiratory depressive effects characterized by slow and shallow breathing, with potential cardiorespiratory arrest and death during overdose. To better understand the mechanisms underlying opioid-induced respiratory depression, thorough knowledge of the regions and cellular subpopulations that may be vulnerable to modulation by opioid drugs is needed. Using in situ hybridization, we determined the distribution and coexpression of Oprm1 (gene encoding MORs) mRNA with glutamatergic (Vglut2) and neurokinin-1 receptor (Tacr1) mRNA in medullary and pontine regions involved in breathing control and modulation. We found that >50% of cells expressed Oprm1 mRNA in the preBötzinger complex (preBötC), nucleus tractus solitarius (NTS), nucleus ambiguus (NA), postinspiratory complex (PiCo), locus coeruleus (LC), Kölliker-Fuse nucleus (KF), and the lateral and medial parabrachial nuclei (LBPN and MPBN, respectively). Among Tacr1 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, Bötzinger complex (BötC), PiCo, LC, raphe magnus nucleus, KF, LPBN, and MPBN, whereas among Vglut2 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, BötC, PiCo, LC, KF, LPBN, and MPBN. Taken together, our study provides a comprehensive map of the distribution and coexpression of Oprm1, Tacr1, and Vglut2 mRNA in brainstem regions that control and modulate breathing and identifies Tacr1 and Vglut2 mRNA-expressing cells as subpopulations with potential vulnerability to modulation by opioid drugs.NEW & NOTEWORTHY Opioid drugs can cause serious respiratory side-effects by binding to µ-opioid receptors (MORs) in brainstem regions that control breathing. To better understand the regions and their cellular subpopulations that may be vulnerable to modulation by opioids, we provide a comprehensive map of Oprm1 (gene encoding MORs) mRNA expression throughout brainstem regions that control and modulate breathing. Notably, we identify glutamatergic and neurokinin-1 receptor-expressing cells as potentially vulnerable to modulation by opioid drugs and worthy of further investigation using targeted approaches.


Asunto(s)
Receptores de Neuroquinina-1 , Receptores Opioides mu , Proteína 2 de Transporte Vesicular de Glutamato , Animales , Receptores Opioides mu/metabolismo , Receptores Opioides mu/genética , Receptores de Neuroquinina-1/metabolismo , Receptores de Neuroquinina-1/genética , Ratones , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Masculino , Tronco Encefálico/metabolismo , Tronco Encefálico/efectos de los fármacos , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , ARN Mensajero/genética , Centro Respiratorio/metabolismo , Centro Respiratorio/efectos de los fármacos
2.
Respir Physiol Neurobiol ; 296: 103810, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34728431

RESUMEN

Systemic 8-OH-DPAT (a 5-HT1A receptor agonist) challenge evokes hyperventilation independent of peripheral 5-HT1A receptors. Though the pre-Botzinger Complex (PBC) is critical in generating respiratory rhythm and activation of local 5-HT1A receptors induces tachypnea via disinhibition of local GABAA neurons, its role in the respiratory response to systemic 8-OH-DPAT challenge is still unclear. In anesthetized rats, 8-OH-DPAT (100 µg/kg, iv) was injected twice to confirm the reproducibility of the evoked responses. The same challenges were performed after bilateral microinjections of (S)-WAY-100135 (a 5-HT1A receptor antagonist) or gabazine (a GABAA receptor antagonist) into the PBC. Our results showed that: 1) 8-OH-DPAT caused reproducible hyperventilation associated with hypotension and bradycardia; 2) microinjections of (S)-WAY-100135 into the PBC attenuated the hyperventilation by ˜60 % without effect on the evoked hypotension and bradycardia; and 3) the same hyperventilatory attenuation was also observed after microinjections of gabazine into the PBC. Our data suggest that PBC 5-HT1A receptors play a key role in the respiratory response to systemic 8-OH-DPAT challenge likely via disinhibiting local GABAergic neurons.


Asunto(s)
8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Antagonistas del GABA/farmacología , Hiperventilación/inducido químicamente , Hiperventilación/tratamiento farmacológico , Bulbo Raquídeo/metabolismo , Receptor de Serotonina 5-HT1A/fisiología , Centro Respiratorio/metabolismo , Antagonistas del Receptor de Serotonina 5-HT1/farmacología , Agonistas de Receptores de Serotonina/farmacología , 8-Hidroxi-2-(di-n-propilamino)tetralin/administración & dosificación , Animales , Modelos Animales de Enfermedad , Masculino , Bulbo Raquídeo/efectos de los fármacos , Piperazinas/farmacología , Piridazinas/farmacología , Ratas , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Centro Respiratorio/efectos de los fármacos
3.
J Neurosci ; 41(48): 9919-9931, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34697095

RESUMEN

Death from opioid overdose is typically caused by opioid-induced respiratory depression (OIRD). A particularly dangerous characteristic of OIRD is its apparent unpredictability. The respiratory consequences of opioids can be surprisingly inconsistent, even within the same individual. Despite significant clinical implications, most studies have focused on average dose-r esponses rather than individual variation, and there remains little insight into the etiology of this apparent unpredictability. The preBötzinger complex (preBötC) in the ventral medulla is an important site for generating the respiratory rhythm and OIRD. Here, using male and female C57-Bl6 mice in vitro, we demonstrate that the preBötC can assume different network states depending on the excitability of the preBötC and the intrinsic membrane properties of preBötC neurons. These network states predict the functional consequences of opioids in the preBötC, and depending on network state, respiratory rhythmogenesis can be either stabilized or suppressed by opioids. We hypothesize that the dynamic nature of preBötC rhythmogenic properties, required to endow breathing with remarkable flexibility, also plays a key role in the dangerous unpredictability of OIRD.SIGNIFICANCE STATEMENT Opioids can cause unpredictable, life-threatening suppression of breathing. This apparent unpredictability makes clinical management of opioids difficult while also making it challenging to define the underlying mechanisms of OIRD. Here, we find in brainstem slices that the preBötC, an opioid-sensitive subregion of the brainstem, has an optimal configuration of cellular and network properties that results in a maximally stable breathing rhythm. These properties are dynamic, and the state of each individual preBötC network relative to the optimal configuration of the network predicts how vulnerable rhythmogenesis is to the effects of opioids. These insights establish a framework for understanding how endogenous and exogenous modulation of the rhythmogenic state of the preBötC can increase or decrease the risk of OIRD.


Asunto(s)
Analgésicos Opioides/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Cultivo de Órganos
4.
Elife ; 102021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34402425

RESUMEN

The analgesic utility of opioid-based drugs is limited by the life-threatening risk of respiratory depression. Opioid-induced respiratory depression (OIRD), mediated by the µ-opioid receptor (MOR), is characterized by a pronounced decrease in the frequency and regularity of the inspiratory rhythm, which originates from the medullary preBötzinger Complex (preBötC). To unravel the cellular- and network-level consequences of MOR activation in the preBötC, MOR-expressing neurons were optogenetically identified and manipulated in transgenic mice in vitro and in vivo. Based on these results, a model of OIRD was developed in silico. We conclude that hyperpolarization of MOR-expressing preBötC neurons alone does not phenocopy OIRD. Instead, the effects of MOR activation are twofold: (1) pre-inspiratory spiking is reduced and (2) excitatory synaptic transmission is suppressed, thereby disrupting network-driven rhythmogenesis. These dual mechanisms of opioid action act synergistically to make the normally robust inspiratory rhythm-generating network particularly prone to collapse when challenged with exogenous opioids.


Asunto(s)
Analgésicos Opioides/efectos adversos , Receptores Opioides mu/genética , Centro Respiratorio/efectos de los fármacos , Insuficiencia Respiratoria/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos , Receptores Opioides mu/metabolismo , Insuficiencia Respiratoria/inducido químicamente , Transmisión Sináptica/fisiología
5.
Respir Physiol Neurobiol ; 293: 103736, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34224867

RESUMEN

The preBötzinger complex (preBötC) is a medullary area essential for normal breathing and widely recognized as necessary and sufficient to generate the inspiratory phase of respiration. It has been studied mainly in rodents. Here we report the main results of our studies revealing the characteristics of the rabbit preBötC identified by means of neuronal recordings, D,L-homocysteic acid microinjections and histological controls. A crucial role in the respiratory rhythmogenesis within this neural substrate is played by excitatory amino acids, but also GABA and glycine display important contributions. Increases in respiratory frequency are induced by microinjections of neurokinins, somatostatin as well by serotonin (5-HT) through an action on 5-HT1A and 5-HT3 receptors or the disinhibition of a GABAergic circuit. Respiratory depression is observed in response to microinjections of the µ-opioid receptor agonist DAMGO. Our results show similarities and differences with the rodent preBötC and emphasize the importance of comparative studies on the mechanisms underlying respiratory rhythmogenesis in different animal species.


Asunto(s)
Generadores de Patrones Centrales/fisiología , Bulbo Raquídeo/fisiología , Neurotransmisores/farmacología , Centro Respiratorio/fisiología , Fenómenos Fisiológicos Respiratorios , Animales , Generadores de Patrones Centrales/efectos de los fármacos , Bulbo Raquídeo/efectos de los fármacos , Conejos , Centro Respiratorio/efectos de los fármacos , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos
6.
Anesth Analg ; 132(5): 1244-1253, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33857966

RESUMEN

Opioids are commonly used for pain management, perioperative procedures, and addiction treatment. There is a current opioid epidemic in North America that is paralleled by a marked increase in related deaths. Since 2000, chronic opioid users have been recognized to have significant central sleep apnea (CSA). After heart failure-related Cheyne-Stokes breathing (CSB), opioid-induced CSA is now the second most commonly seen CSA. It occurs in around 24% of chronic opioid users, typically after opioids have been used for more than 2 months, and usually corresponds in magnitude to opioid dose/plasma concentration. Opioid-induced CSA events often mix with episodes of ataxic breathing. The pathophysiology of opioid-induced CSA is based on dysfunction in respiratory rhythm generation and ventilatory chemoreflexes. Opioids have a paradoxical effect on different brain regions, which result in irregular respiratory rhythm. Regarding ventilatory chemoreflexes, chronic opioid use induces hypoxia that appears to stimulate an augmented hypoxic ventilatory response (high loop gain) and cause a narrow CO2 reserve, a combination that promotes respiratory instability. To date, no direct evidence has shown any major clinical consequence from CSA in chronic opioid users. A line of evidence suggested increased morbidity and mortality in overall chronic opioid users. CSA in chronic opioid users is likely to be a compensatory mechanism to avoid opioid injury and is potentially beneficial. The current treatments of CSA in chronic opioid users mainly focus on continuous positive airway pressure (CPAP) and adaptive servo-ventilation (ASV) or adding oxygen. ASV is more effective in reducing CSA events than CPAP. However, a recent ASV trial suggested an increased all-cause and cardiovascular mortality with the removal of CSA/CSB in cardiac failure patients. A major reason could be counteracting of a compensatory mechanism. No similar trial has been conducted for chronic opioid-related CSA. Future studies should focus on (1) investigating the phenotypes and genotypes of opioid-induced CSA that may have different clinical outcomes; (2) determining if CSA in chronic opioid users is beneficial or detrimental; and (3) assessing clinical consequences on different treatment options on opioid-induced CSA.


Asunto(s)
Analgésicos Opioides/efectos adversos , Encéfalo/efectos de los fármacos , Pulmón/inervación , Trastornos Relacionados con Opioides/complicaciones , Respiración/efectos de los fármacos , Apnea Central del Sueño/inducido químicamente , Encéfalo/fisiopatología , Humanos , Trastornos Relacionados con Opioides/mortalidad , Trastornos Relacionados con Opioides/fisiopatología , Pronóstico , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/fisiopatología , Medición de Riesgo , Factores de Riesgo , Apnea Central del Sueño/mortalidad , Apnea Central del Sueño/fisiopatología
7.
Commun Biol ; 3(1): 583, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33067579

RESUMEN

Hydrogen sulfide (H2S) is constitutively generated in the human body and works as a gasotransmitter in synaptic transmission. In this study, we aimed to evaluate the roles of endogenous H2S in generating eupnea at the respiratory center. We employed an in situ arterially perfused preparation of decerebrated rats and recorded the central respiratory outputs. When the H2S-producing enzyme cystathionine ß-synthase (CBS) was inhibited, respiration switched from the 3-phase eupneic pattern, which consists of inspiration, postinspiration, and expiration, to gasping-like respiration, which consists of inspiration only. On the other hand, when H2S synthesis was inhibited via cystathionine γ-lyase (CSE) or when H2S synthesis was activated via CBS, eupnea remained unchanged. These results suggest that H2S produced by CBS has crucial roles in maintaining the neuronal network to generate eupnea. The mechanism of respiratory pattern generation might be switched from a network-based system to a pacemaker cell-based system in low H2S conditions.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Centro Respiratorio/irrigación sanguínea , Centro Respiratorio/metabolismo , Animales , Seno Carotídeo/efectos de los fármacos , Seno Carotídeo/inervación , Seno Carotídeo/metabolismo , Cistationina betasintasa/antagonistas & inhibidores , Cistationina betasintasa/metabolismo , Desnervación , Ratas , Respiración , Centro Respiratorio/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo
8.
Physiology (Bethesda) ; 35(6): 391-404, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33052772

RESUMEN

Opioids depress minute ventilation primarily by reducing respiratory rate. This results from direct effects on the preBötzinger Complex as well as from depression of the Parabrachial/Kölliker-Fuse Complex, which provides excitatory drive to preBötzinger Complex neurons mediating respiratory phase-switch. Opioids also depress awake drive from the forebrain and chemodrive.


Asunto(s)
Analgésicos Opioides/efectos adversos , Neuronas/efectos de los fármacos , Neuronas/patología , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/patología , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/patología , Animales , Humanos
9.
Respir Physiol Neurobiol ; 280: 103482, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32553889

RESUMEN

The role of the different components of the respiratory network in the mediation of opioid-induced respiratory depression is still unclear. We investigated the contribution of the preBötzinger Complex (preBötC) and the neighbouring Bötzinger Complex (BötC) and inspiratory portion of the ventral respiratory group (iVRG) in anesthetized, vagotomized, paralyzed and artificially ventilated adult rabbits making use of bilateral microinjections (30-50 nl) of the µ-opioid receptor agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO). Dose-dependent effects were observed. In the preBötC and the BötC 0.1 mM DAMGO microinjections caused mainly reductions in peak phrenic amplitude associated with tonic phrenic activity and irregular (ataxic) patterns of breathing that were more pronounced in the preBötC. Apneic effects developed at 0.5 mM. In the iVRG DAMGO provoked decreases in amplitude and frequency of phrenic bursts at 0.1 mM and apnea at 0.5 mM. Local 5 mM naloxone reversed the apneic effects. The results imply that different components of the respiratory network may contribute to opioid-induced respiratory disorders.


Asunto(s)
Analgésicos Opioides/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Bulbo Raquídeo/efectos de los fármacos , Nervio Frénico/efectos de los fármacos , Receptores Opioides mu/metabolismo , Centro Respiratorio/efectos de los fármacos , Insuficiencia Respiratoria/inducido químicamente , Animales , Apnea/inducido químicamente , Apnea/fisiopatología , Bulbo Raquídeo/metabolismo , Microinyecciones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas , Nervio Frénico/fisiopatología , Conejos , Receptores Opioides mu/agonistas , Centro Respiratorio/metabolismo , Insuficiencia Respiratoria/fisiopatología
10.
Respir Physiol Neurobiol ; 277: 103401, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32036030

RESUMEN

The Kölliker-Fuse Nucleus (KF) has been widely investigated for its contribution to "inspiratory off-switch" while more recent studies showed that activation of the Parabrachial Nucleus (PBN) shortened expiratory duration. This study used an adult, in vivo, decerebrate rabbit model to delineate the contribution of each site to inspiratory and expiratory duration through sequential block of glutamatergic excitation with 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). Glutamatergic disfacilitation caused large increases in inspiratory and expiratory duration and minor decrease in peak phrenic activity (PPA). Hypoxia only partially reversed respiratory rate depression but PPA was increased to >200 % of control. The contribution of PBN activity to inspiratory and expiratory duration was equal while block of the KF affected inspiratory duration more than expiratory. We conclude that in the in vivo preparation respiratory rate greatly depends on PBN/KF activity, which contributes to the "inspiratory on- "and "off-switch", but is of minor importance for the magnitude of phrenic motor output.


Asunto(s)
Ácido Glutámico/fisiología , Núcleo de Kölliker-Fuse/fisiología , Núcleos Parabraquiales/fisiología , Centro Respiratorio/fisiología , Frecuencia Respiratoria/fisiología , Animales , Agonistas de Aminoácidos Excitadores/administración & dosificación , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Femenino , Núcleo de Kölliker-Fuse/efectos de los fármacos , Masculino , Microinyecciones/métodos , Núcleos Parabraquiales/efectos de los fármacos , Quinoxalinas/administración & dosificación , Conejos , Centro Respiratorio/efectos de los fármacos , Frecuencia Respiratoria/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/administración & dosificación
11.
Respir Physiol Neurobiol ; 274: 103383, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31923590

RESUMEN

We previously found that maternal cigarette smoke (CS) exposure resulted in impairment of central chemoreception and oxidative stress and mitochondrial dysfunction of parafacial respiratory group (pFRG, a critical site for mammalian central chemoreception) in neonatal rats. The present work was carried out to identify if maternal CS exposure could disturb the glutamate (GLU)-ergic and γ-aminobutyric acid (GABA)-ergic balance in pFRG of neonatal rats. We found that maternal CS exposure induced a decrease in GLU content and consequently in GLU/GABA ratio in pFRG of neonatal rats. Maternal CS exposure also decreased glutamine content and glutaminase and glutamine synthetase activity in offspring pFRG. In addition, expression of vesicular glutamate transporter 2 was depressed, and those of glutamate transporter 1 and GABA transporter 3 were elevated by maternal CS exposure. These results indicate that maternal CS exposure leads to a disturbance of GLU/GABA balance in pFRG of the neonatal rats, which might contribute to the suppression of central chemoreception in maternal CS-exposed offspring.


Asunto(s)
Relojes Biológicos/efectos de los fármacos , Células Quimiorreceptoras , Fumar Cigarrillos/efectos adversos , Ácido Glutámico/metabolismo , Bulbo Raquídeo , Efectos Tardíos de la Exposición Prenatal , Centro Respiratorio , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Células Quimiorreceptoras/efectos de los fármacos , Células Quimiorreceptoras/metabolismo , Femenino , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Sprague-Dawley , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/metabolismo
12.
J Physiol ; 598(1): 189-205, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31589332

RESUMEN

KEY POINTS: The main cause of death from opioid overdose is respiratory depression due to the activation of µ-opioid receptors (MORs). We conditionally deleted MORs from neurons in two key areas of the brainstem respiratory circuitry (the Kölliker-Fuse nucleus (KF) and pre-Bötzinger complex (preBötC)) to determine their role in opioid-induced respiratory disturbances in adult, awake mice. Deletion of MORs from KF neurons attenuated respiratory rate depression at all doses of morphine. Deletion of MORs from preBötC neurons attenuated rate depression at the low dose, but had no effect on rate following high doses of morphine. Instead, high doses of morphine increased the occurrence of apnoeas. The results indicate that opioids affect distributed key areas of the respiratory network in a dose-dependent manner and countering the respiratory effects of high dose opioids via the KF may be an effective approach to combat overdose. ABSTRACT: The primary cause of death from opioid overdose is respiratory failure. High doses of opioids cause severe rate depression and increased risk of fatal apnoea, which correlate with increasing irregularities in breathing pattern. µ-Opioid receptors (MORs) are widely distributed throughout the brainstem respiratory network, but the mechanisms underlying respiratory depression are poorly understood. The medullary pre-Bötzinger complex (preBötC) and the pontine Kölliker-Fuse nucleus (KF) are considered critical for inducing opioid-related respiratory disturbances. We used a conditional knockout approach to investigate the roles and relative contribution of MORs in KF and preBötC neurons in opioid-induced respiratory depression in awake adult mice. The results revealed dose-dependent and region-specific opioid effects on the control of both respiratory rate and pattern. Respiratory depression induced by an anti-nociceptive dose of morphine was significantly attenuated following deletion of MORs from either the KF or the preBötC, suggesting cumulative network effects on respiratory rate control at low opioid doses. Deletion of MORs from KF neurons also relieved rate depression at near-maximal respiratory depressant doses of morphine. Meanwhile, deletion of MORs from the preBötC had no effect on rate following administration of high doses of morphine. Instead, a severe ataxic breathing pattern emerged with many apnoeas. We conclude that opioids affect distributed areas of the respiratory network and opioid-induced respiratory depression cannot be attributed to only one area in isolation. However, countering the effects of near maximal respiratory depressant doses of opioids in the KF may be a powerful approach to combat opioid overdose.


Asunto(s)
Analgésicos Opioides/efectos adversos , Morfina/efectos adversos , Receptores Opioides mu/metabolismo , Centro Respiratorio/efectos de los fármacos , Insuficiencia Respiratoria/inducido químicamente , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Opioides mu/genética , Vigilia
13.
Elife ; 82019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804180

RESUMEN

The rhythm generating network for breathing must continuously adjust to changing metabolic and behavioral demands. Here, we examined network-based mechanisms in the mouse preBötzinger complex using substance P, a potent excitatory modulator of breathing frequency and stability, as a tool to dissect network properties that underlie dynamic breathing. We find that substance P does not alter the balance of excitation and inhibition during breaths or the duration of the resulting refractory period. Instead, mechanisms of recurrent excitation between breaths are enhanced such that the rate that excitation percolates through the network is increased. We propose a conceptual framework in which three distinct phases of inspiration, the burst phase, refractory phase, and percolation phase, can be differentially modulated to control breathing dynamics and stability. Unraveling mechanisms that support this dynamic control may improve our understanding of nervous system disorders that destabilize breathing, many of which involve changes in brainstem neuromodulatory systems.


Asunto(s)
Red Nerviosa/fisiología , Periodicidad , Respiración , Centro Respiratorio/fisiología , Animales , Ratones , Modelos Neurológicos , Red Nerviosa/efectos de los fármacos , Neurotransmisores/metabolismo , Centro Respiratorio/efectos de los fármacos , Sustancia P/metabolismo
14.
Elife ; 82019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31841107

RESUMEN

How mammalian neural circuits generate rhythmic activity in motor behaviors, such as breathing, walking, and chewing, remains elusive. For breathing, rhythm generation is localized to a brainstem nucleus, the preBötzinger Complex (preBötC). Rhythmic preBötC population activity consists of strong inspiratory bursts, which drive motoneuronal activity, and weaker burstlets, which we hypothesize reflect an emergent rhythmogenic process. If burstlets underlie inspiratory rhythmogenesis, respiratory depressants, such as opioids, should reduce burstlet frequency. Indeed, in medullary slices from neonatal mice, the µ-opioid receptor (µOR) agonist DAMGO slowed burstlet generation. Genetic deletion of µORs in a glutamatergic preBötC subpopulation abolished opioid-mediated depression, and the neuropeptide Substance P, but not blockade of inhibitory synaptic transmission, reduced opioidergic effects. We conclude that inspiratory rhythmogenesis is an emergent process, modulated by opioids, that does not rely on strong bursts of activity associated with motor output. These findings also point to strategies for ameliorating opioid-induced depression of breathing.


Asunto(s)
Analgésicos Opioides/farmacología , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/fisiología , Respiración/efectos de los fármacos , Centro Respiratorio/fisiología , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/agonistas , Proteínas de Homeodominio , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/fisiología , Receptores Opioides mu , Centro Respiratorio/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
15.
PLoS Comput Biol ; 15(8): e1006938, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31469828

RESUMEN

The mechanism(s) of action of most commonly used pharmacological blockers of voltage-gated ion channels are well understood; however, this knowledge is rarely considered when interpreting experimental data. Effects of blockade are often assumed to be equivalent, regardless of the mechanism of the blocker involved. Using computer simulations, we demonstrate that this assumption may not always be correct. We simulate the blockade of a persistent sodium current (INaP), proposed to underlie rhythm generation in pre-Bötzinger complex (pre-BötC) respiratory neurons, via two distinct pharmacological mechanisms: (1) pore obstruction mediated by tetrodotoxin and (2) altered inactivation dynamics mediated by riluzole. The reported effects of experimental application of tetrodotoxin and riluzole in respiratory circuits are diverse and seemingly contradictory and have led to considerable debate within the field as to the specific role of INaP in respiratory circuits. The results of our simulations match a wide array of experimental data spanning from the level of isolated pre-BötC neurons to the level of the intact respiratory network and also generate a series of experimentally testable predictions. Specifically, in this study we: (1) provide a mechanistic explanation for seemingly contradictory experimental results from in vitro studies of INaP block, (2) show that the effects of INaP block in in vitro preparations are not necessarily equivalent to those in more intact preparations, (3) demonstrate and explain why riluzole application may fail to effectively block INaP in the intact respiratory network, and (4) derive the prediction that effective block of INaP by low concentration tetrodotoxin will stop respiratory rhythm generation in the intact respiratory network. These simulations support a critical role for INaP in respiratory rhythmogenesis in vivo and illustrate the importance of considering mechanism when interpreting and simulating data relating to pharmacological blockade.


Asunto(s)
Modelos Neurológicos , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/inervación , Bloqueadores de los Canales de Sodio/farmacología , Animales , Biología Computacional , Simulación por Computador , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/fisiología , Sistema Respiratorio/metabolismo , Riluzol/farmacología , Canales de Sodio/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Tetrodotoxina/farmacología
16.
Elife ; 82019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30900989

RESUMEN

Neonatal inflammation is common and has lasting consequences for adult health. We investigated the lasting effects of a single bout of neonatal inflammation on adult respiratory control in the form of respiratory motor plasticity induced by acute intermittent hypoxia, which likely compensates and stabilizes breathing during injury or disease and has significant therapeutic potential. Lipopolysaccharide-induced inflammation at postnatal day four induced lasting impairments in two distinct pathways to adult respiratory plasticity in male and female rats. Despite a lack of adult pro-inflammatory gene expression or alterations in glial morphology, one mechanistic pathway to plasticity was restored by acute, adult anti-inflammatory treatment, suggesting ongoing inflammatory signaling after neonatal inflammation. An alternative pathway to plasticity was not restored by anti-inflammatory treatment, but was evoked by exogenous adenosine receptor agonism, suggesting upstream impairment, likely astrocytic-dependent. Thus, the respiratory control network is vulnerable to early-life inflammation, limiting respiratory compensation to adult disease or injury.


Asunto(s)
Adaptación Fisiológica , Enfermedades del Recién Nacido , Inflamación/complicaciones , Respiración , Centro Respiratorio/patología , Animales , Animales Recién Nacidos , Femenino , Humanos , Recién Nacido , Inflamación/inducido químicamente , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Masculino , Ratas Sprague-Dawley , Centro Respiratorio/efectos de los fármacos
17.
Respir Physiol Neurobiol ; 260: 37-52, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30502519

RESUMEN

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.


Asunto(s)
Ácido Glutámico/metabolismo , Neuronas/fisiología , Respiración , Centro Respiratorio/citología , Centro Respiratorio/fisiología , Mecánica Respiratoria/fisiología , Análisis de Varianza , Animales , Mapeo Encefálico , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Masculino , Microinyecciones , Neuronas/efectos de los fármacos , Periodicidad , Nervio Frénico , Conejos , Respiración/efectos de los fármacos , Centro Respiratorio/efectos de los fármacos , Mecánica Respiratoria/efectos de los fármacos
18.
Brain Res Bull ; 144: 39-45, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30448454

RESUMEN

Preinspiratory (Pre-I) neurons in the parafacial respiratory group (pFRG) comprise one of the respiratory rhythm generators in the medulla of the neonatal rat. A subgroup of pFRG/Pre-I neurons expresses the transcription factor Phox2b. To further analyze detailed neuronal mechanisms of respiratory rhythm generation in the neonatal rat, we developed a transgenic (Tg) rat line in which Phox2b-positive cells expressed archaerhodopsin-3 (Arch). Brainstem-spinal cord preparations were isolated from 0-2-day-old Tg newborn rats and were superfused with artificial cerebrospinal fluid equilibrated with 95% O2 and 5% CO2, pH 7.4, at 25-26 °C. Inspiratory fourth cervical ventral root (C4) activity was monitored, and membrane potentials of neurons in the pFRG including Pre-I and inspiratory neurons were recorded. Phox2b-positive cells in the Tg rats were essentially positive for enhanced green fluorescent protein (EGFP) signals (reporter for Arch) in the pFRG. Continuous photo-stimulation of the rostral ventral medulla for up to 90 s by covering the pFRG with green laser light (532 nm) induced a decrease of respiratory rate measured at C4 accompanied by membrane hyperpolarization of Phox2b-positive pFRG/Pre-I neurons. In contrast, Phox2b-negative inspiratory neurons were not hyperpolarized during the photo-stimulation. Our findings showed that Phox2b-expressing pFRG/Pre-I neurons are involved in the maintenance of the basic respiratory rhythm in neonatal rat.


Asunto(s)
Proteínas Arqueales/biosíntesis , Proteínas de Homeodominio/metabolismo , Red Nerviosa/metabolismo , Centro Respiratorio/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Proteínas Arqueales/genética , Tronco Encefálico/metabolismo , Proteínas de Homeodominio/biosíntesis , Masculino , Bulbo Raquídeo/metabolismo , Neuronas/metabolismo , Ratas , Ratas Transgénicas , Respiración , Centro Respiratorio/metabolismo , Médula Espinal/fisiología , Factores de Transcripción/biosíntesis
19.
Brain Res ; 1704: 26-39, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30244021

RESUMEN

Serotonin (5-HT) has been reported to play excitatory effects on respiration by acting on preBötzinger complex (preBötC) neurons in neonatal or juvenile rodents. However, whether its action is circumscribed to the preBötC and present in other animal species, particularly in adult preparations, is unknown. We investigated the respiratory role of 5-HT within the preBötC and neighbouring respiration-related regions. Experiments were performed on α-chloralose-urethane anesthetized, vagotomized, paralyzed and artificially ventilated rabbits making use of bilateral microinjections (30-50 nl). 5-HT caused excitatory effects on respiratory activity only when applied to the preBötC. These effects were mediated by 5-HT1A and 5-HT3 receptors as shown by microinjections of specific agonists of the different types of 5-HT receptors. Unexpectedly, the blockade of 5-HT1A receptors by methysergide or the specific antagonist (S)-WAY 100135 induced excitatory respiratory effects. Microinjections of the 5-HT3 receptor antagonist ondansetron did not influence respiration, but prevented (S)-WAY 100135-induced responses. The blockade of GABAA receptors by bicuculline within the preBötC prevented the effects of the 5-HT1A receptor agonist 8-OH-DPAT. The involvement of GABAergic inhibition and 5-HT1A receptor-mediated disinhibition is also corroborated by immunohistochemical data. The results show for the first time in an adult animal preparation that 5-HT plays a pivotal role in the modulation of the preBötC activity probably via both presynaptic and postsynaptic mechanisms and highlight the importance of disinhibition phenomena. Present findings may be relevant to some respiratory disorders in which an impairment of central 5-HT mechanisms has been reported, such as sleep apnoea and sudden infant death syndrome.


Asunto(s)
Neuronas/efectos de los fármacos , Respiración/efectos de los fármacos , Centro Respiratorio/efectos de los fármacos , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Antagonistas del Receptor de Serotonina 5-HT1/farmacología , Agonistas del Receptor de Serotonina 5-HT3/farmacología , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Animales , Masculino , Metisergida/farmacología , Microinyecciones , Ondansetrón/farmacología , Piperazinas/farmacología , Conejos
20.
Respir Physiol Neurobiol ; 265: 9-18, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30075288

RESUMEN

Inflammation inhibits the expression of some, but not all forms of respiratory motor plasticity. For example, systemic application of lipopolysaccharide (LPS) inhibits the phrenic long-term facilitation induced by moderate-intermittent hypoxia in vivo. There are multiple pro-inflammatory processes triggered by the systemic application of LPS, including neuroinflammation in the CNS. Considering that microglia can be activated by the systemic application of LPS, it is likely that this cell type influences the response of the respiratory circuits to intermittent hypoxia (IH). Thus, we aimed to test whether modulators of microglial function would affect the response to IH of the preBötzinger complex (preBötC) isolated in a brainstem slice preparation. This experimental approach avoids the systemic influences of these microglial modulators and limits their effects on cells, mostly microglia, included in the slice. First, we found that IH (3 × 5-min episodes of bubbling with 95% N2 and 5% CO2, mixed with 5-min normoxic intervals by bubbling with 95% O2 and 5% CO2) induces a long-lasting increase in the respiratory rhythm frequency recorded directly from the preBötC, called in vitro long-term facilitation (LTF), which occurs simultaneously with a long-lasting decrease in burst amplitude. Moreover, we found that bath applications of "microglial activators" (LPS and fractalkine), "microglial inhibitors" (minocycline and fucoidan) and a microgliotoxin (liposomal clodronate) partially reduce in vitro LTF. These findings reveal a complex scenario in which both the activation and the inhibition of microglia halts IH-induced preBötC plasticity and suggest that experimental or pathological conditions that affect this cell type, almost in any way, could affect breathing and its plastic responses.


Asunto(s)
Hipoxia , Factores Inmunológicos/farmacología , Inflamación , Microglía/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Periodicidad , Respiración/efectos de los fármacos , Centro Respiratorio/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Interneuronas/efectos de los fármacos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA