Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
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
3.
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
4.
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.

5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Personal Disord ; 11(3): 222-229, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31724409

RESUMEN

This study examined the relationship between borderline personality disorder assessed as self-reported borderline features (Morey, 1991), opioid use, and Hepatitis C virus (HCV) in pregnant women. There were 55 women in the opioid use group and 38 in the comparison group who were at high risk due to medical issues that did not include drug use. Women were in their 2nd or 3rd trimester. All women received Medicaid and were racially representative of the geographic area (84% White). We assessed opioid use severity from medical records based on urine assays and prescriptions for opioids. The results revealed that women who scored in the clinical range of total borderline features, which is associated with a diagnosis of borderline personality disorder (Trull, 1995), had 2.83 greater odds of being opioid users (prescribed and nonprescribed) than had individuals below the cutoff. The borderline features of affective instability, identity disturbance, negative relationships, and self-harm/impulsivity were significantly correlated with opioid use severity. Negative relationships and self-harm/impulsivity contributed significant variance in opioid use severity over and above affective instability and identity disturbance. Women in the clinical range of borderline features were more likely to have HCV than were women below the cutoff, and the borderline feature of negative relationships specifically was associated with HCV. We discuss implications for interventions to address negative relationships and self-harm/impulsivity and interventions to help prevent opioid misuse in women before they become pregnant. (PsycInfo Database Record (c) 2020 APA, all rights reserved).


Asunto(s)
Trastorno de Personalidad Limítrofe/epidemiología , Hepatitis C/epidemiología , Trastornos Relacionados con Opioides/epidemiología , Complicaciones del Embarazo/epidemiología , Adulto , Analgésicos Opioides/efectos adversos , Trastorno de Personalidad Limítrofe/complicaciones , Femenino , Hepacivirus , Hepatitis C/complicaciones , Humanos , Conducta Impulsiva , Trastornos Relacionados con Opioides/complicaciones , Inventario de Personalidad , Embarazo , Conducta Autodestructiva , Adulto Joven
12.
J Vis Exp ; (153)2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31789318

RESUMEN

Current web resources provide limited, user friendly tools to compute spectrograms for visualizing and quantifying electroencephalographic (EEG) data. This paper describes a Windows-based, open source code for creating EEG multitaper spectrograms. The compiled program is accessible to Windows users without software licensing. For Macintosh users, the program is limited to those with a MATLAB software license. The program is illustrated via EEG spectrograms that vary as a function of states of sleep and wakefulness, and opiate-induced alterations in those states. The EEGs of C57BL/6J mice were wirelessly recorded for 4 h after intraperitoneal injection of saline (vehicle control) and antinociceptive doses of morphine, buprenorphine, and fentanyl. Spectrograms showed that buprenorphine and morphine caused similar changes in EEG power at 1-3 Hz and 8-9 Hz. Spectrograms after administration of fentanyl revealed maximal average power bands at 3 Hz and 7 Hz. The spectrograms unmasked differential opiate effects on EEG frequency and power. These computer-based methods are generalizable across drug classes and can be readily modified to quantify and display a wide range of rhythmic biological signals.


Asunto(s)
Electroencefalografía , Programas Informáticos , Análisis Espectral , Animales , Electrodos , Electromiografía , Ratones , Alcaloides Opiáceos/administración & dosificación , Sueño , Vigilia
13.
Sleep Med Rev ; 48: 101207, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31505369

RESUMEN

This systematic review with meta-analysis and trial sequential analysis of randomized clinical trials aimed to clarify the efficacy of sleep and circadian interventions on preventing postoperative delirium. The search and screening identified 13 trials with great heterogeneity in interventions, surgery types as well as methods for evaluating delirium, sleep and circadian rhythms. Meta-analyses revealed that sleep and circadian interventions were associated with decreased incidences of postoperative delirium (pooled relative risk (RR) = 0.48, 95% confidence interval (CI) = 0.29 to 0.78) compared with control. The pooled incidences of delirium for patients receiving interventions and no intervention (control) were 8.6% and 20.7% respectively. Results of the trial sequential analysis supported the interpretation that sleep and circadian interventions significantly diminished delirium compared to control. Subgroup analysis found that interventions that showed positive efficacy on sleep and circadian outcomes (p < 0.001), but not those without improvements (p = 0.114) or without assessments (p = 0.858), were associated with decreased risk of delirium. Dexmedetomidine sedation (p < 0.001) and timed bright light exposure (p = 0.006) appeared to reduce postoperative delirium. In summary, currently only limited evidence suggests strategies targeted at sleep and circadian health as a useful way to prevent postoperative delirium.


Asunto(s)
Ritmo Circadiano/fisiología , Delirio/prevención & control , Complicaciones Posoperatorias/prevención & control , Ensayos Clínicos Controlados Aleatorios como Asunto , Sueño/fisiología , Delirio/tratamiento farmacológico , Dexmedetomidina/uso terapéutico , Humanos , Hipnóticos y Sedantes/uso terapéutico
14.
Sleep ; 41(11)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30107617

RESUMEN

Study Objectives: This study tested the hypothesis that sleep fragmentation (SF) delays wound healing in obese B6.BKS(D)-Leprdb/J (db/db) mice with impaired leptin signaling and type 2 diabetes compared with wild-type C57BL/6J (B6) mice. Methods: Adult male mice (n = 34) were anesthetized and bilateral full-thickness excisional wounds were created on the back of each mouse. Half of the db/db and B6 mice were housed in SF cages equipped with a bar that moved across the cage floor every 2 min, 12 hr/day for 23 days. The other half of each group of mice was housed in the same room and did not experience SF. The dependent measures were number of days required to achieve wound closure, mRNA expression of four inflammatory mediators, blood glucose, insulin, and corticosterone. Results: SF in the db/db mice caused a significant delay in wound healing relative to db/db mice with no SF. Days to achieve 50 per cent wound healing were 13.3 ± 0.4 with SF compared with 10.3 ± 0.7 without SF. All B6 mice achieved 50 per cent wound healing within 6 days and complete healing after 16 days. SF caused a significant increase in wound levels of TNF-α mRNA only in the db/db mice and an increase in corticosterone only in the B6 mice. Conclusions: The delayed wound healing in obese, diabetic mice caused by SF is homologous to delayed wound healing in some patients with type 2 diabetes. The results support the interpretation that altered leptinergic signaling and inflammatory proteins contribute to delayed wound healing.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Obesidad/patología , Privación de Sueño/patología , Cicatrización de Heridas/fisiología , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 2/sangre , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/sangre , Privación de Sueño/sangre
15.
Sci Rep ; 8(1): 11225, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30046159

RESUMEN

By identifying endogenous molecules in brain extracellular fluid metabolomics can provide insight into the regulatory mechanisms and functions of sleep. Here we studied how the cortical metabolome changes during sleep, sleep deprivation and spontaneous wakefulness. Mice were implanted with electrodes for chronic sleep/wake recording and with microdialysis probes targeting prefrontal and primary motor cortex. Metabolites were measured using ultra performance liquid chromatography-high resolution mass spectrometry. Sleep/wake changes in metabolites were evaluated using partial least squares discriminant analysis, linear mixed effects model analysis of variance, and machine-learning algorithms. More than 30 known metabolites were reliably detected in most samples. When used by a logistic regression classifier, the profile of these metabolites across sleep, spontaneous wake, and enforced wake was sufficient to assign mice to their correct experimental group (pair-wise) in 80-100% of cases. Eleven of these metabolites showed significantly higher levels in awake than in sleeping mice. Some changes extend previous findings (glutamate, homovanillic acid, lactate, pyruvate, tryptophan, uridine), while others are novel (D-gluconate, N-acetyl-beta-alanine, N-acetylglutamine, orotate, succinate/methylmalonate). The upregulation of the de novo pyrimidine pathway, gluconate shunt and aerobic glycolysis may reflect a wake-dependent need to promote the synthesis of many essential components, from nucleic acids to synaptic membranes.


Asunto(s)
Metabolómica , Corteza Prefrontal/metabolismo , Sueño/fisiología , Vigilia/fisiología , Animales , Ácido Glutámico/metabolismo , Ácido Homovanílico/metabolismo , Humanos , Ácido Láctico/metabolismo , Ratones , Corteza Motora/metabolismo , Corteza Motora/fisiopatología , Corteza Prefrontal/fisiopatología , Ácido Pirúvico/metabolismo , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Triptófano/metabolismo , Uridina/metabolismo
16.
Methods Enzymol ; 603: 237-255, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29673529

RESUMEN

Anesthetic mechanisms that eliminate consciousness and perception of pain are products of the nervous system. Chemical approaches to the study of anesthetic mechanisms have the potential to serve as an ideal interface between basic and clinical neuroscience. There are disproportionately more basic neurochemical studies than clinical studies of anesthetic mechanisms. Even within neuroscience, the study of anesthetic mechanisms is sparse. The Society for Neuroscience hosts one of the world's largest and most vibrant scientific meetings, yet the content themes of that meeting do not include anesthesia. One goal of this chapter is to facilitate neurochemical studies of anesthetic mechanisms by outlining user-friendly descriptions of existing and emerging techniques. The introduction provides a context for chapter goals. The second portion of this chapter focuses on microdialysis methods that enable the humane acquisition of neurochemical samples from intact, behaving animals during anesthetic induction, maintenance, and emergence. No single neurotransmitter and no single brain region regulate the physiological and behavioral traits characteristic of any anesthetic state. This limitation is being addressed via application of new instrumentation and techniques in analytic chemistry. The final third of this chapter highlights selected omics approaches that are now being applied to the neurochemical study of anesthetic mechanisms. We hope that this brief chapter can stimulate basic and clinical metabolomic approaches aiming to elucidate the mechanisms of anesthetic action.


Asunto(s)
Anestesia General/métodos , Anestésicos/farmacocinética , Metaboloma/fisiología , Microdiálisis/métodos , Neuroquímica/métodos , Neurotransmisores/análisis , Animales , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Química Encefálica/fisiología , Mapeo Encefálico , Técnicas de Química Analítica , Cromatografía Líquida de Alta Presión , Humanos , Bombas de Infusión , Límite de Detección , Microdiálisis/instrumentación , Red Nerviosa/anatomía & histología , Red Nerviosa/fisiología , Neuroquímica/instrumentación , Neurotransmisores/metabolismo , Ratas , Técnicas Estereotáxicas , Vigilia/fisiología
17.
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
18.
Neurosci Lett ; 660: 29-33, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28893589

RESUMEN

Buprenorphine is an opiate used for pain management and to treat opiate addiction. The cytokine leptin can modulate nociception, but the extent to which buprenorphine-induced antinociception varies as a function of leptin signaling has not been characterized. Four congenic mouse lines with phenotypes that include differences in body weight and leptin status were used to test the hypothesis that the antinociceptive effects of buprenorphine vary as function of sex and leptin signaling. Each mouse line was comprised of males (n=12) and females (n=12) for a total of 96 animals. Groups included C57BL/6J (B6) mice (wild type), B6 mice with diet-induced obesity (DIO), obese B6.Cg-Lepob/J (ob/ob) mice lacking leptin, and obese B6.BKS(D)-Leprdb/J (db/db) mice with dysfunctional leptin receptors. The dependent measure was tail flick latency (TFL) in seconds for mouse-initiated tail removal from a warm water bath. Independent variables were intraperitoneal administration of saline (control) or buprenorphine (0.3mg/kg). Within every mouse line, buprenorphine significantly increased TFL relative to saline. Compared to the other mouse lines, db/db mice with dysfunctional leptin receptors had a significantly longer TFL after saline and after buprenorphine. TFL did not vary significantly by body weight or sex. The results provide novel support for the interpretation that acute thermal nociception is associated with altered leptin signaling.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Buprenorfina/administración & dosificación , Leptina/fisiología , Obesidad/fisiopatología , Receptores de Leptina/fisiología , Animales , Femenino , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Umbral del Dolor/efectos de los fármacos , Receptores de Leptina/genética , Caracteres Sexuales
19.
Sleep ; 39(2): 393-404, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26564126

RESUMEN

STUDY OBJECTIVES: This study tested the hypothesis that Regulators of G protein Signaling (RGS) proteins contribute to the regulation of wakefulness, non-rapid eye movement (NREM) sleep, and rapid eye movement (REM) sleep, and to sleep disruption caused by volatile anesthetics. METHODS: The three groups used in this study included wild-type (WT; n = 7) mice and knock-in mice that were heterozygous (+/GS; n = 7) or homozygous (GS/GS; n = 7) for an RGS-insensitive allele that causes prolonged Gαi2 signaling. Mice were implanted with electrodes for recording sleep and conditioned for 1 week or more to sleep in the laboratory. Using within and between groups designs, 24-h recordings of wakefulness, NREM sleep, and REM sleep were compared across three interventions: (1) baseline (control) and after 3 h of being anesthetized with (2) isoflurane or (3) sevoflurane. RESULTS: Baseline recordings during the light phase revealed that relative to WT mice, homozygous RGS-insensitive (GS/GS) mice exhibit significantly increased wakefulness and decreased NREM and REM sleep. During the dark phase, these state-specific differences remained significant but reversed direction of change. After cessation of isoflurane and sevoflurane anesthesia there was a long-lasting and significant disruption of sleep and wakefulness. The durations of average episodes of wakefulness, NREM sleep, and REM sleep were significantly altered as a function of genotype and isoflurane and sevoflurane anesthesia. CONCLUSIONS: RGS proteins and Gαi2 play a significant role in regulating states of wakefulness, NREM sleep, and REM sleep. Genotype-specific differences demonstrate that RGS proteins modulate sleep disruption caused by isoflurane and sevoflurane anesthesia. The results also support the conclusion that isoflurane and sevoflurane anesthesia do not satisfy the homeostatic drive for sleep.


Asunto(s)
Anestesia , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Isoflurano/farmacología , Éteres Metílicos/farmacología , Proteínas RGS/metabolismo , Sueño/efectos de los fármacos , Sueño/fisiología , Vigilia/efectos de los fármacos , Vigilia/fisiología , Animales , Electrodos Implantados , Electroencefalografía , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Homeostasis/efectos de los fármacos , Masculino , Ratones , Sevoflurano , Sueño REM/efectos de los fármacos , Sueño REM/fisiología
20.
Sleep ; 38(1): 73-84, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25325438

RESUMEN

STUDY OBJECTIVES: Dexmedetomidine is used clinically to induce states of sedation that have been described as homologous to nonrapid eye movement (NREM) sleep. A better understanding of the similarities and differences between NREM sleep and dexmedetomidine-induced sedation is essential for efforts to clarify the relationship between these two states. This study tested the hypothesis that dexmedetomidine-induced sedation is homologous to sleep. DESIGN: This study used between-groups and within-groups designs. SETTING: University of Michigan. PARTICIPANTS: Adult male Sprague Dawley rats (n = 40). INTERVENTIONS: Independent variables were administration of dexmedetomidine and saline or Ringer's solution (control). Dependent variables included time spent in states of wakefulness, sleep, and sedation, electroencephalographic (EEG) power, adenosine levels in the substantia innominata (SI), and activation of pCREB and c-Fos in sleep related forebrain regions. MEASUREMENTS AND RESULTS: Dexmedetomidine significantly decreased time spent in wakefulness (-49%), increased duration of sedation (1995%), increased EEG delta power (546%), and eliminated the rapid eye movement (REM) phase of sleep for 16 h. Sedation was followed by a rebound increase in NREM and REM sleep. Systemically administered dexmedetomidine significantly decreased (-39%) SI adenosine levels. Dialysis delivery of dexmedetomidine into SI did not decrease adenosine level. Systemic delivery of dexmedetomidine did not alter c-Fos or pCREB expression in the horizontal diagonal band, or ventrolateral, median, and medial preoptic areas of the hypothalamus. CONCLUSIONS: Dexmedetomidine significantly altered normal sleep phenotypes, and the dexmedetomidine-induced state did not compensate for sleep need. Thus, in the Sprague Dawley rat, dexmedetomidine-induced sedation is characterized by behavioral, electrographic, and immunohistochemical phenotypes that are distinctly different from similar measures obtained during sleep.


Asunto(s)
Dexmedetomidina/farmacología , Hipnóticos y Sedantes/farmacología , Fenotipo , Sueño/fisiología , Adenosina/metabolismo , Animales , Prosencéfalo Basal/efectos de los fármacos , Prosencéfalo Basal/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dexmedetomidina/administración & dosificación , Diálisis , Electroencefalografía , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Sustancia Innominada/efectos de los fármacos , Sustancia Innominada/metabolismo , Factores de Tiempo , Vigilia/efectos de los fármacos , Vigilia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA