Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(50): e2212195119, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36469762

RESUMEN

Biological supramolecular assemblies, such as phospholipid bilayer membranes, have been used to demonstrate signal processing via short-term synaptic plasticity (STP) in the form of paired pulse facilitation and depression, emulating the brain's efficiency and flexible cognitive capabilities. However, STP memory in lipid bilayers is volatile and cannot be stored or accessed over relevant periods of time, a key requirement for learning. Using droplet interface bilayers (DIBs) composed of lipids, water and hexadecane, and an electrical stimulation training protocol featuring repetitive sinusoidal voltage cycling, we show that DIBs displaying memcapacitive properties can also exhibit persistent synaptic plasticity in the form of long-term potentiation (LTP) associated with capacitive energy storage in the phospholipid bilayer. The time scales for the physical changes associated with the LTP range between minutes and hours, and are substantially longer than previous STP studies, where stored energy dissipated after only a few seconds. STP behavior is the result of reversible changes in bilayer area and thickness. On the other hand, LTP is the result of additional molecular and structural changes to the zwitterionic lipid headgroups and the dielectric properties of the lipid bilayer that result from the buildup of an increasingly asymmetric charge distribution at the bilayer interfaces.


Asunto(s)
Potenciación a Largo Plazo , Fosfolípidos , Potenciación a Largo Plazo/fisiología , Fosfolípidos/química , Membrana Dobles de Lípidos/química , Plasticidad Neuronal/fisiología , Agua/química
2.
Physiology (Bethesda) ; 36(4): 203-219, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34159803

RESUMEN

Obtundation of wakefulness caused by opioids and loss of wakefulness caused by anesthetics and sleep significantly alter concentrations of molecules comprising the prefrontal cortex (PFC) metabolome. Quantifying state-selective changes in the PFC metabolome is essential for advancing functional metabolomics. Diverse functions of the PFC suggest the PFC metabolome as a potential therapeutic entry point for countermeasures to state-selective autonomic dysfunction.


Asunto(s)
Analgésicos Opioides , Anestesia , Humanos , Metaboloma , Corteza Prefrontal , Sueño
3.
J Neurophysiol ; 126(4): 1265-1275, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34469699

RESUMEN

The electroencephalogram (EEG) provides an objective, neural correlate of consciousness. Opioid receptors modulate mammalian neuronal excitability, and this fact was used to characterize how opioids administered to mice alter EEG power and states of consciousness. The present study tested the hypothesis that antinociceptive doses of fentanyl, morphine, or buprenorphine differentially alter the EEG and states of sleep and wakefulness in adult, male C57BL/6J mice. Mice were anesthetized and implanted with telemeters that enabled wireless recordings of cortical EEG and electromyogram (EMG). After surgical recovery, EEG and EMG were used to objectively score states of consciousness as wakefulness, rapid eye movement (REM) sleep, or non-REM (NREM) sleep. Measures of EEG power (dB) were quantified as δ (0.5-4 Hz), θ (4-8 Hz), α (8-13 Hz), σ (12-15 Hz), ß (13-30 Hz), and γ (30-60 Hz). Compared with saline (control), fentanyl and morphine decreased NREM sleep, morphine eliminated REM sleep, and buprenorphine eliminated NREM sleep and REM sleep. Opioids significantly and differentially disrupted the temporal organization of sleep/wake states, altered specific EEG frequency bands, and caused dissociated states of consciousness. The results are discussed relative to the fact that opioids, pain, and sleep modulate interacting states of consciousness.NEW & NOTEWORTHY This study discovered that antinociceptive doses of fentanyl, morphine, and buprenorphine significantly and differentially disrupt EEG-defined states of consciousness in C57BL/6J mice. These data are noteworthy because: 1) buprenorphine is commonly used in medication-assisted therapy for opioid addiction, and 2) there is evidence that disordered sleep can promote addiction relapse. The results contribute to community phenotyping efforts by making publicly available all descriptive and inferential statistics from this study (Supplemental Tables S1-S8).


Asunto(s)
Analgésicos Opioides/farmacología , Analgésicos/farmacología , Ondas Encefálicas/efectos de los fármacos , Buprenorfina/farmacología , Estado de Conciencia/efectos de los fármacos , Trastornos Disociativos/inducido químicamente , Electrocorticografía/efectos de los fármacos , Fentanilo/farmacología , Morfina/farmacología , Fases del Sueño/efectos de los fármacos , Vigilia/efectos de los fármacos , Analgésicos/administración & dosificación , Analgésicos Opioides/administración & dosificación , Animales , Buprenorfina/administración & dosificación , Modelos Animales de Enfermedad , Electroencefalografía , Electromiografía , Fentanilo/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación
4.
J Neurophysiol ; 125(5): 1899-1919, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33826874

RESUMEN

Opioid-induced respiratory depression (OIRD) represents the primary cause of death associated with therapeutic and recreational opioid use. Within the United States, the rate of death from opioid abuse since the early 1990s has grown disproportionally, prompting the classification as a nationwide "epidemic." Since this time, we have begun to unravel many fundamental cellular and systems-level mechanisms associated with opioid-related death. However, factors such as individual vulnerability, neuromodulatory compensation, and redundancy of opioid effects across central and peripheral nervous systems have created a barrier to a concise, integrative view of OIRD. Within this review, we bring together multiple perspectives in the field of OIRD to create an overarching viewpoint of what we know, and where we view this essential topic of research going forward into the future.


Asunto(s)
Analgésicos Opioides/farmacología , Generadores de Patrones Centrales/efectos de los fármacos , Bulbo Raquídeo/efectos de los fármacos , Trastornos Relacionados con Opioides/complicaciones , Insuficiencia Respiratoria/inducido químicamente , Analgésicos Opioides/efectos adversos , Animales , Humanos
5.
J Neurophysiol ; 124(6): 2012-2021, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33112692

RESUMEN

Identifying similarities and differences in the brain metabolome during different states of consciousness has broad relevance for neuroscience and state-dependent autonomic function. This study focused on the prefrontal cortex (PFC) as a brain region known to modulate states of consciousness. Anesthesia was used as a tool to eliminate wakefulness. Untargeted metabolomic analyses were performed on microdialysis samples obtained from mouse PFC during wakefulness and during isoflurane anesthesia. Analyses detected 2,153 molecules, 91 of which could be identified. Analytes were grouped as detected during both wakefulness and anesthesia (n = 61) and as unique to wakefulness (n = 23) or anesthesia (n = 7). Data were analyzed using univariate and multivariate approaches. Relative to wakefulness, during anesthesia there was a significant (q < 0.0001) fourfold change in 21 metabolites. During anesthesia 11 of these 21 molecules decreased and 10 increased. The Kyoto Encyclopedia of Genes and Genomes database was used to relate behavioral state-specific changes in the metabolome to metabolic pathways. Relative to wakefulness, most of the amino acids and analogs measured were significantly decreased during isoflurane anesthesia. Nucleosides and analogs were significantly increased during anesthesia. Molecules associated with carbohydrate metabolism, maintenance of lipid membranes, and normal cell functions were significantly decreased during anesthesia. Significant state-specific changes were also discovered among molecules comprising lipids and fatty acids, monosaccharides, and organic acids. Considered together, these molecules regulate point-to-point transmission, volume conduction, and cellular metabolism. The results identify a novel ensemble of candidate molecules in PFC as putative modulators of wakefulness and the loss of wakefulness.NEW & NOTEWORTHY The loss of wakefulness caused by a single concentration of isoflurane significantly altered levels of interrelated metabolites in the prefrontal cortex. The results support the interpretation that states of consciousness reflect dynamic interactions among cortical neuronal networks involving a humbling number of molecules that comprise the brain metabolome.


Asunto(s)
Anestesia , Anestésicos por Inhalación/farmacología , Estado de Conciencia/efectos de los fármacos , Isoflurano/farmacología , Metaboloma/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Vigilia/efectos de los fármacos , Anestésicos por Inhalación/administración & dosificación , Animales , Cromatografía Liquida , Isoflurano/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Microdiálisis , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Espectrometría de Masas en Tándem
6.
J Neurophysiol ; 123(6): 2285-2296, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32347157

RESUMEN

This study quantified eight small-molecule neurotransmitters collected simultaneously from prefrontal cortex of C57BL/6J mice (n = 23) during wakefulness and during isoflurane anesthesia (1.3%). Using isoflurane anesthesia as an independent variable enabled evaluation of the hypothesis that isoflurane anesthesia differentially alters concentrations of multiple neurotransmitters and their interactions. Machine learning was applied to reveal higher order interactions among neurotransmitters. Using a between-subjects design, microdialysis was performed during wakefulness and during anesthesia. Concentrations (nM) of acetylcholine, adenosine, dopamine, GABA, glutamate, histamine, norepinephrine, and serotonin in the dialysis samples are reported (means ± SD). Relative to wakefulness, acetylcholine concentration was lower during isoflurane anesthesia (1.254 ± 1.118 vs. 0.401 ± 0.134, P = 0.009), and concentrations of adenosine (29.456 ± 29.756 vs. 101.321 ± 38.603, P < 0.001), dopamine (0.0578 ± 0.0384 vs. 0.113 ± 0.084, P = 0.036), and norepinephrine (0.126 ± 0.080 vs. 0.219 ± 0.066, P = 0.010) were higher during anesthesia. Isoflurane reconfigured neurotransmitter interactions in prefrontal cortex, and the state of isoflurane anesthesia was reliably predicted by prefrontal cortex concentrations of adenosine, norepinephrine, and acetylcholine. A novel finding to emerge from machine learning analyses is that neurotransmitter concentration profiles in mouse prefrontal cortex undergo functional reconfiguration during isoflurane anesthesia. Adenosine, norepinephrine, and acetylcholine showed high feature importance, supporting the interpretation that interactions among these three transmitters may play a key role in modulating levels of cortical and behavioral arousal.NEW & NOTEWORTHY This study discovered that interactions between neurotransmitters in mouse prefrontal cortex were altered during isoflurane anesthesia relative to wakefulness. Machine learning further demonstrated that, relative to wakefulness, higher order interactions among neurotransmitters were disrupted during isoflurane administration. These findings extend to the neurochemical domain the concept that anesthetic-induced loss of wakefulness results from a disruption of neural network connectivity.


Asunto(s)
Acetilcolina/metabolismo , Adenosina/metabolismo , Anestesia , Anestésicos por Inhalación/farmacología , Isoflurano/farmacología , Aprendizaje Automático , Red Nerviosa , Norepinefrina/metabolismo , Corteza Prefrontal , Inconsciencia/metabolismo , Vigilia/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Microdiálisis , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología
7.
Anesthesiology ; 128(5): 984-991, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29394163

RESUMEN

BACKGROUND: Opiate-induced respiratory depression is sexually dimorphic and associated with increased risk among the obese. The mechanisms underlying these associations are unknown. The present study evaluated the two-tailed hypothesis that sex, leptin status, and obesity modulate buprenorphine-induced changes in breathing. METHODS: Mice (n = 40 male and 40 female) comprising four congenic lines that differ in leptin signaling and body weight were injected with saline and buprenorphine (0.3 mg/kg). Whole-body plethysmography was used to quantify the effects on minute ventilation. The data were evaluated using three-way analysis of variance, regression, and Poincaré analyses. RESULTS: Relative to B6 mice with normal leptin, buprenorphine decreased minute ventilation in mice with diet-induced obesity (37.2%; P < 0.0001), ob/ob mice that lack leptin (62.6%; P < 0.0001), and db/db mice with dysfunctional leptin receptors (65.9%; P < 0.0001). Poincaré analyses showed that buprenorphine caused a significant (P < 0.0001) collapse in minute ventilation variability that was greatest in mice with leptin dysfunction. There was no significant effect of sex or body weight on minute ventilation. CONCLUSIONS: The results support the interpretation that leptin status but not body weight or sex contributed to the buprenorphine-induced decrease in minute ventilation. Poincaré plots illustrate that the buprenorphine-induced decrease in minute ventilation variability was greatest in mice with impaired leptin signaling. This is relevant because normal respiratory variability is essential for martialing a compensatory response to ventilatory challenges imposed by disease, obesity, and surgical stress.


Asunto(s)
Analgésicos Opioides/efectos adversos , Buprenorfina/efectos adversos , Leptina/fisiología , Obesidad/fisiopatología , Insuficiencia Respiratoria/inducido químicamente , Transducción de Señal/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Factores Sexuales
8.
Eur J Neurosci ; 40(1): 2264-73, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24674578

RESUMEN

The oral part of the pontine reticular formation (PnO) contributes to the regulation of sleep, anesthesia and pain. The role of PnO γ-aminobutyric acid (GABA) in modulating these states remains incompletely understood. The present study used time to loss and time to resumption of righting response (LoRR and RoRR) as surrogate measures of loss and resumption of consciousness. This study tested three hypotheses: (i) pharmacologically manipulating GABA levels in rat PnO alters LoRR, RoRR and nociception; (ii) propofol decreases GABA levels in the PnO; and (iii) inhibiting GABA synthesis in the PnO blocks hyperalgesia caused by sleep deprivation. Administering a GABA synthesis inhibitor [3-mercaptopropionic acid (3-MPA)] or a GABA uptake inhibitor [nipecotic acid (NPA)] into rat PnO significantly altered LoRR caused by propofol. 3-MPA significantly decreased LoRR for propofol (-18%). NPA significantly increased LoRR during administration of propofol (36%). Neither 3-MPA nor NPA altered RoRR following cessation of propofol or isoflurane delivery. The finding that LoRR was decreased by 3-MPA and increased by NPA is consistent with measures showing that extracellular GABA levels in the PnO were decreased (41%) by propofol. Thermal nociception was significantly decreased by 3-MPA and increased by NPA, and 3-MPA blocked the hyperalgesia caused by sleep deprivation. The results demonstrate that GABA levels in the PnO regulate the time for loss of consciousness caused by propofol, extend the concept that anesthetic induction and emergence are not inverse processes, and suggest that GABAergic transmission in the PnO mediates hyperalgesia caused by sleep loss.


Asunto(s)
Anestésicos Generales/farmacología , Tegmento Pontino/efectos de los fármacos , Tegmento Pontino/fisiopatología , Privación de Sueño/fisiopatología , Ácido gamma-Aminobutírico/metabolismo , Ácido 3-Mercaptopropiónico/farmacología , Animales , Estado de Conciencia/efectos de los fármacos , Estado de Conciencia/fisiología , GABAérgicos/farmacología , Inhibidores de Recaptación de GABA/farmacología , Calor , Hiperalgesia/fisiopatología , Isoflurano/farmacología , Masculino , Ácidos Nipecóticos/farmacología , Nocicepción/efectos de los fármacos , Nocicepción/fisiología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Propofol/farmacología , Ratas Sprague-Dawley , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/fisiología , Factores de Tiempo
9.
Anesth Analg ; 118(6): 1293-300, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24842176

RESUMEN

BACKGROUND: Agonist binding at the benzodiazepine site of γ-aminobutric acid type A receptors diminishes anxiety and insomnia by actions in the amygdala. The neurochemical effects of benzodiazepine site agonists remain incompletely understood. Cholinergic neurotransmission modulates amygdala function, and this study tested the hypothesis that benzodiazepine site agonists alter acetylcholine (ACh) release in the amygdala. METHODS: Microdialysis and high-performance liquid chromatography quantified ACh release in the amygdala of Sprague-Dawley rats (n = 33). ACh was measured before and after IV administration (3 mg/kg) of midazolam or eszopiclone, with and without anesthesia. ACh in isoflurane-anesthetized rats during dialysis with Ringer's solution (control) was compared with ACh release during dialysis with Ringer's solution containing (100 µM) midazolam, diazepam, eszopiclone, or zolpidem. RESULTS: In unanesthetized rats, ACh in the amygdala was decreased by IV midazolam (-51.1%; P = 0.0029; 95% confidence interval [CI], -73.0% to -29.2%) and eszopiclone (-39.6%; P = 0.0222; 95% CI, -69.8% to -9.3%). In anesthetized rats, ACh in the amygdala was decreased by IV administration of midazolam (-46.2%; P = 0.0041; 95% CI, -67.9% to -24.5%) and eszopiclone (-34.0%; P = 0.0009; 95% CI, -44.7% to -23.3%), and increased by amygdala delivery of diazepam (43.2%; P = 0.0434; 95% CI, 2.1% to 84.3%) and eszopiclone (222.2%; P = 0.0159; 95% CI, 68.5% to 375.8%). CONCLUSIONS: ACh release in the amygdala was decreased by IV delivery of midazolam and eszopiclone. Dialysis delivery directly into the amygdala caused either increased (eszopiclone and diazepam) or likely no significant change (midazolam and zolpidem) in ACh release. These contrasting effects of delivery route on ACh release support the interpretation that systemically administered midazolam and eszopiclone decrease ACh release in the amygdala by acting on neuronal systems outside the amygdala.


Asunto(s)
Acetilcolina/metabolismo , Amígdala del Cerebelo/metabolismo , Agonistas del GABA/farmacología , Receptores de GABA-A/efectos de los fármacos , Amígdala del Cerebelo/efectos de los fármacos , Anestesia por Inhalación , Anestésicos por Inhalación , Animales , Compuestos de Azabiciclo/farmacología , Cromatografía Líquida de Alta Presión , Diazepam/farmacología , Eszopiclona , Agonistas del GABA/administración & dosificación , Inyecciones Intravenosas , Isoflurano , Masculino , Microdiálisis , Midazolam/farmacología , Piperazinas/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Zolpidem
10.
Anesthesiology ; 118(2): 327-36, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23263018

RESUMEN

BACKGROUND: Clinical and preclinical data demonstrate the analgesic actions of adenosine. Central administration of adenosine agonists, however, suppresses arousal and breathing by poorly understood mechanisms. This study tested the two-tailed hypothesis that adenosine A1 receptors in the pontine reticular formation (PRF) of C57BL/6J mice modulate breathing, behavioral arousal, and PRF acetylcholine release. METHODS: Three sets of experiments used 51 mice. First, breathing was measured by plethysmography after PRF microinjection of the adenosine A1 receptor agonist N-sulfophenyl adenosine (SPA) or saline. Second, mice were anesthetized with isoflurane and the time to recovery of righting response (RoRR) was quantified after a PRF microinjection of SPA or saline. Third, acetylcholine release in the PRF was measured before and during microdialysis delivery of SPA, the adenosine A1 receptor antagonist 1, 3-dipropyl-8-cyclopentylxanthine, or SPA and 1, 3-dipropyl-8-cyclopentylxanthine. RESULTS: First, SPA significantly decreased respiratory rate (-18%), tidal volume (-12%), and minute ventilation (-16%). Second, SPA concentration accounted for 76% of the variance in RoRR. Third, SPA concentration accounted for a significant amount of the variance in acetylcholine release (52%), RoRR (98%), and breathing rate (86%). 1, 3-dipropyl-8-cyclopentylxanthine alone caused a concentration-dependent increase in acetylcholine, a decrease in RoRR, and a decrease in breathing rate. Coadministration of SPA and 1, 3-dipropyl-8-cyclopentylxanthine blocked the SPA-induced decrease in acetylcholine and increase in RoRR. CONCLUSIONS: Endogenous adenosine acting at adenosine A1 receptors in the PRF modulates breathing, behavioral arousal, and acetylcholine release. The results support the interpretation that an adenosinergic-cholinergic interaction within the PRF comprises one neurochemical mechanism underlying the wakefulness stimulus for breathing.


Asunto(s)
Acetilcolina/metabolismo , Periodo de Recuperación de la Anestesia , Puente/metabolismo , Receptor de Adenosina A1/efectos de los fármacos , Respiración/efectos de los fármacos , Formación Reticular/metabolismo , Agonistas del Receptor de Adenosina A1/farmacología , Antagonistas del Receptor de Adenosina A1/farmacología , Anestesia , Animales , Nivel de Alerta/fisiología , Cromatografía Líquida de Alta Presión , Condicionamiento Operante/efectos de los fármacos , Electroquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microdiálisis , Microinyecciones , Puente/efectos de los fármacos , Equilibrio Postural/efectos de los fármacos , Reflejo/efectos de los fármacos , Formación Reticular/efectos de los fármacos
11.
J Neurosci ; 31(7): 2649-56, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21325533

RESUMEN

Studies using drugs that increase or decrease GABAergic transmission suggest that GABA in the pontine reticular formation (PRF) promotes wakefulness and inhibits rapid eye movement (REM) sleep. Cholinergic transmission in the PRF promotes REM sleep, and levels of endogenous acetylcholine (ACh) in the PRF are significantly greater during REM sleep than during wakefulness or non-REM (NREM) sleep. No previous studies have determined whether levels of endogenous GABA in the PRF vary as a function of sleep and wakefulness. This study tested the hypothesis that GABA levels in cat PRF are greatest during wakefulness and lowest during REM sleep. Extracellular GABA levels were measured during wakefulness, NREM sleep, REM sleep, and the REM sleep-like state (REM(Neo)) caused by microinjecting neostigmine into the PRF. GABA levels varied significantly as a function of sleep and wakefulness, and decreased significantly below waking levels during REM sleep (-42%) and REM(Neo) (-63%). The decrease in GABA levels during NREM sleep (22% below waking levels) was not statistically significant. Compared with NREM sleep, GABA levels decreased significantly during REM sleep (-27%) and REM(Neo) (-52%). Comparisons of REM sleep and REM(Neo) revealed no differences in GABA levels or cortical EEG power. GABA levels did not vary significantly as a function of dialysis site within the PRF. The inverse relationship between changes in PRF levels of GABA and ACh during REM sleep indicates that low GABAergic tone combined with high cholinergic tone in the PRF contributes to the generation of REM sleep.


Asunto(s)
Formación Reticular/metabolismo , Sueño REM/fisiología , Vigilia/fisiología , Ácido gamma-Aminobutírico/metabolismo , Acetilcolina/metabolismo , Animales , Gatos , Cromatografía Líquida de Alta Presión/métodos , Electroencefalografía/métodos , Masculino , Microdiálisis/métodos , Factores de Tiempo
12.
Anesth Analg ; 114(2): 297-302, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22167771

RESUMEN

BACKGROUND: Morbid obesity affects the pharmacokinetics and pharmacodynamics of anesthetics, which may result in inappropriate dosing. We hypothesized that obesity significantly alters the minimum alveolar concentration (MAC) for isoflurane and sevoflurane. To test this hypothesis, we used a rodent model of human metabolic syndrome developed through artificial selection for inherent low aerobic capacity runners (LCR) and high aerobic capacity runners (HCR). The LCR rats are obese, display phenotypes homologous to those characteristic of human metabolic syndrome, and exhibit low running endurance. In contrast, HCR rats have high running endurance and are characterized by improved cardiovascular performance and overall health. METHODS: Male and female LCR (n = 10) and HCR (n = 10) rats were endotracheally intubated and maintained on mechanical ventilation with either isoflurane or sevoflurane. A bracketing design was used to determine MAC; sensory stimulation was induced by tail clamping. An equilibration period of 30 minutes was provided before and between the consecutive tail clamps. Two-tailed parametric (unpaired t test) and nonparametric (Mann-Whitney test) statistics were used for the comparison of MAC between LCR and HCR rats. The data are reported as mean ± sd along with the 95% confidence interval. A P value of <0.05 was considered statistically significant. RESULTS: The MAC for isoflurane in LCR rats (1.52% ± 0.13%) was similar to previously reported isoflurane-MAC for normal rats (1.51% ± 0.12%). The HCR rats showed a significantly higher isoflurane-MAC (1.90% ± 0.19%) than did the LCR rats (1.52% ± 0.13%) (P = 0.0001). The MAC for sevoflurane was not significantly different between LCR and HCR rats and was similar to the previously published sevoflurane-MAC for normal rats (2.4% ± 0.30%). There was no influence of sex on the MAC of either isoflurane or sevoflurane. CONCLUSION: Obesity and associated comorbidities do not affect anesthetic requirements as measured by MAC in a rodent model of metabolic syndrome. By contrast, high aerobic capacity is associated with a higher MAC for isoflurane and may be a risk factor for subtherapeutic dosing.


Asunto(s)
Anestésicos por Inhalación/farmacología , Isoflurano/farmacocinética , Síndrome Metabólico/metabolismo , Éteres Metílicos/farmacocinética , Obesidad/metabolismo , Alveolos Pulmonares/metabolismo , Administración por Inhalación , Anestésicos por Inhalación/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Isoflurano/administración & dosificación , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/fisiopatología , Éteres Metílicos/administración & dosificación , Obesidad/genética , Obesidad/fisiopatología , Umbral del Dolor/efectos de los fármacos , Resistencia Física/genética , Ratas , Sevoflurano , Cola (estructura animal)/inervación
13.
Respir Physiol Neurobiol ; 303: 103924, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35662641

RESUMEN

Opioids impair many functions modulated by the prefrontal cortex (PFC), including wakefulness, cognition, and breathing. In contrast, cholinergic activity in the PFC increases wakefulness. This study tested the hypothesis that microinjecting the opioid fentanyl and the acetylcholinesterase inhibitor neostigmine into the PFC of awake C57BL/6J male mice (n = 27) alters breathing. The lateral and medial PFC were unilaterally microinjected with saline (control) and fentanyl. The medial PFC received additional microinjections of neostigmine. The results show that fentanyl caused site-specific changes in breathing. Fentanyl delivered to the lateral PFC significantly decreased minute ventilation variability, whereas fentanyl delivered to the medial PFC significantly increased tidal volume and duty cycle. Neostigmine microinjected into the medial PFC significantly increased respiratory rate, tidal volume, and minute ventilation. A final series of experiments revealed that decreased minute ventilation caused by systemic fentanyl administration was mitigated by PFC microinjection of neostigmine.


Asunto(s)
Fentanilo , Neostigmina , Acetilcolinesterasa , Analgésicos Opioides/farmacología , Animales , Fentanilo/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neostigmina/farmacología , Corteza Prefrontal
14.
Respir Physiol Neurobiol ; 297: 103834, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34954128

RESUMEN

The opioid buprenorphine alters breathing and the cytokine leptin stimulates breathing. Obesity increases the risk for respiratory disorders and can lead to leptin resistance. This study tested the hypothesis that buprenorphine causes dose-dependent changes in breathing that vary as a function of obesity, leptin status, and sex. Breathing measures were acquired from four congenic mouse lines: female and male wild type C57BL/6J (B6) mice, obese db/db and ob/ob mice with leptin dysfunction, and male B6 mice with diet-induced obesity. Mice were injected intraperitoneally with saline (control) and five doses of buprenorphine (0.1, 0.3, 1.0, 3.0, 10 mg/kg). Buprenorphine caused dose-dependent decreases in respiratory frequency while increasing tidal volume, minute ventilation, and respiratory duty cycle. The effects of buprenorphine varied significantly with leptin status and sex. Buprenorphine decreased minute ventilation variability in all mice. The present findings highlight leptin status as an important modulator of respiration and encourage future studies aiming to elucidate the mechanisms through which leptin status alters breathing.


Asunto(s)
Analgésicos Opioides/farmacología , Buprenorfina/farmacología , Leptina/metabolismo , Obesidad/fisiopatología , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Buprenorfina/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ventilación Pulmonar/efectos de los fármacos , Frecuencia Respiratoria/efectos de los fármacos , Caracteres Sexuales , Volumen de Ventilación Pulmonar
15.
ISME Commun ; 2(1): 66, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-37938724

RESUMEN

There are known associations between opioids, obesity, and the gut microbiome, but the molecular connection/mediation of these relationships is not understood. To better clarify the interplay of physiological, genetic, and microbial factors, this study investigated the microbiome and host inflammatory responses to chronic opioid administration in genetically obese, diet-induced obese, and lean mice. Samples of feces, urine, colon tissue, and plasma were analyzed using targeted LC-MS/MS quantification of metabolites, immunoassays of inflammatory cytokine levels, genome-resolved metagenomics, and metaproteomics. Genetic obesity, diet-induced obesity, and morphine treatment in lean mice each showed increases in distinct inflammatory cytokines. Metagenomic assembly and binning uncovered over 400 novel gut bacterial genomes and species. Morphine administration impacted the microbiome's composition and function, with the strongest effect observed in lean mice. This microbiome effect was less pronounced than either diet or genetically driven obesity. Based on inferred microbial physiology from the metaproteome datasets, a high-fat diet transitioned constituent microbes away from harvesting diet-derived nutrients and towards nutrients present in the host mucosal layer. Considered together, these results identified novel host-dependent phenotypes, differentiated the effects of genetic obesity versus diet induced obesity on gut microbiome composition and function, and showed that chronic morphine administration altered the gut microbiome.

16.
J Neurosci ; 30(37): 12301-9, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20844126

RESUMEN

Drugs that potentiate transmission at GABA(A) receptors are widely used to enhance sleep and to cause general anesthesia. The mechanisms underlying these effects are unknown. This study tested the hypothesis that GABA(A) receptors in the pontine reticular nucleus, oral part (PnO) of mouse modulate five phenotypes of arousal: sleep and wakefulness, cortical electroencephalogram (EEG) activity, acetylcholine (ACh) release in the PnO, breathing, and recovery time from general anesthesia. Microinjections into the PnO of saline (vehicle control), the GABA(A) receptor agonist muscimol, muscimol with the GABA(A) receptor antagonist bicuculline, and bicuculline alone were performed in male C57BL/6J mice (n = 33) implanted with EEG recording electrodes. Muscimol caused a significant increase in wakefulness and decrease in rapid eye movement (REM) and non-REM (NREM) sleep. These effects were reversed by coadministration of bicuculline. Bicuculline administered alone caused a significant decrease in wakefulness and increase in NREM sleep and REM sleep. Muscimol significantly increased EEG power in the delta range (0.5-4 Hz) during wakefulness and in the theta range (4-9 Hz) during REM sleep. Dialysis delivery of bicuculline to the PnO of male mice (n = 18) anesthetized with isoflurane significantly increased ACh release in the PnO, decreased breathing rate, and increased anesthesia recovery time. All drug effects were concentration dependent. The effects on phenotypes of arousal support the conclusion that GABA(A) receptors in the PnO promote wakefulness and suggest that increasing GABAergic transmission in the PnO may be one mechanism underlying the phenomenon of paradoxical behavioral activation by some benzodiazepines.


Asunto(s)
Conducta Animal/fisiología , Electroencefalografía , Fenotipo , Puente/metabolismo , Receptores de GABA-A/fisiología , Formación Reticular/metabolismo , Vigilia/fisiología , Animales , Conducta Animal/efectos de los fármacos , Bicuculina/administración & dosificación , Electroencefalografía/efectos de los fármacos , Agonistas de Receptores de GABA-A , Masculino , Ratones , Ratones Endogámicos C57BL , Microdiálisis , Microinyecciones , Muscimol/administración & dosificación , Puente/citología , Puente/fisiología , Receptores de GABA-A/genética , Formación Reticular/citología , Formación Reticular/efectos de los fármacos , Vigilia/genética , Ácido gamma-Aminobutírico/fisiología
17.
J Neurochem ; 118(4): 571-80, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21679185

RESUMEN

The oral part of the pontine reticular formation (PnO) is a component of the ascending reticular activating system and plays a role in the regulation of sleep and wakefulness. The PnO receives glutamatergic and GABAergic projections from many brain regions that regulate behavioral state. Indirect, pharmacological evidence has suggested that glutamatergic and GABAergic signaling within the PnO alters traits that characterize wakefulness and sleep. No previous studies have simultaneously measured endogenous glutamate and GABA from rat PnO in relation to sleep and wakefulness. The present study utilized in vivo microdialysis coupled on-line to capillary electrophoresis with laser-induced fluorescence to test the hypothesis that concentrations of glutamate and GABA in the PnO vary across the sleep/wake cycle. Concentrations of glutamate and GABA were significantly higher during wakefulness than during non-rapid eye movement sleep and rapid eye movement sleep. Regression analysis revealed that decreases in glutamate and GABA accounted for a significant portion of the variance in the duration of non-rapid eye movement sleep and rapid eye movement sleep episodes. These data provide novel support for the hypothesis that endogenous glutamate and GABA in the PnO contribute to the regulation of sleep duration.


Asunto(s)
Ácido Glutámico/metabolismo , Puente/metabolismo , Puente/fisiología , Formación Reticular/metabolismo , Formación Reticular/fisiología , Sueño/fisiología , Ácido gamma-Aminobutírico/metabolismo , Aminoácidos/metabolismo , Animales , Conducta Animal/fisiología , Electroencefalografía , Electromiografía , Electroforesis Capilar , Masculino , Microdiálisis , Ratas , Ratas Sprague-Dawley , Sueño/genética , Sueño REM/genética , Sueño REM/fisiología , Vigilia/fisiología
18.
Anesthesiology ; 115(4): 836-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21666435

RESUMEN

BACKGROUND: The current study was designed to test the hypothesis that the increased duration of analgesia caused by adding dexmedetomidine to local anesthetic results from blockade of the hyperpolarization-activated cation (I(h)) current. METHODS: In this randomized, blinded, controlled study, the analgesic effects of peripheral nerve blocks using 0.5% ropivacaine alone or 0.5% ropivacaine plus dexmedetomidine (34 µM or 6 µg/kg) were assessed with or without the pretreatment of α(1)- and α(2)-adrenoceptor antagonists (prazosin and idazoxan, respectively) and antagonists and agonists of the I(h) current (ZD 7288 and forskolin, respectively). Sciatic nerve blocks were performed, and analgesia was measured by paw withdrawal latency to a thermal stimulus every 30 min for 300 min postblock. RESULTS: The analgesic effect of dexmedetomidine added to ropivacaine was not reversed by either prazosin or idazoxan. There were no additive or attenuated effects from the pretreatment with ZD 7288 (I(h) current blocker) compared with dexmedetomidine added to ropivacaine. When forskolin was administered as a pretreatment to ropivacaine plus dexmedetomidine, there were statistically significant reductions in duration of analgesia at time points 90-180 min (P < 0.0001 for each individual comparison). The duration of blockade for the forskolin (768 µM) followed by ropivacaine plus dexmedetomidine group mirrored the pattern of the ropivacaine alone group, thereby implying a reversal effect. CONCLUSION: Dexmedetomidine added to ropivacaine caused approximately a 75% increase in the duration of analgesia, which was reversed by pretreatment with an I(h) current enhancer. The analgesic effect of dexmedetomidine was not reversed by an α(2)-adrenoceptor antagonist.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Amidas/farmacología , Analgesia , Anestésicos Locales/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/antagonistas & inhibidores , Dexmedetomidina/farmacología , Hipnóticos y Sedantes/farmacología , Bloqueo Nervioso , Bloqueadores de los Canales de Potasio , Nervio Ciático/efectos de los fármacos , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Animales , Conducta Animal/efectos de los fármacos , Colforsina/farmacología , Relación Dosis-Respuesta a Droga , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Idazoxan/farmacología , Masculino , Dimensión del Dolor/efectos de los fármacos , Canales de Potasio , Prazosina/farmacología , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Ropivacaína
19.
Anesthesiology ; 115(4): 743-53, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21857500

RESUMEN

BACKGROUND: Buprenorphine, a partial µ-opioid receptor agonist and κ-opioid receptor antagonist, is an effective analgesic. The effects of buprenorphine on sleep have not been well characterized. This study tested the hypothesis that an antinociceptive dose of buprenorphine decreases sleep and decreases adenosine concentrations in regions of the basal forebrain and pontine brainstem that regulate sleep. METHODS: Male Sprague Dawley rats were implanted with intravenous catheters and electrodes for recording states of wakefulness and sleep. Buprenorphine (1 mg/kg) was administered systemically via an indwelling catheter and sleep-wake states were recorded for 24 h. In additional rats, buprenorphine was delivered by microdialysis to the pontine reticular formation and substantia innominata of the basal forebrain while adenosine was simultaneously measured. RESULTS: An antinociceptive dose of buprenorphine caused a significant increase in wakefulness (25.2%) and a decrease in nonrapid eye movement sleep (-22.1%) and rapid eye movement sleep (-3.1%). Buprenorphine also increased electroencephalographic delta power during nonrapid eye movement sleep. Coadministration of the sedative-hypnotic eszopiclone diminished the buprenorphine-induced decrease in sleep. Dialysis delivery of buprenorphine significantly decreased adenosine concentrations in the pontine reticular formation (-14.6%) and substantia innominata (-36.7%). Intravenous administration of buprenorphine significantly decreased (-20%) adenosine in the substantia innominata. CONCLUSIONS: Buprenorphine significantly increased time spent awake, decreased nonrapid eye movement sleep, and increased latency to sleep onset. These disruptions in sleep architecture were mitigated by coadministration of the nonbenzodiazepine sedative-hypnotic eszopiclone. The buprenorphine-induced decrease in adenosine concentrations in basal forebrain and pontine reticular formation is consistent with the interpretation that decreasing adenosine in sleep-regulating brain regions is one mechanism by which opioids disrupt sleep.


Asunto(s)
Adenosina/metabolismo , Analgésicos Opioides/farmacología , Química Encefálica/efectos de los fármacos , Buprenorfina/farmacología , Sueño/efectos de los fármacos , Sueño/fisiología , Animales , Compuestos de Azabiciclo/farmacología , Conducta Animal/efectos de los fármacos , Ritmo Delta/efectos de los fármacos , Electroencefalografía/efectos de los fármacos , Eszopiclona , Hipnóticos y Sedantes/farmacología , Masculino , Microdiálisis , Dimensión del Dolor/efectos de los fármacos , Piperazinas/farmacología , Polisomnografía/efectos de los fármacos , Puente/efectos de los fármacos , Puente/metabolismo , Ratas , Ratas Sprague-Dawley , Formación Reticular/efectos de los fármacos , Formación Reticular/metabolismo , Sustancia Innominada/efectos de los fármacos , Sustancia Innominada/metabolismo , Vigilia/efectos de los fármacos
20.
J Neurosci ; 29(3): 871-81, 2009 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19158311

RESUMEN

During prolonged intervals of wakefulness, brain adenosine levels rise within the basal forebrain and cortex. The view that adenosine promotes sleep is supported by the corollary that N-methylated xanthines such as caffeine increase brain and behavioral arousal by blocking adenosine receptors. The four subtypes of adenosine receptors are distributed heterogeneously throughout the brain, yet the neurotransmitter systems and brain regions through which adenosine receptor blockade causes arousal are incompletely understood. This study tested the hypothesis that adenosine A(1) and A(2A) receptors in the prefrontal cortex contribute to the regulation of behavioral and cortical arousal. Dependent measures included acetylcholine (ACh) release in the prefrontal cortex, cortical electroencephalographic (EEG) power, and time to waking after anesthesia. Sleep and wakefulness were also quantified after microinjecting an adenosine A(1) receptor antagonist into the prefrontal cortex. The results showed that adenosine A(1) and A(2A) receptors in the prefrontal cortex modulate cortical ACh release, behavioral arousal, EEG delta power, and sleep. Additional dual microdialysis studies revealed that ACh release in the pontine reticular formation is significantly altered by dialysis delivery of adenosine receptor agonists and antagonists to the prefrontal cortex. These data, and early brain transection studies demonstrating that the forebrain is not needed for sleep cycle generation, suggest that the prefrontal cortex modulates EEG and behavioral arousal via descending input to the pontine brainstem. The results provide novel evidence that adenosine A(1) receptors within the prefrontal cortex comprise part of a descending system that inhibits wakefulness.


Asunto(s)
Acetilcolina/metabolismo , Nivel de Alerta/fisiología , Corteza Prefrontal/metabolismo , Receptor de Adenosina A1/fisiología , Receptores de Adenosina A2/fisiología , Adenosina/análogos & derivados , Adenosina/farmacología , Antagonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A2 , Análisis de Varianza , Animales , Conducta Animal , Cafeína/farmacología , Cromatografía Líquida de Alta Presión/métodos , Electroencefalografía/métodos , Electromiografía/métodos , Masculino , Ratones , Microdiálisis/métodos , Fenetilaminas/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Análisis Espectral , Triazinas/farmacología , Triazoles/farmacología , Xantinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA