Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurophysiol ; 123(6): 2285-2296, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32347157

RESUMO

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.


Assuntos
Acetilcolina/metabolismo , Adenosina/metabolismo , Anestesia , Anestésicos Inalatórios/farmacologia , Isoflurano/farmacologia , Aprendizado de Máquina , Rede Nervosa , Norepinefrina/metabolismo , Córtex Pré-Frontal , Inconsciência/metabolismo , Vigília/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/metabolismo , Rede Nervosa/fisiopatologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia
2.
Sleep Med Rev ; 48: 101207, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31505369

RESUMO

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.


Assuntos
Ritmo Circadiano/fisiologia , Delírio/prevenção & controle , Complicações Pós-Operatórias/prevenção & controle , Ensaios Clínicos Controlados Aleatórios como Assunto , Sono/fisiologia , Delírio/tratamento farmacológico , Dexmedetomidina/uso terapêutico , Humanos , Hipnóticos e Sedativos/uso terapêutico
3.
Sleep ; 38(1): 73-84, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25325438

RESUMO

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.


Assuntos
Dexmedetomidina/farmacologia , Hipnóticos e Sedativos/farmacologia , Fenótipo , Sono/fisiologia , Adenosina/metabolismo , Animais , Prosencéfalo Basal/efeitos dos fármacos , Prosencéfalo Basal/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dexmedetomidina/administração & dosagem , Diálise , Eletroencefalografia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Sono REM/efeitos dos fármacos , Sono REM/fisiologia , Substância Inominada/efeitos dos fármacos , Substância Inominada/metabolismo , Fatores de Tempo , Vigília/efeitos dos fármacos , Vigília/fisiologia
4.
Anesthesiology ; 118(2): 327-36, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23263018

RESUMO

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.


Assuntos
Acetilcolina/metabolismo , Período de Recuperação da Anestesia , Ponte/metabolismo , Receptor A1 de Adenosina/efeitos dos fármacos , Respiração/efeitos dos fármacos , Formação Reticular/metabolismo , Agonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Anestesia , Animais , Nível de Alerta/fisiologia , Cromatografia Líquida de Alta Pressão , Condicionamento Operante/efeitos dos fármacos , Eletroquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Microinjeções , Ponte/efeitos dos fármacos , Equilíbrio Postural/efeitos dos fármacos , Reflexo/efeitos dos fármacos , Formação Reticular/efeitos dos fármacos
5.
Anesthesiology ; 115(4): 743-53, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21857500

RESUMO

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.


Assuntos
Adenosina/metabolismo , Analgésicos Opioides/farmacologia , Química Encefálica/efeitos dos fármacos , Buprenorfina/farmacologia , Sono/efeitos dos fármacos , Sono/fisiologia , Animais , Compostos Azabicíclicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Eletroencefalografia/efeitos dos fármacos , Zopiclona , Hipnóticos e Sedativos/farmacologia , Masculino , Microdiálise , Medição da Dor/efeitos dos fármacos , Piperazinas/farmacologia , Polissonografia/efeitos dos fármacos , Ponte/efeitos dos fármacos , Ponte/metabolismo , Ratos , Ratos Sprague-Dawley , Formação Reticular/efeitos dos fármacos , Formação Reticular/metabolismo , Substância Inominada/efeitos dos fármacos , Substância Inominada/metabolismo , Vigília/efeitos dos fármacos
6.
J Pain ; 11(6): 535-44, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20015707

RESUMO

UNLABELLED: Clinical and preclinical data concur that sleep disruption causes hyperalgesia, but the brain mechanisms through which sleep and pain interact remain poorly understood. Evidence that pontine components of the ascending reticular activating system modulate sleep and nociception encouraged the present study testing the hypothesis that hypocretin-1 (orexin-A) and an adenosine receptor agonist administered into the pontine reticular nucleus, oral part (PnO) each alter thermal nociception. Adult male rats (n = 23) were implanted with microinjection guide tubes aimed for the PnO. The PnO was microinjected with saline (control), hypocretin-1, the adenosine A(1) receptor agonist N(6)-p-sulfophenyladenosine (SPA), the hypocretin receptor-1 antagonist N-(2-Methyl-6-benzoxazolyl)-N''-1,5-naphthyridin-4-yl-urea (SB-334867), and hypocretin-1 plus SB-334867. As an index of antinociceptive behavior, the latency (in seconds) to paw withdrawal away from a thermal stimulus was measured following each microinjection. Compared to control, antinociception was significantly increased by hypocretin-1 and by SPA. SB-334867 increased nociceptive responsiveness, and administration of hypocretin-1 plus SB-334867 blocked the antinociception caused by hypocretin-1. These results suggest for the first time that hypocretin receptors in rat PnO modulate nociception. PERSPECTIVE: Widely distributed and overlapping neural networks regulate states of sleep and pain. Specifying the brain regions and neurotransmitters through which pain and sleep interact is an essential step for developing adjunctive therapies that diminish pain without disrupting states of sleep and wakefulness.


Assuntos
Agonistas do Receptor A1 de Adenosina , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neuropeptídeos/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Ponte/efeitos dos fármacos , Formação Reticular/efeitos dos fármacos , Adenosina/análogos & derivados , Adenosina/farmacologia , Envelhecimento , Animais , Benzoxazóis/farmacologia , Temperatura Alta , Masculino , Microinjeções , Naftiridinas , Receptores de Orexina , Orexinas , Medição da Dor , Ponte/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/antagonistas & inibidores , Receptores de Neuropeptídeos/metabolismo , Formação Reticular/metabolismo , Fatores de Tempo , Ureia/análogos & derivados , Ureia/farmacologia
7.
Anesthesiology ; 111(6): 1327-33, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19934879

RESUMO

BACKGROUND: Opioids disrupt sleep and adenosine promotes sleep, but no studies have characterized the effects of opioids on adenosine levels in brain regions known to regulate states of arousal. Delivering opioids to the pontine reticular formation (PRF) and substantia innominata (SI) region of the basal forebrain disrupts sleep. In contrast, administering adenosine agonists to the PRF or SI increases sleep. These findings encouraged the current study testing the hypothesis that microdialysis delivery of opioids to the PRF or SI decreases adenosine levels in the PRF or SI, respectively. METHODS: A microdialysis probe was placed in the PRF of isoflurane anesthetized rats and perfused with Ringer's solution (control) followed by Ringer's solution containing morphine (0, 10, 30, 100, or 300 microm), fentanyl (100 microm), morphine (100 microm) and the adenosine deaminase inhibitor EHNA (100 microm), or naloxone (10 microm) and morphine (100 microm). Additional experiments measured adenosine levels in the SI before and during microdialysis delivery of morphine, fentanyl, and morphine plus EHNA. RESULTS: Morphine caused a significant (P < 0.05) concentration-dependent decrease in PRF adenosine levels. The significant decrease (-20%) in adenosine caused by 100 microm morphine was blocked by coadministration of naloxone. Fentanyl also significantly decreased (-13.3%) PRF adenosine. SI adenosine levels were decreased by morphine (-26.8%) and fentanyl (-27.4%). In both PRF and SI, coadministration of morphine and EHNA prevented the significant decrease in adenosine levels caused by morphine alone. CONCLUSIONS: These data support the interpretation that decreased adenosine levels in sleep-regulating brain regions may be one of the mechanisms by which opioids disrupt sleep.


Assuntos
Inibidores de Adenosina Desaminase , Adenosina/metabolismo , Analgésicos Opioides/farmacologia , Química Encefálica/efeitos dos fármacos , Animais , Cromatografia Líquida de Alta Pressão , Cognição/efeitos dos fármacos , Masculino , Microdiálise , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Formação Reticular/efeitos dos fármacos , Formação Reticular/metabolismo , Sono/efeitos dos fármacos , Espectrofotometria Ultravioleta
8.
J Pain ; 10(8): 836-43, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19380255

RESUMO

UNLABELLED: A single gene deletion causes lack of leptin and obesity in B6.V-Lep(ob) (obese; ob) mice compared with wild-type C57BL/6J (B6) mice. This study compared the phenotype of nociception and supraspinal antinociception in obese and B6 mice by testing 2 hypotheses: (1) microinjection of cholinomimetics or an adenosine receptor agonist, but not morphine, into the pontine reticular formation (PRF) is antinociceptive in B6 but not obese mice, and (2) leptin replacement in obese mice attenuates differences in nociceptive responses between obese and B6 mice. Adult male mice (n = 22) were implanted with microinjection guide tubes aimed for the PRF. The PRF was injected with neostigmine, carbachol, nicotine, N(6)-p-sulfophenyladenosine (SPA), morphine, or saline (control), and latency to paw withdrawal (PWL) from a thermal stimulus was recorded. B6 and ob mice did not differ in PWL after saline microinjection into the PRF. Neostigmine, carbachol, and SPA caused PWL to increase significantly in B6 but not obese mice. An additional 15 obese mice were implanted with osmotic pumps that delivered leptin for 7 days. Leptin replacement in obese mice restored the analgesic effect of PRF neostigmine to the level displayed by B6 mice. The results show for the first time that leptin significantly alters supraspinal cholinergic antinociception. PERSPECTIVE: This study specifies a brain region (the pontine reticular formation), cholinergic neurotransmission, and a protein (leptin) modulating thermal nociception. The results are relevant for efforts to understand the association between obesity, disordered sleep, and hyperalgesia.


Assuntos
Acetilcolina/metabolismo , Leptina/metabolismo , Obesidade/fisiopatologia , Dor/fisiopatologia , Ponte/fisiopatologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Inibidores da Colinesterase/farmacologia , Temperatura Alta , Leptina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfina/farmacologia , Neostigmina/farmacologia , Nicotina/farmacologia , Obesidade/genética , Dor/tratamento farmacológico , Medição da Dor , Ponte/efeitos dos fármacos , Fatores de Tempo
9.
J Neurosci ; 29(3): 871-81, 2009 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19158311

RESUMO

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.


Assuntos
Acetilcolina/metabolismo , Nível de Alerta/fisiologia , Córtex Pré-Frontal/metabolismo , Receptor A1 de Adenosina/fisiologia , Receptores A2 de Adenosina/fisiologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Análise de Variância , Animais , Comportamento Animal , Cafeína/farmacologia , Cromatografia Líquida de Alta Pressão/métodos , Eletroencefalografia/métodos , Eletromiografia/métodos , Masculino , Camundongos , Microdiálise/métodos , Fenetilaminas/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Análise Espectral , Triazinas/farmacologia , Triazóis/farmacologia , Xantinas/farmacologia
10.
J Neurochem ; 96(6): 1750-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16539690

RESUMO

In vivo microdialysis in C57BL/6J (B6) mouse was used to test the hypothesis that activating adenosine A(2A) receptors in the pontine reticular formation (PRF) increases acetylcholine (ACh) release and rapid eye movement (REM) sleep. Eight concentrations of the adenosine A(2A) receptor agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine hydrochloride (CGS 21680; CGS) were delivered to the PRF and ACh in the PRF was quantified. ACh release was significantly increased by dialysis with 3 mum CGS and significantly decreased by dialysis with 10 and 100 microm CGS. Co-administration of the adenosine A(2A) receptor antagonist 4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol (ZM 241385; 30 nM) blocked the CGS-induced increase in ACh release. In a second series of experiments, CGS (3 microm) was delivered by dialysis to the PRF for 2 h while recording sleep and wakefulness. CGS significantly decreased time in wakefulness (-51% in h 1; -54% in h 2), increased time in non-rapid eye movement (NREM) sleep (90% in h 1; 151% in h 2), and increased both time in REM sleep (331% in h 2) and the number of REM sleep episodes (488% in h 2). The enhancement of REM sleep is consistent with the interpretation that adenosine A(2A) receptors in the PRF of the B6 mouse contribute to REM sleep regulation, in part, by increasing ACh release in the PRF. A(2A) receptor activation may promote NREM sleep via GABAergic inhibition of arousal promoting neurons in the PRF.


Assuntos
Acetilcolina/metabolismo , Adenosina/metabolismo , Ponte/metabolismo , Receptor A2A de Adenosina/metabolismo , Formação Reticular/metabolismo , Sono/fisiologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Animais , Relação Dose-Resposta a Droga , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microdiálise , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Fenetilaminas/farmacologia , Ponte/efeitos dos fármacos , Formação Reticular/efeitos dos fármacos , Sono/efeitos dos fármacos , Sono REM/efeitos dos fármacos , Sono REM/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Triazinas/farmacologia , Triazóis/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Vigília/efeitos dos fármacos , Vigília/fisiologia , Ácido gama-Aminobutírico/metabolismo
11.
Anesthesiology ; 98(4): 912-20, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12657853

RESUMO

BACKGROUND: Adenosine modulates cell excitability, acetylcholine release, nociception, and sleep. Pontine cholinergic neurotransmission contributes to the generation and maintenance of electroencephalographic and behavioral arousal. Adenosine A(1) receptors inhibit arousal-promoting, pontine cholinergic neurons, and adenosine enhances sleep. No previous studies have determined whether pontine adenosine also modulates recovery from anesthesia. Therefore, the current study tested the hypotheses that dialysis delivery of the adenosine A(1) receptor agonist N6-p-sulfophenyladenosine (SPA) into the pontine reticular formation would decrease acetylcholine release and increase the time needed for recovery from halothane anesthesia. METHODS: A microdialysis probe was positioned in the pontine reticular formation of halothane-anesthetized cats. Probes were perfused with Ringer's solution (control) followed by the adenosine A(1) receptor agonist SPA (0.088 or 8.8 mm). Dependent measures included acetylcholine release and a numeric assessment of recovery from anesthesia. An intensive, within-subjects design and analysis of variance evaluated SPA's main effect on acetylcholine release and anesthetic recovery. The adenosine A(1) receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 100 microm) was coadministered with SPA to test for antagonist blocking of SPA's effects. RESULTS: SPA significantly (P < 0.0001) decreased acetylcholine release in the pontine reticular formation and significantly (P < 0.0001) delayed recovery from anesthesia. Coadministration of SPA and DPCPX caused no decrease in acetylcholine release or delay in postanesthetic recovery. Dialysis delivery of SPA into the cerebellar cortex confirmed that the SPA effects were site-specific to the pontine reticular formation. CONCLUSION: The results provide a novel extension of the sleep-promoting effects of adenosine by showing that pontine delivery of an adenosine A(1) receptor agonist delays resumption of wakefulness following halothane anesthesia. This extension is consistent with a potentially larger relevance of the current findings for efforts to specify neurons and molecules causing physiologic and behavioral traits comprising anesthetic states. These data support the conclusion that adenosine A(1) receptors in medial regions of the pontine reticular formation, known to modulate sleep, also contribute to the generation and/or maintenance of halothane anesthesia.


Assuntos
Acetilcolina/metabolismo , Período de Recuperação da Anestesia , Ponte/efeitos dos fármacos , Agonistas do Receptor Purinérgico P1 , Formação Reticular/efeitos dos fármacos , Estimulação Acústica , Animais , Gatos , Cromatografia Líquida de Alta Pressão , Eletroquímica , Eletroencefalografia/efeitos dos fármacos , Eletromiografia/efeitos dos fármacos , Eletroculografia/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Microdiálise , Atividade Motora/efeitos dos fármacos , Oxigênio/sangue , Fluxo Sanguíneo Regional/efeitos dos fármacos , Pele/irrigação sanguínea , Xantinas/farmacologia
12.
Anesthesiology ; 97(6): 1597-601, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12459690

RESUMO

BACKGROUND: Both pain and the pharmacologic management of pain can cause the undesirable effect of sleep disruption. One goal of basic and clinical neuroscience is to facilitate rational drug development by identifying the brain regions and neurochemical modulators of sleep and pain. Adenosine is thought to be an endogenous sleep promoting substance and adenosinergic compounds can contribute to pain management. In the pontine brain stem adenosine promotes sleep but the effects of pontine adenosine on pain have not been studied. This study tested the hypothesis that an adenosine agonist would cause antinociception when microinjected into pontine reticular formation regions that regulate sleep. METHODS: The tail flick latency (TFL) test quantified the time in seconds for an animal to move its tail away from a thermal stimulus created by a beam of light. TFL measures were used to evaluate the antinociceptive effects of the adenosine A1 receptor agonist N6-p-sulfophenyladenosine (SPA). Pontine microinjection of SPA (0.1 microg/0.25 microl, 0.88 mm) was followed by TFL measures as a function of time after drug delivery and across the sleep-wake cycle. RESULTS: Compared with saline (control), pontine administration of the adenosine agonist significantly increased latency to tail withdrawal (P < 0.0001). The increase in antinociceptive behavior evoked by the adenosine agonist SPA was blocked by pretreatment with the adenosine A1 receptor antagonist 8-cyclopentyl-1, 3-dipropylxanthine (DPCPX, 0.75 ng/0.25 microl, 10 microm). CONCLUSIONS: These preclinical data encourage additional research on the cellular mechanisms by which adenosine in the pontine reticular formation contributes to the supraspinal modulation of pain.


Assuntos
Analgésicos/agonistas , Dor/prevenção & controle , Agonistas do Receptor Purinérgico P1 , Sono/efeitos dos fármacos , Xantinas/farmacologia , Animais , Gatos , Temperatura Alta/efeitos adversos , Masculino , Microinjeções , Dor/etiologia , Cauda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA