Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Neurophysiol ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38748514

ABSTRACT

µ-Opioid receptors (MORs) are responsible for mediating both the analgesic and respiratory effects of opioids 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 opioids is needed. Using in situ hybridization, we determined the distribution and co-expression 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), locus coeruleus (LC), Kölliker-Fuse nucleus (KF), and the lateral and medial parabrachial nuclei (LBPN and MPBN, respectively). Among Tacr1 mRNA-expressing cells, >50% co-expressed Oprm1 mRNA in the preBötC, NTS, NA, Bötzinger Complex (BötC), LC, raphe magnus nucleus, KF, LPBN, and MPBN, whereas among Vglut2 mRNA-expressing cells, >50% co-expressed Oprm1 mRNA in the preBötC, NTS, NA, BötC, LC, KF, LPBN, and MPBN. Taken together, our study provides a comprehensive map of the distribution and co-expression 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 opioids.

2.
Elife ; 122023 07 17.
Article in English | MEDLINE | ID: mdl-37458576

ABSTRACT

Rhythmic breathing is generated by neural circuits located in the brainstem. At its core is the preBötzinger Complex (preBötC), a region of the medulla, necessary for the generation of rhythmic breathing in mammals. The preBötC is comprised of various neuronal populations expressing neurokinin-1 receptors, the cognate G-protein-coupled receptor of the neuropeptide substance P (encoded by the tachykinin precursor 1 or Tac1). Neurokinin-1 receptors are highly expressed in the preBötC and destruction or deletion of neurokinin-1 receptor-expressing preBötC neurons severely impair rhythmic breathing. Although, the application of substance P to the preBötC stimulates breathing in rodents, substance P is also involved in nociception and locomotion in various brain regions, suggesting that Tac1 neurons found in the preBötC may have diverse functional roles. Here, we characterized the role of Tac1-expressing preBötC neurons in the generation of rhythmic breathing in vivo, as well as motor behaviors. Using a cre-lox recombination approach, we injected adeno-associated virus containing the excitatory channelrhodopsin-2 ChETA in the preBötC region of Tac1-cre mice. Employing a combination of histological, optogenetics, respiratory, and behavioral assays, we showed that stimulation of glutamatergic or Tac1 preBötC neurons promoted rhythmic breathing in both anesthetized and freely moving animals, but also triggered locomotion and overcame respiratory depression by opioid drugs. Overall, our study identified a population of excitatory preBötC with major roles in rhythmic breathing and behaviors.


Subject(s)
Receptors, Neurokinin-1 , Substance P , Mice , Animals , Receptors, Neurokinin-1/genetics , Neurons/physiology , Medulla Oblongata/physiology , Respiration , Respiratory Center/physiology , Mammals
3.
eNeuro ; 10(6)2023 06.
Article in English | MEDLINE | ID: mdl-37364996

ABSTRACT

Opioid drugs are widely used as analgesics but cause respiratory depression, a potentially lethal side effect with overdose, by acting on µ-opioid receptors (MORs) expressed in brainstem regions involved in the control of breathing. Although many brainstem regions have been shown to regulate opioid-induced respiratory depression, the types of neurons involved have not been identified. Somatostatin is a major neuropeptide found in brainstem circuits regulating breathing, but it is unknown whether somatostatin-expressing circuits regulate respiratory depression by opioids. We examined the coexpression of Sst (gene encoding somatostatin) and Oprm1 (gene encoding MORs) mRNAs in brainstem regions involved in respiratory depression. Interestingly, Oprm1 mRNA expression was found in the majority (>50%) of Sst-expressing cells in the preBötzinger Complex, the nucleus tractus solitarius, the nucleus ambiguus, and the Kölliker-Fuse nucleus. We then compared respiratory responses to fentanyl between wild-type and Oprm1 full knock-out mice and found that the lack of MORs prevented respiratory rate depression from occurring. Next, using transgenic knock-out mice lacking functional MORs specifically in Sst-expressing cells, we compared respiratory responses to fentanyl between control and the conditional knock-out mice. We found that respiratory rate depression by fentanyl was preserved when MORs were deleted only in Sst-expressing cells. Our results show that despite coexpression of Sst and Oprm1 in respiratory circuits and the importance of somatostatin-expressing cells in the regulation of breathing, these cells do not mediate opioid-induced respiratory rate depression. Instead, MORs found in respiratory cell populations other than Sst-expressing cells likely contribute to the respiratory effects of fentanyl.


Subject(s)
Fentanyl , Respiratory Insufficiency , Mice , Animals , Fentanyl/pharmacology , Analgesics, Opioid/pharmacology , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Neurons/metabolism , Mice, Knockout , Somatostatin/metabolism , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Respiratory Insufficiency/metabolism
4.
Pediatr Res ; 94(3): 971-978, 2023 09.
Article in English | MEDLINE | ID: mdl-37185965

ABSTRACT

BACKGROUND: Leptin augments central CO2 chemosensitivity and stabilizes breathing in adults. Premature infants have unstable breathing and low leptin levels. Leptin receptors are on CO2 sensitive neurons in the Nucleus Tractus Solitarius (NTS) and locus coeruleus (LC). We hypothesized that exogenous leptin improves hypercapnic respiratory response in newborn rats by improving central CO2 chemosensitivity. METHODS: In rats at postnatal day (p)4 and p21, hyperoxic and hypercapnic ventilatory responses, and pSTAT and SOCS3 protein expression in the hypothalamus, NTS and LC were measured before and after treatment with exogenous leptin (6 µg/g). RESULTS: Exogenous leptin increased the hypercapnic response in p21 but not in p4 rats (P ≤ 0.001). At p4, leptin increased pSTAT expression only in the LC, and SOCS3 expression in the NTS and LC; while at p21 pSTAT and SOCS3 levels were higher in the hypothalamus, NTS, and LC (P ≤ 0.05). CONCLUSIONS: We describe the developmental profile of the effect of exogenous leptin on CO2 chemosensitivity. Exogenous leptin does not augment central CO2 sensitivity during the first week of life in newborn rats. The translational implication of these findings is that low plasma leptin levels in premature infants may not be contributing to respiratory instability. IMPACT: Exogenous leptin does not augment CO2 sensitivity during the first week of life in newborn rats, similar to the developmental period when feeding behavior is resistant to leptin. Exogenous leptin increases CO2 chemosensitivity in newborn rats after the 3rd week of life and upregulates the expression of pSTAT and SOC3 in the hypothalamus, NTS and LC. Low plasma leptin levels in premature infants are unlikely contributors to respiratory instability via decreased CO2 sensitivity in premature infants. Thus, it is highly unlikely that exogenous leptin would alter this response.


Subject(s)
Carbon Dioxide , Leptin , Rats , Animals , Carbon Dioxide/metabolism , Animals, Newborn , Leptin/pharmacology , Hypercapnia , Respiration
5.
Front Physiol ; 14: 1043581, 2023.
Article in English | MEDLINE | ID: mdl-37089428

ABSTRACT

Opioid medications are the mainstay of pain management but present substantial side-effects such as respiratory depression which can be lethal with overdose. Most opioid drugs, such as fentanyl, act on opioid receptors such as the G-protein-coupled µ-opioid receptors (MOR). G-protein-coupled receptors activate pertussis toxin-sensitive G-proteins to inhibit neuronal activity. Binding of opioid ligands to MOR and subsequent activation G proteins ßγ is modulated by regulator of G-protein signaling (RGS). The roles of G-proteins ßγ and RGS in MOR-mediated inhibition of the respiratory network are not known. Using rodent models to pharmacologically modulate G-protein signaling, we aim to determine the roles of ßγ G-proteins and RGS4. We showed that inhibition of ßγ G-proteins using gallein perfused in the brainstem circuits regulating respiratory depression by opioid drugs results in complete reversal of respiratory depression. Blocking of RGS4 using CCG55014 did not change the respiratory depression induced by MOR activation despite co-expression of RGS4 and MORs in the brainstem. Our results suggest that neuronal inhibition by opioid drugs is mediated by G-proteins, but not by RGS4, which supports the concept that ßγ G-proteins could be molecular targets to develop opioid overdose antidotes without the risks of re-narcotization often found with highly potent opioid drugs. On the other hand, RGS4 mediates opioid analgesia, but not respiratory depression, and RGS4 may be molecular targets to develop pain therapies without respiratory liability.

7.
Handb Clin Neurol ; 188: 339-355, 2022.
Article in English | MEDLINE | ID: mdl-35965031

ABSTRACT

Opiates, such as morphine, and synthetic opioids, such as fentanyl, constitute a class of drugs acting on opioid receptors which have been used therapeutically and recreationally for centuries. Opioid drugs have strong analgesic properties and are used to treat moderate to severe pain, but also present side effects including opioid dependence, tolerance, addiction, and respiratory depression, which can lead to lethal overdose if not treated. This chapter explores the pathophysiology, the neural circuits, and the cellular mechanisms underlying opioid-induced respiratory depression and provides a translational perspective of the most recent research. The pathophysiology discussed includes the effects of opioid drugs on the respiratory system in patients, as well as the animal models used to identify underlying mechanisms. Using a combination of gene editing and pharmacology, the neural circuits and molecular pathways mediating neuronal inhibition by opioids are examined. By using pharmacology and neuroscience approaches, new therapies to prevent or reverse respiratory depression by opioid drugs have been identified and are currently being developed. Considering the health and economic burden associated with the current opioid epidemic, innovative research is needed to better understand the side effects of opioid drugs and to discover new therapeutic solutions to reduce the incidence of lethal overdoses.


Subject(s)
Analgesics, Opioid , Respiratory Insufficiency , Analgesics, Opioid/adverse effects , Animals , Humans , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy
8.
Elife ; 102021 03 15.
Article in English | MEDLINE | ID: mdl-33720013

ABSTRACT

An opioid epidemic is spreading in North America with millions of opioid overdoses annually. Opioid drugs, like fentanyl, target the mu opioid receptor system and induce potentially lethal respiratory depression. The challenge in opioid research is to find a safe pain therapy with analgesic properties but no respiratory depression. Current discoveries are limited by lack of amenable animal models to screen candidate drugs. Zebrafish (Danio rerio) is an emerging animal model with high reproduction and fast development, which shares remarkable similarity in their physiology and genome to mammals. However, it is unknown whether zebrafish possesses similar opioid system, respiratory and analgesic responses to opioids than mammals. In freely-behaving larval zebrafish, fentanyl depresses the rate of respiratory mandible movements and induces analgesia, effects reversed by µ-opioid receptor antagonists. Zebrafish presents evolutionary conserved mechanisms of action of opioid drugs, also found in mammals, and constitute amenable models for phenotype-based drug discovery.


When it comes to treating severe pain, a doctor's arsenal is somewhat limited: synthetic or natural opioids such as morphine, fentanyl or oxycodone are often one of the only options available to relieve patients. Yet these compounds can make breathing slower and shallower, quickly depriving the body of oxygen and causing death. This lethal side-effect is particularly devastating as opioids misuse has reached dangerously high levels in the United States, creating an 'opioid epidemic' which has claimed the lives of over 80,000 Americans in 2020. It is therefore crucial to find safer drugs that do not have this effect on breathing, but this research has been slowed down by the lack of animal models in which to study the effect of new compounds. Zebrafish are small freshwater fish that reproduce and develop fast, yet they are also remarkably genetically similar to mammals and feature a complex nervous system. However, it is not known whether the effect of opioids on zebrafish is comparable to mammals, and therefore whether these animals can be used to test new drugs for pain relief. To investigate this question, Zaig et al. exposed zebrafish larvae to fentanyl, showing that the fish then exhibited slower lower jaw movements ­ a sign of decreased breathing. The fish also could also tolerate a painful stimulus for longer, suggesting that this opioid does reduce pain in the animals. Together, these results point towards zebrafish and mammals sharing similar opioid responses, demonstrating that the fish could be used to test potential pain medications. The methods Zaig et al. have developed to establish these results could be harnessed to quickly assess large numbers of drug compounds, as well as decipher how pain emerges and can be stopped.


Subject(s)
Analgesia , Analgesics, Opioid/pharmacology , Respiration/drug effects , Respiratory Insufficiency/physiopathology , Zebrafish/physiology , Animals , Larva/drug effects , Larva/growth & development , Larva/physiology , Respiratory Insufficiency/chemically induced , Zebrafish/growth & development
9.
Sleep Med Rev ; 58: 101441, 2021 08.
Article in English | MEDLINE | ID: mdl-33567395

ABSTRACT

We conducted a systematic review to address limited evidence suggesting that opioids may induce or aggravate obstructive sleep apnea (OSA). All clinical trials or observational studies on adults from 1946 to 2018 found through MEDLINE, EMBASE, CINAHL, PsycINFO, Cochrane Databases were eligible. We assessed the quality of the studies using published guidelines. Fifteen studies (six clinical trials and nine observational) with only two of good quality were included. Fourteen studies investigated the impact of opioids on the presence or severity of OSA, four addressed the effects of treatment for OSA  in opioid users, and none explored the consequences of opioid use in individuals with OSA. Eight of 14 studies found no significant relationship between opioid use or dose and apnea-hypopnea index (AHI) or degree of nocturnal desaturation. A random-effects meta-analysis (n = 10) determined the pooled mean change in AHI associated with opioid use of 1.47/h (-2.63-5.57; I2 = 65%). Three of the four studies found that continuous positive airway pressure (CPAP) therapy reduced AHI by 17-30/h in opioid users with OSA. Bilevel therapy with a back-up rate and adaptive servo-ventilation (ASV) without mandatory pressure support successfully normalized AHI (≤5) in opioid users. Limited by a paucity of good-quality studies, our review did not show a significant relationship between opioid use and the severity of OSA. There was some evidence that CPAP, Bilevel therapy, and ASV alleviate OSA for opioid users, with higher failure rates observed in patients on CPAP in opioid users.


Subject(s)
Analgesics, Opioid , Sleep Apnea, Obstructive , Adult , Analgesics, Opioid/adverse effects , Continuous Positive Airway Pressure , Humans , Sleep Apnea, Obstructive/therapy
10.
Sci Rep ; 10(1): 550, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953471

ABSTRACT

Motoneurons are the final output pathway for the brain's influence on behavior. Here we identify properties of hypoglossal motor output to the tongue musculature. Tongue motor control is critical to the pathogenesis of obstructive sleep apnea, a common and serious sleep-related breathing disorder. Studies were performed on mice expressing a light sensitive cation channel exclusively on cholinergic neurons (ChAT-ChR2(H134R)-EYFP). Discrete photostimulations under isoflurane-induced anesthesia from an optical probe positioned above the medullary surface and hypoglossal motor nucleus elicited discrete increases in tongue motor output, with the magnitude of responses dependent on stimulation power (P < 0.001, n = 7) and frequency (P = 0.002, n = 8, with responses to 10 Hz stimulation greater than for 15-25 Hz, P < 0.022). Stimulations during REM sleep elicited significantly reduced responses at powers 3-20 mW compared to non-rapid eye movement (non-REM) sleep and wakefulness (each P < 0.05, n = 7). Response thresholds were also greater in REM sleep (10 mW) compared to non-REM and waking (3 to 5 mW, P < 0.05), and the slopes of the regressions between input photostimulation powers and output motor responses were specifically reduced in REM sleep (P < 0.001). This study identifies that variations in photostimulation input produce tunable changes in hypoglossal motor output in-vivo and identifies REM sleep specific suppression of net motor excitability and responsivity.


Subject(s)
Channelrhodopsins/genetics , Choline O-Acetyltransferase/genetics , Hypoglossal Nerve/physiology , Motor Neurons/physiology , Tongue/innervation , Animals , Bacterial Proteins/genetics , Isoflurane/administration & dosage , Luminescent Proteins/genetics , Male , Mice , Mice, Transgenic , Sleep, REM , Tongue/physiology , Wakefulness/physiology
11.
Sci Rep ; 9(1): 14122, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31575947

ABSTRACT

Opioid drugs are the mainstay of pain management but present the side-effect of respiratory depression that can be lethal with overdose. In addition to their respiratory effect, opioids also induce a profound sedative state and produce electrocortical features characteristic of a state of reduced brain arousal, similar to anaesthesia or sleep. In such states, respiratory activity depends more on the integrity of the brainstem respiratory network than it does during wakefulness. Accordingly, we propose that sedation by fentanyl induces specific electrocortical changes consistent with reduced brain arousal, and that the magnitude of respiratory depression is associated with distinct electrocortical changes. To these aims, we determined the effects of systemic injections of fentanyl (dosage 100 µg ·kg) versus control on electrocortical  and respiratory activities of freely-behaving rats. We found that fentanyl induced electrocortical changes that differed from those observed in sleep or wakefulness. Fentanyl increased δ (1-3 Hz) frequency power (P < 0.001), but reduced α (7.5-13.5 Hz) and ß2 (20-30 Hz) powers (P = 0.012 and P < 0.001, respectively), when compared to wakefulness. Interestingly, respiratory rate depression by fentanyl was significantly correlated with increased θ power (R = 0.61, P < 0.001), therefore showing a clear association between electrocortical activity and the magnitude of respiratory rate depression. Overall, we provide new evidence linking specific electrocortical changes to the severity of respiratory depression by opioids, which highlights the importance of considering the cortical and subcortical effects of opioids in addition to their impacts on breathing when evaluating opioid-induced respiratory depression.


Subject(s)
Analgesics, Opioid/pharmacology , Fentanyl/pharmacology , Respiratory Insufficiency/drug therapy , Respiratory Rate/drug effects , Anesthesia/methods , Animals , Arousal/drug effects , Brain Stem/drug effects , Male , Rats , Rats, Wistar
13.
Nature ; 562(7727): 419-422, 2018 10.
Article in English | MEDLINE | ID: mdl-30305735

ABSTRACT

Dysfunctional breathing is the main cause of morbidity and mortality after traumatic injury of the cervical spinal cord1,2 and often necessitates assisted ventilation, thus stressing the need to develop strategies to restore breathing. Cervical interneurons that form synapses on phrenic motor neurons, which control the main inspiratory muscle, can modulate phrenic motor output and diaphragmatic function3-5. Here, using a combination of pharmacogenetics and respiratory physiology assays in different models of spinal cord injury, we show that mid-cervical excitatory interneurons are essential for the maintenance of breathing in mice with non-traumatic cervical spinal cord injury, and are also crucial for promoting respiratory recovery after traumatic spinal cord injury. Although these interneurons are not necessary for breathing under normal conditions, their stimulation in non-injured animals enhances inspiratory amplitude. Immediately after spinal cord injury, pharmacogenetic stimulation of cervical excitatory interneurons restores respiratory motor function. Overall, our results demonstrate a strategy to restore breathing after central nervous system trauma by targeting a neuronal subpopulation.


Subject(s)
Interneurons/physiology , Respiration , Spinal Cord Injuries/physiopathology , Animals , Diaphragm/innervation , Diaphragm/physiology , Female , Inhalation/physiology , Interneurons/metabolism , Mice , Motor Neurons/physiology
14.
Sci Rep ; 7(1): 18105, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273726

ABSTRACT

Persistent and stable respiratory activity across behavioral states is key to homeostasis. Extrasynaptic δ-subunit containing GABAA receptors (δGABAARs) mediate tonic inhibition and regulate network activity. However, the influence of δGABAARs on respiratory rhythm and motor outputs is unknown. We manipulated extra-synaptic GABAA receptor function in the preBötzinger Complex (preBötC), a site central to the generation of inspiratory motor activity in mammals. Activation of preBötC δGABAARs in anesthetized rats and wild-type mice decreased breathing rate. In δGABAAR knockout (Gabrd -/-) mice, however, δGABAARs activation had no effect on breathing rate. We then found that during active wakefulness associated with behaviors and movements, diaphragm activation was higher in the Gabrd -/- compared to wild-type mice, but not in other states. These findings identify that δGABAARs modulate the respiratory network, which is critical to understand how δGABAARs change breathing in pathological conditions affecting extra-synaptic GABAA receptor function such as exposure to anesthetics and neurosteroids.


Subject(s)
Medulla Oblongata/physiology , Neck Muscles/physiology , Neurons/physiology , Receptors, GABA-A/metabolism , Respiratory Rate/physiology , Animals , Behavior, Animal/physiology , Mice , Mice, Knockout , Rats
15.
Epilepsia ; 58(9): 1637-1644, 2017 09.
Article in English | MEDLINE | ID: mdl-28691204

ABSTRACT

OBJECTIVE: Sudden unexplained death in epilepsy is the leading cause of death in young adult epilepsy patients, typically occurring during the early postictal period, presumably resulting from brainstem and cardiorespiratory dysfunction. We hypothesized that ictal discharges in the brainstem disrupt the cardiorespiratory network, causing mortality. To study this hypothesis, we chose an animal model comprising focal unilateral hippocampal injection of 4-aminopyridine (4-AP), which produced focal recurrent hippocampal seizures with secondary generalization in awake, behaving rats. METHODS: We studied ictal and interictal intracranial electrographic activity (iEEG) in 23 rats implanted with a custom electrode array into the hippocampus, the contralateral cortex, and brainstem. The hippocampal electrodes contained a cannula to administer the potassium channel blocker and convulsant (4-AP). iEEG was recorded continuously before, during, and after seizures induced by 4-AP infusion into the hippocampus. RESULTS: The control group (n = 5) was monitored for 2-3 months, and the weekly baseline iEEG recordings showed long-term stability. The low-dose group (1 µL 4-AP, 40 mm, n = 5) exhibited local electrographic seizures without spread to the contralateral cerebral cortex or brainstem. The high-dose group (5 µL 4-AP, 40 mm, n = 3) had several hippocampal electrographic seizures, which spread contralaterally and triggered brainstem discharges within 40 min, and were associated with violent motor seizures followed by dyspnea and respiratory arrest, with cortical and hippocampal iEEG flattening. The group that received high-dose 4-AP without brainstem implantation (n = 5) had similar seizure-related respiratory difficulties. Finally, five rats that received high-dose 4-AP without EEG recording also developed violent motor seizures with postictal respiratory arrest. Following visualized respiratory arrest in groups III, IV, and V, manual respiratory resuscitation was successful in five of 13 animals. SIGNIFICANCE: These studies show that hippocampal seizure activity can spread or trigger brainstem epileptiform discharges that may cause mortality, possibly mediated by respiratory network dysfunction.


Subject(s)
4-Aminopyridine/pharmacology , Brain Stem/drug effects , Hippocampus/drug effects , Seizures/chemically induced , Animals , Electroencephalography/drug effects , Male , Rats , Rats, Wistar , Recurrence , Seizures/mortality
16.
Sci Rep ; 7: 45860, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28383527

ABSTRACT

Reduced tongue muscle tone precipitates obstructive sleep apnea (OSA), and activation of the tongue musculature can lessen OSA. The hypoglossal motor nucleus (HMN) innervates the tongue muscles but there is no pharmacological agent currently able to selectively manipulate a channel (e.g., Kir2.4) that is highly restricted in its expression to cranial motor pools such as the HMN. To model the effect of manipulating such a restricted target, we introduced a "designer" receptor into the HMN and selectively modulated it with a "designer" drug. We used cre-dependent viral vectors (AAV8-hSyn-DIO-hM3Dq-mCherry) to transduce hypoglossal motoneurons of ChAT-Cre+ mice with hM3Dq (activating) receptors. We measured sleep and breathing in three conditions: (i) sham, (ii) after systemic administration of clozapine-N-oxide (CNO; 1 mg/kg) or (iii) vehicle. CNO activates hM3Dq receptors but is otherwise biologically inert. Systemic administration of CNO caused significant and sustained increases in tongue muscle activity in non-REM (261 ± 33% for 10 hrs) and REM sleep (217 ± 21% for 8 hrs), both P < 0.01 versus controls. Responses were specific and selective for the tongue with no effects on diaphragm or postural muscle activities, or sleep-wake states. These results support targeting a selective and restricted "druggable" target at the HMN (e.g., Kir2.4) to activate tongue motor activity during sleep.


Subject(s)
Sleep Apnea, Obstructive/physiopathology , Sleep/physiology , Tongue/physiology , Animals , Clozapine/administration & dosage , Clozapine/analogs & derivatives , Diaphragm/innervation , Diaphragm/physiology , Efferent Pathways/drug effects , Efferent Pathways/physiology , Electromyography , Facial Muscles/innervation , Facial Muscles/physiology , Hypoglossal Nerve/physiology , Motor Neurons/physiology , Rats , Rats, Wistar , Serotonin/metabolism , Sleep/drug effects , Sleep Apnea, Obstructive/drug therapy , Tongue/drug effects , Tongue/innervation , Wakefulness/drug effects , Wakefulness/physiology
17.
Best Pract Res Clin Anaesthesiol ; 31(4): 469-485, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29739536

ABSTRACT

The increasing use of opioids in the perioperative period has increased opioid-associated morbidity and mortality. There is a well-established connection between opioids, sleep-disordered breathing (SDB), and respiratory depression. The treatment of postoperative pain with opioids in patients with SDB may result in respiratory depression. In an unmonitored setting, it may lead to life-threatening respiratory events. More studies are required to evaluate the effective management and prevention of respiratory depression in patients with SDB. This review summarizes the current state of knowledge relating to the pathophysiology of respiratory depression by opioids and opioid-related respiratory depression and appraises the association between opioids and SDB.


Subject(s)
Analgesics, Opioid/adverse effects , Pain, Postoperative/prevention & control , Respiratory Insufficiency/chemically induced , Sleep Apnea Syndromes/chemically induced , Analgesics, Opioid/administration & dosage , Animals , Brain/drug effects , Brain/physiology , Humans , Nerve Net/drug effects , Nerve Net/physiology , Pain, Postoperative/diagnosis , Pain, Postoperative/physiopathology , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/physiopathology , Sleep Apnea Syndromes/diagnosis , Sleep Apnea Syndromes/physiopathology
18.
Sci Rep ; 6: 32707, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27599866

ABSTRACT

Breathing is generated by a respiratory network in the brainstem. At its core, a population of neurons expressing neurokinin-1 receptors (NK1R) and the peptide somatostatin (SST) form the preBötzinger Complex (preBötC), a site essential for the generation of breathing. PreBötC interneurons generate rhythm and follower neurons shape motor outputs by activating upper airway respiratory muscles. Since NK1R-expressing preBötC neurons are preferentially inhibited by µ-opioid receptors via activation of GIRK channels, NK1R stimulation may also involve GIRK channels. Hence, we identify the contribution of GIRK channels to rhythm, motor output and respiratory modulation by NK1Rs and SST. In adult rats, GIRK channels were identified in NK1R-expressing preBötC cells. Their activation decreased breathing rate and genioglossus muscle activity, an important upper airway muscle. NK1R activation increased rhythmic breathing and genioglossus muscle activity in wild-type mice, but not in mice lacking GIRK2 subunits (GIRK2(-/-)). Conversely, SST decreased rhythmic breathing via SST2 receptors, reduced genioglossus muscle activity likely through SST4 receptors, but did not involve GIRK channels. In summary, NK1R stimulation of rhythm and motor output involved GIRK channels, whereas SST inhibited rhythm and motor output via two SST receptor subtypes, therefore revealing separate circuits mediating rhythm and motor output.


Subject(s)
Brain Stem/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Receptors, Neurokinin-1/physiology , Somatostatin/physiology , Animals , Immunohistochemistry , Male , Mice , Rats , Rats, Wistar , Respiration
19.
Anesthesiology ; 125(5): 889-903, 2016 11.
Article in English | MEDLINE | ID: mdl-27541315

ABSTRACT

BACKGROUND: Opioid analgesia is an essential component of perioperative care, but effective analgesia can be limited by excessive sedation and respiratory depression. The cortical signatures associated with sedation by opioids and the relationship between changes in cortical activity and respiratory function are not well understood. The objectives of this study were to identify the electroencephalogram signatures of sedation and respiratory changes induced by morphine in a pediatric population after elective surgery. METHODS: After otologic surgery, patients (14.8 ± 2.8 yr, n = 10) stayed overnight for pain relief with morphine (3 to 10 mg), hydration, and clinical observation. Electroencephalogram activity and polysomnography were performed before and after morphine, and electroencephalogram spectral properties and cardiorespiratory activities were analyzed. RESULTS: Compared to wakefulness and non-rapid eye movement sleep, morphine reduced high-frequency ß1 (13.5 to 20 Hz) and ß2 (20 to 30Hz) electroencephalogram powers (n = 10) and decreased coherence between frontal and occipital ß2 electroencephalogram activities (n = 9), therefore indicating that morphine induced a deep sedative state. Morphine also reduced respiratory rate by 8.3% (n = 10). Interestingly, there was a significant correlation between the reduction in ß1 electroencephalogram activity and the depression in respiratory rate induced by morphine (R = 0.715, n = 10). With significant reduction in ß1 power, respiratory rate was decreased by more than 25%, suggesting that reduction in cortical arousal is associated with the severity of respiratory rate depression. CONCLUSIONS: Analgesic doses of morphine are associated with reduction in respiratory rate when accompanied by reduction in ß1 electroencephalogram power, indicating a powerful effect of cortical arousal state per se in respiratory rate depression by morphine.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , Consciousness/drug effects , Morphine/pharmacology , Respiratory Insufficiency/physiopathology , Respiratory Rate/drug effects , Adolescent , Child , Child, Preschool , Electroencephalography/drug effects , Female , Humans , Male , Polysomnography/drug effects
20.
Anesthesiology ; 124(3): 641-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26675532

ABSTRACT

BACKGROUND: Drugs acting on µ-opioid receptors (MORs) are widely used as analgesics but present side effects including life-threatening respiratory depression. MORs are G-protein-coupled receptors inhibiting neuronal activity through calcium channels, adenylyl cyclase, and/or G-protein-gated inwardly rectifying potassium (GIRK) channels. The pathways underlying MOR-dependent inhibition of rhythmic breathing are unknown. METHODS: By using a combination of genetic, pharmacological, and physiological tools in rodents in vivo, the authors aimed to identify the role of GIRK channels in MOR-mediated inhibition of respiratory circuits. RESULTS: GIRK channels were expressed in the ventrolateral medulla, a neuronal population regulating rhythmic breathing, and GIRK channel activation with flupirtine reduced respiratory rate in rats (percentage of baseline rate in mean ± SD: 79.4 ± 7.4%, n = 7), wild-type mice (82.6 ± 3.8%, n = 3), but not in mice lacking the GIRK2 subunit, an integral subunit of neuronal GIRK channels (GIRK2, 101.0 ± 1.9%, n = 3). Application of the MOR agonist [D-Ala, N-MePhe, Gly-ol]-enkephalin (DAMGO) to the ventrolateral medulla depressed respiratory rate, an effect partially reversed by the GIRK channel blocker Tertiapin-Q (baseline: 42.1 ± 7.4 breath/min, DAMGO: 26.1 ± 13.4 breath/min, Tertiapin-Q + DAMGO: 33.9 ± 9.8 breath/min, n = 4). Importantly, DAMGO applied to the ventrolateral medulla failed to reduce rhythmic breathing in GIRK2 mice (percentage of baseline rate: 103.2 ± 12.1%, n = 4), whereas it considerably reduced rate in wild-type mice (62.5 ± 17.7% of baseline, n = 4). Respiratory rate depression by systemic injection of the opioid analgesic fentanyl was markedly reduced in GIRK2 (percentage of baseline: 12.8 ± 15.8%, n = 5) compared with wild-type mice (72.9 ± 27.3%). CONCLUSIONS: Overall, these results identify that GIRK channels contribute to respiratory inhibition by MOR, an essential step toward understanding respiratory depression by opioids.


Subject(s)
Analgesics, Opioid/toxicity , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/metabolism , Animals , Bee Venoms/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/toxicity , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , Male , Mice , Mice, Knockout , Rats , Rats, Wistar , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...