Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
J Neurosci ; 44(29)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38744530

RESUMEN

Sleep disorders affect millions of people around the world and have a high comorbidity with psychiatric disorders. While current hypnotics mostly increase non-rapid eye movement sleep (NREMS), drugs acting selectively on enhancing rapid eye movement sleep (REMS) are lacking. This polysomnographic study in male rats showed that the first-in-class selective melatonin MT1 receptor partial agonist UCM871 increases the duration of REMS without affecting that of NREMS. The REMS-promoting effects of UCM871 occurred by inhibiting, in a dose-response manner, the firing activity of the locus ceruleus (LC) norepinephrine (NE) neurons, which express MT1 receptors. The increase of REMS duration and the inhibition of LC-NE neuronal activity by UCM871 were abolished by MT1 pharmacological antagonism and by an adeno-associated viral (AAV) vector, which selectively knocked down MT1 receptors in the LC-NE neurons. In conclusion, MT1 receptor agonism inhibits LC-NE neurons and triggers REMS, thus representing a novel mechanism and target for REMS disorders and/or psychiatric disorders associated with REMS impairments.


Asunto(s)
Locus Coeruleus , Ratas Sprague-Dawley , Receptor de Melatonina MT1 , Sueño REM , Animales , Masculino , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Locus Coeruleus/fisiología , Ratas , Receptor de Melatonina MT1/agonistas , Receptor de Melatonina MT1/metabolismo , Sueño REM/fisiología , Sueño REM/efectos de los fármacos , Norepinefrina/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Neuronas Adrenérgicas/fisiología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología
2.
J Neurosci ; 43(13): 2338-2348, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36849414

RESUMEN

Photoaffinity ligands are best known as tools used to identify the specific binding sites of drugs to their molecular targets. However, photoaffinity ligands have the potential to further define critical neuroanatomic targets of drug action. In the brains of WT male mice, we demonstrate the feasibility of using photoaffinity ligands in vivo to prolong anesthesia via targeted yet spatially restricted photoadduction of azi-m-propofol (aziPm), a photoreactive analog of the general anesthetic propofol. Systemic administration of aziPm with bilateral near-ultraviolet photoadduction in the rostral pons, at the border of the parabrachial nucleus and locus coeruleus, produced a 20-fold increase in the duration of sedative and hypnotic effects compared with control mice without UV illumination. Photoadduction that missed the parabrachial-coerulean complex also failed to extend the sedative or hypnotic actions of aziPm and was indistinguishable from nonadducted controls. Paralleling the prolonged behavioral and EEG consequences of on target in vivo photoadduction, we conducted electrophysiologic recordings in rostral pontine brain slices. Using neurons within the locus coeruleus to further highlight the cellular consequences of irreversible aziPm binding, we demonstrate transient slowing of spontaneous action potentials with a brief bath application of aziPm that becomes irreversible on photoadduction. Together, these findings suggest that photochemistry-based strategies are a viable new approach for probing CNS physiology and pathophysiology.SIGNIFICANCE STATEMENT Photoaffinity ligands are drugs capable of light-induced irreversible binding, which have unexploited potential to identify the neuroanatomic sites of drug action. We systemically administer a centrally acting anesthetic photoaffinity ligand in mice, conduct localized photoillumination within the brain to covalently adduct the drug at its in vivo sites of action, and successfully enrich irreversible drug binding within a restricted 250 µm radius. When photoadduction encompassed the pontine parabrachial-coerulean complex, anesthetic sedation and hypnosis was prolonged 20-fold, thus illustrating the power of in vivo photochemistry to help unravel neuronal mechanisms of drug action.


Asunto(s)
Anestésicos Intravenosos , Encéfalo , Hipnosis , Hipnóticos y Sedantes , Ligandos , Etiquetas de Fotoafinidad , Propofol , Animales , Masculino , Ratones , Neuronas Adrenérgicas/efectos de los fármacos , Anestesia Intravenosa , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de la radiación , Electrocorticografía , Electroencefalografía , Hipnosis/métodos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/química , Hipnóticos y Sedantes/farmacología , Hipnóticos y Sedantes/efectos de la radiación , Locus Coeruleus/citología , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Locus Coeruleus/efectos de la radiación , Ratones Endogámicos C57BL , Núcleos Parabraquiales/efectos de los fármacos , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/efectos de la radiación , Etiquetas de Fotoafinidad/química , Etiquetas de Fotoafinidad/efectos de la radiación , Propofol/administración & dosificación , Propofol/análogos & derivados , Propofol/farmacología , Propofol/efectos de la radiación , Factores de Tiempo , Rayos Ultravioleta , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/química , Anestésicos Intravenosos/farmacología , Anestésicos Intravenosos/efectos de la radiación
3.
J Neurophysiol ; 132(1): 68-77, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38838298

RESUMEN

The prepositus hypoglossi nucleus (PHN) and the interstitial nucleus of Cajal (INC) are involved in the control of horizontal and vertical gaze, respectively. A previous study showed that PHN neurons exhibit depolarized or hyperpolarized responses to noradrenaline (NA). However, the adrenoceptor types that participate in NA-induced responses and the effects of NA on INC neurons have not yet been investigated. Furthermore, the relationship between NA-induced responses and neuron types defined by neurotransmitter phenotypes has not been determined. In this study, we investigated NA-induced current responses in PHN and INC neurons and the relationships between these responses and neuron types using whole cell recordings in wild-type and transgenic rat brainstem slices. Local application of NA to the cell soma induced slow inward (SI) and slow outward (SO) currents that were mainly mediated by α1 and α2 adrenoceptors, respectively. These current responses were observed in both PHN and INC neurons, although the proportion of INC neurons that responded to NA was low. Analyses of the distributions of the current responses revealed that in the PHN, all fluorescently identified inhibitory neurons exhibited SI currents, whereas glutamatergic and cholinergic neurons exhibited both SI and SO currents. In the INC, glutamatergic and inhibitory neurons preferentially exhibited SI and SO currents, respectively. When the PHN and INC neurons were characterized by their firing pattern, we found that the proportions of the currents depended on their firing pattern. These results suggest that various modes of noradrenergic modulation in horizontal and vertical neural integrators are dependent on neuron type.NEW & NOTEWORTHY Noradrenergic modulation of oculomotor neural integrators involved in gaze control has not been elucidated. Here, we report that noradrenaline (NA)-induced slow inward (SI) and outward (SO) currents are mediated mainly by α1 and α2 adrenoceptors in neurons that participate in horizontal and vertical gaze control. The NA-induced current responses differed depending on the neurotransmitter phenotype and firing pattern. These results suggest various modes of noradrenergic modulation in horizontal and vertical integrator neurons.


Asunto(s)
Norepinefrina , Animales , Norepinefrina/farmacología , Ratas , Masculino , Ratas Transgénicas , Neuronas/fisiología , Neuronas/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 1/fisiología , Neuronas Adrenérgicas/fisiología , Neuronas Adrenérgicas/efectos de los fármacos , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos alfa 2/fisiología , Técnicas de Placa-Clamp , Tronco Encefálico/fisiología , Tronco Encefálico/citología , Tronco Encefálico/efectos de los fármacos , Neuronas Colinérgicas/fisiología , Neuronas Colinérgicas/efectos de los fármacos
4.
J Neurosci Res ; 102(10): e25390, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39373381

RESUMEN

Visceral feedback from the body is often subconscious, but plays an important role in guiding motivated behaviors. Vagal sensory neurons relay "gut feelings" to noradrenergic (NA) neurons in the caudal nucleus of the solitary tract (cNTS), which in turn project to the anterior ventrolateral bed nucleus of the stria terminalis (vlBNST) and other hypothalamic-limbic forebrain regions. Prior work supports a role for these circuits in modulating memory consolidation and extinction, but a potential role in retrieval of conditioned avoidance remains untested. To examine this, adult male rats underwent passive avoidance conditioning. We then lesioned gut-sensing vagal afferents by injecting cholecystokinin-conjugated saporin toxin (CSAP) into the vagal nodose ganglia (Experiment 1), or lesioned NA inputs to the vlBNST by injecting saporin toxin conjugated to an antibody against dopamine-beta hydroxylase (DSAP) into the vlBNST (Experiment 2). When avoidance behavior was later assessed, rats with vagal CSAP lesions or NA DSAP lesions displayed significantly increased conditioned passive avoidance. These new findings support the view that gut vagal afferents and the cNTSNA-to-vlBNST circuit play a role in modulating the expression/retrieval of learned passive avoidance. Overall, our data suggest a dynamic modulatory role of vagal sensory feedback to the limbic forebrain in integrating interoceptive signals with contextual cues that elicit conditioned avoidance behavior.


Asunto(s)
Reacción de Prevención , Ratas Sprague-Dawley , Nervio Vago , Animales , Masculino , Reacción de Prevención/fisiología , Reacción de Prevención/efectos de los fármacos , Ratas , Nervio Vago/fisiología , Nervio Vago/efectos de los fármacos , Nervio Vago/metabolismo , Saporinas , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/fisiología , Neuronas Adrenérgicas/metabolismo , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Norepinefrina/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Recuerdo Mental/fisiología , Recuerdo Mental/efectos de los fármacos , Memoria/fisiología , Memoria/efectos de los fármacos , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Núcleos Septales/fisiología
5.
Pflugers Arch ; 473(6): 859-872, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33855632

RESUMEN

The pontine A5 noradrenergic group contributes to the maturation of the respiratory system before birth in rats. These neurons are connected to the neural network responsible for respiratory rhythmogenesis. In the present study, we investigated the participation of A5 noradrenergic neurons in neonates (P7-8 and P14-15) in the control of ventilation during hypoxia and hypercapnia in in vivo experiments using conjugated saporin anti-dopamine beta-hydroxylase (DßH-SAP) to specifically ablate noradrenergic neurons. Thus, DßH-SAP (420 ng/µL) or saporin (SAP, control) was injected into the A5 region of neonatal male Wistar rats. Hypoxia reduced respiratory variability in control animals; however, A5 lesion prevented this effect in P7-8 rats. Our data suggest that noradrenergic neurons of the A5 region in neonate rats do not participate in the control of ventilation under baseline and hypercapnic conditions, but exert an inhibitory modulation on breathing variability under hypoxic challenge in early life (P7-8).


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Tronco Encefálico/citología , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Respiración , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/fisiología , Animales , Animales Recién Nacidos , Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/fisiopatología , Dopamina beta-Hidroxilasa/farmacología , Masculino , Ratas , Ratas Wistar , Saporinas/farmacología
6.
Neurobiol Learn Mem ; 178: 107362, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33333316

RESUMEN

Trauma patients treated with ketamine during emergency care present aggravated early post- traumatic stress reaction which is highly predictive of post-traumatic stress disorder (PTSD) development and severity. The use of ketamine in the acute trauma phase may directly or indirectly interfere with neural processes of memory consolidation of the traumatic event, thus leading to the formation of maladaptive memories, a hallmark symptom of PTSD. We have recently shown that ketamine anesthesia, immediately after a traumatic event, enhances memory consolidation and leads to long-lasting alterations of social behavior in rats. Based on the evidence that ketamine induces a robust central and peripheral adrenergic/noradrenergic potentiation and that activation of this system is essential for the formation of memory for stressful events, we explored the possibility that the strong sympathomimetic action of ketamine might underlie its memory enhancing effects. We found that rats given immediate, but not delayed, post-training ketamine anesthesia (125 mg/kg) presented enhanced 48-h memory retention in an inhibitory avoidance task and that these effects were blocked by adrenal medullectomy, lesions of the locus coeruleus, systemic or intra-basolateral amygdala ß-adrenergic receptor antagonism. Thus, the memory enhancing effects of ketamine anesthesia are time-dependent and mediated by a combined peripheral-central sympathomimetic action. We elucidated a mechanism by which ketamine exacerbates acute post-traumatic reaction, possibly leading to development of PTSD symptomatology later in life. These findings will help guide for a better management of sedation/anesthesia in emergency care to promote the prophylaxis and reduce the risk of developing trauma-related disorders in trauma victims.


Asunto(s)
Neuronas Adrenérgicas/efectos de los fármacos , Anestésicos Disociativos/administración & dosificación , Complejo Nuclear Basolateral/efectos de los fármacos , Miedo/efectos de los fármacos , Ketamina/administración & dosificación , Consolidación de la Memoria/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Animales , Nivel de Alerta/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Ratas , Trastornos por Estrés Postraumático/metabolismo
7.
Int J Neuropsychopharmacol ; 24(7): 570-579, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-33674836

RESUMEN

BACKGROUND: Clinical studies have shown that the rapid antidepressant effect of the glutamate N-methyl-D-aspartate receptor antagonist ketamine generally disappears within 1 week but can be maintained by repeated administration. Preclinical studies showed that a single ketamine injection immediately increases the firing and burst activity of norepinephrine (NE) neurons, but not that of serotonin (5-HT) neurons. It also enhances the population activity of dopamine (DA) neurons. In the present study, we investigated whether such alterations of monoamine neuronal firing are still present 1 day after a single injection, and whether they can be maintained by repeated injections. METHODS: Rats received a single ketamine injection or 6 over 2 weeks and the firing activity of dorsal raphe nucleus 5-HT, locus coeruleus NE, and ventral tegmental area DA neurons was assessed. RESULTS: One day following a single injection of ketamine, there was no change in the firing activity of 5-HT, NE, or DA neurons. One day after repeated ketamine administration, however, there was a robust increase of the firing activity of NE neurons and an enhancement of burst and population activities of DA neurons, but still no change in firing parameters of 5-HT neurons. The increased activity of NE neurons was no longer present 3 days after the last injection, whereas that of DA neurons was still present. DA neurons were firing normally 7 days after repeated injections. CONCLUSION: These results imply that the enhanced activity of NE and DA neurons may play a significant role in the maintenance of the antidepressant action of ketamine.


Asunto(s)
Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Ketamina/farmacología , Tegmento Mesencefálico/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Ketamina/administración & dosificación , Locus Coeruleus/efectos de los fármacos , Masculino , Núcleos del Rafe/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Neuronas Serotoninérgicas/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos
8.
Int J Mol Sci ; 22(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34681746

RESUMEN

Noradrenaline (NE) is a catecholamine acting as both a neurotransmitter and a hormone, with relevant effects in modulating feeding behavior and satiety. Several studies have assessed the relationship between the noradrenergic system and Eating Disorders (EDs). This systematic review aims to report the existing literature on the role of the noradrenergic system in the development and treatment of EDs. A total of 35 studies were included. Preclinical studies demonstrated an involvement of the noradrenergic pathways in binge-like behaviors. Genetic studies on polymorphisms in genes coding for NE transporters and regulating enzymes have shown conflicting evidence. Clinical studies have reported non-unanimous evidence for the existence of absolute alterations in plasma NE values in patients with Anorexia Nervosa (AN) and Bulimia Nervosa (BN). Pharmacological studies have documented the efficacy of noradrenaline-modulating therapies in the treatment of BN and Binge Eating Disorder (BED). Insufficient evidence was found concerning the noradrenergic-mediated genetics of BED and BN, and psychopharmacological treatments targeting the noradrenergic system in AN. According to these data, further studies are required to expand the existing knowledge on the noradrenergic system as a potential target for treatments of EDs.


Asunto(s)
Encéfalo/metabolismo , Trastornos de Alimentación y de la Ingestión de Alimentos/tratamiento farmacológico , Trastornos de Alimentación y de la Ingestión de Alimentos/etiología , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/genética , Norepinefrina/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Conducta Alimentaria/fisiología , Trastornos de Alimentación y de la Ingestión de Alimentos/diagnóstico por imagen , Humanos , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo
9.
Int J Mol Sci ; 22(24)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34948196

RESUMEN

Although guanethidine (GUA) was used in the past as a drug to suppress hyperactivity of the sympathetic nerve fibers, there are no available data concerning the possible action of this substance on the sensory component of the peripheral nervous system supplying the urinary bladder. Thus, the present study was aimed at disclosing the influence of intravesically instilled GUA on the distribution, relative frequency, and chemical coding of dorsal root ganglion neurons associated with the porcine urinary bladder. The investigated sensory neurons were visualized with a retrograde tracing method using Fast Blue (FB), while their chemical profile was disclosed with single-labeling immunohistochemistry using antibodies against substance P (SP), calcitonin gene-related peptide (CGRP), pituitary adenylate cyclase activating polypeptide (PACAP), galanin (GAL), neuronal nitric oxide synthase (nNOS), somatostatin (SOM), and calbindin (CB). After GUA treatment, a slight decrease in the number of FB+ neurons containing SP was observed when compared with untreated animals (34.6 ± 6.5% vs. 45.6 ± 1.3%), while the number of retrogradely traced cells immunolabeled for GAL, nNOS, and CB distinctly increased (12.3 ± 1.0% vs. 7.4 ± 0.6%, 11.9 ± 0.6% vs. 5.4 ± 0.5% and 8.6 ± 0.5% vs. 2.7 ± 0.4%, respectively). However, administration of GUA did not change the number of FB+ neurons containing CGRP, PACAP, or SOM. The present study provides evidence that GUA significantly modifies the sensory innervation of the porcine urinary bladder wall and thus may be considered a potential tool for studying the plasticity of this subdivision of the bladder innervation.


Asunto(s)
Ganglios Espinales/metabolismo , Guanetidina/farmacología , Vejiga Urinaria/inervación , Antagonistas Adrenérgicos/farmacología , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Animales , Calbindinas/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Femenino , Galanina/metabolismo , Ganglios Espinales/efectos de los fármacos , Guanetidina/metabolismo , Neurotoxinas/farmacología , Óxido Nítrico Sintasa/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Células Receptoras Sensoriales/metabolismo , Somatostatina/metabolismo , Sustancia P/metabolismo , Porcinos , Vejiga Urinaria/efectos de los fármacos
10.
Neurobiol Dis ; 134: 104616, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31678403

RESUMEN

The pontine nucleus locus coeruleus (LC) is the primary source of noradrenergic (NE) projections to the brain and is important for working memory, attention, and cognitive flexibility. Individuals with Down syndrome (DS) develop Alzheimer's disease (AD) with high penetrance and often exhibit working memory deficits coupled with degeneration of LC-NE neurons early in the progression of AD pathology. Designer receptors exclusively activated by designer drugs (DREADDs) are chemogenetic tools that allow targeted manipulation of discrete neuronal populations in the brain without the confounds of off-target effects. We utilized male Ts65Dn mice (a mouse model for DS), and male normosomic (NS) controls to examine the effects of inhibitory DREADDs delivered via an AAV vector under translational control of the synthetic PRSx8, dopamine ß hydroxylase (DßH) promoter. This chemogenetic tool allowed LC inhibition upon administration of the inert DREADD ligand, clozapine-N-oxide (CNO). DREADD-mediated LC inhibition impaired performance in a novel object recognition task and reversal learning in a spatial task. DREADD-mediated LC inhibition gave rise to an elevation of α-adrenoreceptors both in NS and in Ts65Dn mice. Further, microglial markers showed that the inhibitory DREADD stimulation led to increased microglial activation in the hippocampus in Ts65Dn but not in NS mice. These findings strongly suggest that LC signaling is important for intact memory and learning in Ts65Dn mice and disruption of these neurons leads to increased inflammation and dysregulation of adrenergic receptors.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Síndrome de Down/metabolismo , Locus Coeruleus/metabolismo , Trastornos de la Memoria/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Animales , Drogas de Diseño , Modelos Animales de Enfermedad , Síndrome de Down/complicaciones , Locus Coeruleus/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos
11.
FASEB J ; 33(6): 7252-7260, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30860868

RESUMEN

Electroencephalogram monitoring during propofol (PRO) anesthesia typically features low-frequency oscillations, which may be involved with thalamic reticular nucleus (TRN) modulation. TRN receives noradrenergic inputs from the locus coeruleus (LC). We hypothesized that specific noradrenergic connections in the TRN may contribute to the emergence from PRO anesthesia. Intranuclei norepinephrine (NE) injections (n = 10) and designer receptors exclusively activated by designer drugs (DREADDs) (n = 10) were used to investigate the role of noradrenergic inputs from the LC to the TRN during PRO anesthesia. Whole-cell recording in acute brain slice preparations was used to identify the type of adrenoceptor that regulates noradrenergic innervation in the TRN. An intracerebral injection of NE into the TRN delays arousal in mice recovering from PRO anesthesia (means ± sd; 486.6 ± 57.32 s for the NE injection group vs. 422.4 ± 48.19 s for the control group; P = 0.0143) and increases the cortical-δ (0.1-4 Hz, 25.4 ± 2.9 for the NE injection group vs. 21.0 ± 1.7 for the control group; P = 0.0094) oscillation. An intra-TRN injection of NE also decreased the EC50 of PRO-induced unconsciousness (57.05 ± 1.78 mg/kg for the NE injection group vs. 72.44 ± 3.23 mg/kg for the control group; P = 0.0096). Moreover, the activation of LC-noradrenergic nerve terminals in the TRN using DREADDs increased the recovery time [466.1 ± 44.57 s for the clozapine N-oxide (CNO) injection group vs. 426.1 ± 38.75 s for the control group; P = 0.0033], decreased the EC50 of PRO-induced unconsciousness (64.77 ± 3.40 mg/kg for the CNO injection group vs. 74.00 ± 2.08 mg/kg for the control group; P = 0.0081), and increased the cortical-δ oscillation during PRO anesthesia (23.29 ± 2.58 for the CNO injection group vs. 19.56 ± 1.9 for the control group; P = 0.0213). In addition, whole-cell recording revealed that NE augmented the inhibitory postsynaptic currents in the TRN neurons via the α1-adrenoceptor. Our data indicated that enhanced NE signaling at the noradrenergic terminals of the LC-TRN projection delays arousal from general anesthesia, which is likely mediated by the α1-adrenoceptor activation. Our findings open a door for further understanding of the functions of various LC targets in both anesthesia and arousal.-Zhang, Y., Fu, B., Liu, C., Yu, S., Luo, T., Zhang, L., Zhou, W., Yu, T. Activation of noradrenergic terminals in the reticular thalamus delays arousal from propofol anesthesia in mice.


Asunto(s)
Neuronas Adrenérgicas/fisiología , Anestesia General , Nivel de Alerta/fisiología , Retraso en el Despertar Posanestésico/fisiopatología , Núcleos Talámicos Intralaminares/fisiopatología , Terminaciones Nerviosas/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Neuronas Adrenérgicas/efectos de los fármacos , Anestésicos Intravenosos , Animales , Clozapina/análogos & derivados , Clozapina/farmacología , Drogas de Diseño/farmacología , Electroencefalografía , Vectores Genéticos/administración & dosificación , Núcleos Talámicos Intralaminares/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Terminaciones Nerviosas/efectos de los fármacos , Norepinefrina/farmacología , Técnicas de Placa-Clamp , Propofol , Distribución Aleatoria , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/fisiología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Reflejo de Enderezamiento/efectos de los fármacos , Método Simple Ciego , Organismos Libres de Patógenos Específicos , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/fisiología
12.
Circ Res ; 122(7): 928-932, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29436388

RESUMEN

RATIONALE: DPP-4 (dipeptidyl peptidase-4) inhibitors have increased the risk of heart failure events in both randomized clinical trials and observational studies, but the mechanisms that underlie their deleterious effect remain to be elucidated. Previous work has implicated a role of these drugs to promote cardiac fibrosis. OBJECTIVE: This article postulates that DPP-4 inhibitors increase the risk of heart failure events by activating the sympathetic nervous system to stimulate cardiomyocyte cell death, and it crystallizes the findings from both experimental studies and clinical trials that support the hypothesis. METHODS AND RESULTS: Inhibition of DPP-4 not only potentiates the actions of GLP-1 (glucagon-like peptide-1; which can increase myocardial cAMP) but also potentiates the actions of SDF-1 (stromal cell-derived factor 1), NPY (neuropeptide Y), and substance P to activate the sympathetic nervous system and stimulate ß-adrenergic receptors to cause cardiomyocyte apoptosis, presumably through a CaMKII (Ca++/calmodulin-dependent protein kinase II) pathway. An action of SDF-1 to interfere with cAMP and protein kinase A signaling may account for the absence of a clinically overt positive chronotropic effect. This conceptual framework is supported by the apparent ability of ß-blocking drugs to attenuate the increased risk of DPP-4 inhibitors in a large-scale clinical trial. CONCLUSIONS: Sympathetic activation may explain the increased risk of heart failure produced by DPP-4 inhibitors. The proposed mechanism has major implications for clinical care because in the treatment of patients with type 2 diabetes mellitus, DPP-4 inhibitors are widely prescribed, but ß-blockers are underutilized because of fears that they might mask hypoglycemia.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/toxicidad , Insuficiencia Cardíaca/etiología , Hipoglucemiantes/toxicidad , Sistema Nervioso Simpático/efectos de los fármacos , Neuronas Adrenérgicas/efectos de los fármacos , Animales , Cardiotoxicidad , Ensayos Clínicos como Asunto , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Humanos , Hipoglucemiantes/farmacología , Sistema Nervioso Simpático/citología
13.
J Nucl Cardiol ; 27(3): 931-939, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-30569409

RESUMEN

BACKGROUND: Doxorubicin is the mainstay of curative lymphoma treatment but is associated with a dose-dependent cardiotoxicity that is often recognized too late to avoid substantial irreversible cardiac injury. Iodine-123 metaiodobenzylguanidine (123I-MIBG) is a gamma-emitting tracer that mimics noradrenaline uptake, storage, and release mechanisms in adrenergic presynaptic neurons. 123I-MIBG scintigraphy can be used for assessment of doxorubicin-induced injury to myocardial adrenergic neurons during treatment and could be the tool for early detection of doxorubicin cardiotoxicity, which is currently lacking. METHODS AND RESULTS: A total of 37 lymphoma patients scheduled for doxorubicin treatment were included in our study. 123I-MIBG imaging was performed prior to chemotherapy and after a median of 4 cycles of doxorubicin. Early and late heart-to-mediastinum ratios (H/Mearly and H/Mlate) and washout rate (WOR) were used for evaluation of cardiotoxicity. The prognostic value of 123I-MIBG results was assessed using left ventricular ejection fraction (LVEF) as measured by cardiac magnetic resonance at 1-year follow-up. We found a post-therapy increase in WOR (including nine patients with > 10% increase), which was not statistically significant (18.6 vs 23.4%, P = 0.09). The difference appeared to be driven by an increase in H/Mearly. LVEF decreased from baseline to 1-year follow-up (64 vs 58%, P = 0.03). LVEF change was not associated with changes in WOR (P = 0.5). CONCLUSION: The present study does not provide evidence for 123I-MIBG imaging as a clinically applicable tool for early detection of doxorubicin-induced cardiotoxicity.


Asunto(s)
3-Yodobencilguanidina , Doxorrubicina/efectos adversos , Corazón/efectos de los fármacos , Corazón/diagnóstico por imagen , Radioisótopos de Yodo , Linfoma/diagnóstico por imagen , Neuronas Adrenérgicas/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Cardiotoxicidad , Femenino , Corazón/inervación , Cardiopatías/inducido químicamente , Humanos , Linfoma/complicaciones , Linfoma/tratamiento farmacológico , Masculino , Mediastino/diagnóstico por imagen , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Cintigrafía , Volumen Sistólico , Adulto Joven
14.
J Pharmacol Exp Ther ; 371(2): 453-475, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31492824

RESUMEN

In December 2018, the Centers for Disease Control declared fentanyl the deadliest drug in America. Opioid overdose is the single greatest cause of death in the United States adult population (ages 18-50), and fentanyl and its analogs [fentanyl/fentanyl analogs (F/FAs)] are currently involved in >50% of these deaths. Anesthesiologists in the United States were introduced to fentanyl in the early 1970s when it revolutionized surgical anesthesia by combining profound analgesia with hemodynamic stability. However, they quickly had to master its unique side effect. F/FAs can produce profound rigidity in the diaphragm, chest wall and upper airway within an extremely narrow dosing range. This clinical effect was called wooden chest syndrome (WCS) by anesthesiologists and is not commonly known outside of anesthesiology or to clinicians or researchers in addiction research/medicine. WCS is almost routinely fatal without expert airway management. This review provides relevant clinical human pharmacology and animal data demonstrating that the significant increase in the number of F/FA-induced deaths may involve α-adrenergic and cholinergic receptor-mediated mechanical failure of the respiratory and cardiovascular systems with rapid development of rigidity and airway closure. Although morphine and its prodrug, heroin, can cause mild rigidity in abdominal muscles at high doses, neither presents with the distinct and rapid respiratory failure seen with F/FA-induced WCS, separating F/FA overdose from the slower onset of respiratory depression caused by morphine-derived alkaloids. This distinction has significant consequences for the design and implementation of new pharmacologic strategies to effectively prevent F/FA-induced death. SIGNIFICANCE STATEMENT: Deaths from fentanyl and F/FAs are increasing in spite of availability and awareness of the opioid reversal drug naloxone. This article reviews literature suggesting that naloxone may be ineffective against centrally mediated noradrenergic and cholinergic effects of F/FAs, which clinically manifest as severe muscle rigidity and airway compromise (e.g., wooden chest syndrome) that is rapid and distinct from respiratory depression seen with morphine-derived alkaloids. A physiologic model is proposed and implications for new drug development and treatment are discussed.


Asunto(s)
Neuronas Adrenérgicas/efectos de los fármacos , Analgésicos Opioides/efectos adversos , Fentanilo/efectos adversos , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Epidemia de Opioides/prevención & control , Neuronas Adrenérgicas/metabolismo , Analgésicos Opioides/metabolismo , Sobredosis de Droga/metabolismo , Sobredosis de Droga/prevención & control , Fentanilo/metabolismo , Humanos , Rigidez Muscular/inducido químicamente , Rigidez Muscular/tratamiento farmacológico , Rigidez Muscular/metabolismo , Naloxona/metabolismo , Antagonistas de Narcóticos/metabolismo , Epidemia de Opioides/tendencias , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/metabolismo , Tiempo de Tratamiento/tendencias
15.
Alcohol Clin Exp Res ; 43(8): 1695-1701, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31141179

RESUMEN

BACKGROUND: Relapse is a critical barrier to effective long-term treatment of alcoholism, and stress is often cited as a key trigger to relapse. Numerous studies suggest that stress-induced reinstatement to drug-seeking behaviors is mediated by norepinephrine (NE) and corticotropin-releasing factor (CRF) signaling interactions in the bed nucleus of the stria terminalis (BNST), a brain region critical to many behavioral and physiologic responses to stressors. Here, we sought to directly examine the effects of NE on BNST CRF neuron activity and determine whether these effects may be modulated by chronic intermittent EtOH (CIE) exposure or a single restraint stress. METHODS: Adult male CRF-tomato reporter mice were treatment-naïve, or either exposed to CIE for 2 weeks or to a single 1-hour restraint stress. Effects of application of exogenous NE on BNST CRF neuron activity were assessed via whole-cell patch-clamp electrophysiological techniques. RESULTS: We found that NE depolarized BNST CRF neurons in naïve mice in a ß-adrenergic receptor (AR)-dependent mechanism. CRF neurons from CIE- or stress-exposed mice had significantly elevated basal resting membrane potential compared to naïve mice. Furthermore, CIE and stress individually disrupted the ability of NE to depolarize CRF neurons, suggesting that both stress and CIE utilize ß-AR signaling to modulate BNST CRF neurons. Neither stress nor CIE altered the ability of exogenous NE to inhibit evoked glutamatergic transmission onto BNST CRF neurons as shown in naïve mice, a mechanism previously shown to be α-AR-dependent. CONCLUSIONS: Altogether, these findings suggest that stress and CIE interact with ß-AR signaling to modulate BNST CRF neuron activity, potentially disrupting the α/ß-AR balance of BNST CRF neuronal excitability. Restoration of α/ß-AR balance may lead to novel therapies for the alleviation of many stress-related disorders.


Asunto(s)
Neuronas Adrenérgicas/fisiología , Hormona Liberadora de Corticotropina/fisiología , Etanol/efectos adversos , Norepinefrina/farmacología , Restricción Física/fisiología , Núcleos Septales/fisiología , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Animales , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Técnicas de Sustitución del Gen , Ácido Glutámico/fisiología , Ácido Quinurénico/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Norepinefrina/antagonistas & inhibidores , Picrotoxina/farmacología , Propranolol/farmacología , Núcleos Septales/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/fisiopatología
16.
Addict Biol ; 24(3): 509-521, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-29480583

RESUMEN

Effects of stress on the reward system are well established in the literature. Although previous studies have revealed that stress can reinstate extinguished addictive behaviors related to cocaine, the effects of stress on the rewarding memory of cocaine are not fully understood. Here, we provide evidence that stress potentiates the expression of rewarding memory of cocaine via the activation of brainstem-reward circuitry using a cocaine-induced conditioned place preference (CPP) paradigm combined with restraint stress in rats. The rats exposed to 30-minute restraint stress immediately before posttest exhibited significantly larger CPP scores compared with non-stressed rats. Intra-laterodorsal tegmental nucleus (LDT) microinjection of a ß or α2 adrenoceptor antagonist attenuated the stress-induced enhancement of cocaine CPP. Consistent with this observation, intra-LDT microinjection of a ß or α2 adrenoceptor agonist before posttest increased cocaine CPP. Additionally, intra-ventral tegmental area (VTA) microinjection of antagonists for the muscarinic acetylcholine, nicotinic acetylcholine or glutamate receptors attenuated the stress-induced enhancement of cocaine CPP. Finally, intra-medial prefrontal cortex (mPFC) microinjection of a D1 receptor antagonist also reduced the stress-induced enhancement of cocaine CPP. These findings suggest a mechanism wherein the LDT is activated by noradrenergic input from the locus coeruleus, leading to the activation of VTA dopamine neurons via both cholinergic and glutamatergic transmission and the subsequent excitation of the mPFC to enhance the memory of cocaine-induced reward value.


Asunto(s)
Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Memoria/efectos de los fármacos , Recompensa , Neuronas Adrenérgicas/efectos de los fármacos , Antagonistas Adrenérgicos alfa/farmacología , Animales , Tronco Encefálico/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Idazoxan/análogos & derivados , Idazoxan/farmacología , Masculino , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 2/fisiología , Receptores Adrenérgicos beta/fisiología , Restricción Física , Estrés Psicológico/fisiopatología , Tegmento Mesencefálico/efectos de los fármacos , Timolol/farmacología
17.
Neural Plast ; 2019: 3152129, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30804990

RESUMEN

Prenatal stress (PS) has long-term sequelae for the morphological and functional status of the central nervous system of the progeny. A PS-induced proinflammatory status of the organism may result in an impairment of both hippocampal synaptic plasticity and hippocampus-dependent memory formation in adults. We addressed here the question of how PS-induced alterations in the immune response in young and old mice may contribute to changes in hippocampal function in aging. Immune stimulation (via LPS injection) significantly affected the ability of the hippocampal CA3-CA1 synapse of PS mice to undergo long-term potentiation (LTP). Elevated corticosterone level in the blood of aged PS mice that is known to influence LTP magnitude indicates a chronic activation of the HPA axis due to the in utero stress exposure. We investigated the contribution of adrenergic receptors to the modulation of hippocampal synaptic plasticity of aged mice and found that impaired LTP in the PS-LPS group was indeed rescued by application of isoproterenol (a nonspecific noradrenergic agonist). Further exploration of the mechanisms of the observed phenomena will add to our understanding of the interaction between PS and proinflammatory immune activation and its contribution to the functional and structural integrity of the aging brain.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Hipocampo/metabolismo , Lipopolisacáridos/farmacología , Potenciación a Largo Plazo/fisiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Estrés Psicológico/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Animales , Corticosterona/sangre , Femenino , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Ratones , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Embarazo
18.
Croat Med J ; 60(4): 352-360, 2019 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-31483121

RESUMEN

AIM: To analyze the effects of glutamatergic agonists and antagonists on the activation of the A1 and A2 noradrenergic neurons localized in caudal ventrolateral medulla and nucleus tractus solitarii, respectively. METHODS: Rats were injected with glutamatergic agonists - kainic acid, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), or N-methyl-D-aspartic acid (NMDA), and the brain sections were prepared for immunohistochemistry. Before agonist injections, antagonists - 6-cyano-7-nitroquinoxaline-2,3-dione or dizocilpine were administered. The expression of c-Fos, as the neuronal activation marker, and tyrosine hydroxylase (TH), as the marker of noradrenergic neurons was assessed with dual immunohistochemistry. The percentage of c-Fos-positive noradrenergic neurons relative to all TH-positive neurons in the respective areas of the brain stem was calculated. RESULTS: All three glutamatergic agonists significantly increased the number of the c-Fos-positive noradrenergic neurons in both the A1 and A2 area when compared with control animals. Kainic acid injection activated about 57% of TH-positive neurons in A1 and 40% in A2, AMPA activated 26% in A1 and 38% in A2, and NMDA 77% in A1 and 22% in A2. The injections of appropriate glutamatergic antagonists greatly decreased the number of activated noradrenergic neurons. CONCLUSION: Our results suggest that noradrenergic neurons are regulated and/or activated by glutamatergic system and that these neurons express functional glutamate receptors.


Asunto(s)
Neuronas Adrenérgicas/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Fármacos actuantes sobre Aminoácidos Excitadores/agonistas , Fármacos actuantes sobre Aminoácidos Excitadores/antagonistas & inhibidores , Animales , Femenino , Inmunohistoquímica , Ácido Kaínico/farmacología , N-Metilaspartato/farmacología , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/efectos de los fármacos , Tirosina 3-Monooxigenasa/biosíntesis , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
19.
J Neurosci ; 37(29): 6938-6945, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28626012

RESUMEN

State-dependent activity of locus ceruleus (LC) neurons has long suggested a role for noradrenergic modulation of arousal. However, in vivo insights into noradrenergic arousal circuitry have been constrained by the fundamental inaccessibility of the human brain for invasive studies. Functional magnetic resonance imaging (fMRI) studies performed during site-specific pharmacological manipulations of arousal levels may be used to study brain arousal circuitry. Dexmedetomidine is an anesthetic that alters the level of arousal by selectively targeting α2 adrenergic receptors on LC neurons, resulting in reduced firing rate and norepinephrine release. Thus, we hypothesized that dexmedetomidine-induced altered arousal would manifest with reduced functional connectivity between the LC and key brain regions involved in the regulation of arousal. To test this hypothesis, we acquired resting-state fMRI data in right-handed healthy volunteers 18-36 years of age (n = 15, 6 males) at baseline, during dexmedetomidine-induced altered arousal, and recovery states. As previously reported, seed-based resting-state fMRI analyses revealed that the LC was functionally connected to a broad network of regions including the reticular formation, basal ganglia, thalamus, posterior cingulate cortex (PCC), precuneus, and cerebellum. Functional connectivity of the LC to only a subset of these regions (PCC, thalamus, and caudate nucleus) covaried with the level of arousal. Functional connectivity of the PCC to the ventral tegmental area/pontine reticular formation and thalamus, in addition to the LC, also covaried with the level of arousal. We propose a framework in which the LC, PCC, thalamus, and basal ganglia comprise a functional arousal circuitry.SIGNIFICANCE STATEMENT Electrophysiological studies of locus ceruleus (LC) neurons have long suggested a role for noradrenergic mechanisms in mediating arousal. However, the fundamental inaccessibility of the human brain for invasive studies has limited a precise understanding of putative brain regions that integrate with the LC to regulate arousal. Our results suggest that the PCC, thalamus, and basal ganglia are key components of a LC-noradrenergic arousal circuit.


Asunto(s)
Neuronas Adrenérgicas/fisiología , Nivel de Alerta/fisiología , Dexmedetomidina/administración & dosificación , Locus Coeruleus/fisiología , Red Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Adolescente , Neuronas Adrenérgicas/efectos de los fármacos , Agonistas de Receptores Adrenérgicos alfa 2 , Adulto , Nivel de Alerta/efectos de los fármacos , Conectoma/métodos , Femenino , Humanos , Hipnóticos y Sedantes/administración & dosificación , Locus Coeruleus/efectos de los fármacos , Imagen por Resonancia Magnética/métodos , Masculino , Red Nerviosa/efectos de los fármacos , Vías Nerviosas/diagnóstico por imagen , Vías Nerviosas/fisiología , Plasticidad Neuronal/efectos de los fármacos , Adulto Joven
20.
Pflugers Arch ; 470(11): 1659-1672, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30054719

RESUMEN

Optogenetic stimulation of the adrenergic C1 neurons produces cardiorespiratory activation, and selective depletion of these cells attenuates breathing responses induced by hypoxia. The preBötzinger complex (preBötC) is a group of neurons located in the intermediate aspect of the ventrolateral medulla, critical for respiratory rhythmogenesis, and is modulated by glutamate and catecholamines. Our hypothesis is that selective activation of C1 neurons leads to breathing responses by excitatory connections with the preBötC neurons. Anatomical connection between C1 cells and preBötC was evaluated using retrograde (Cholera Toxin b; preBötC) and anterograde (LVV-PRSx8-ChR2-eYFP; C1 region) tracers. LVV-PRSx8-ChR2-eYFP (viral vector that expresses channelrhodopsin-2 (ChR2) under the control of the catecholaminergic neuron-preferring promoter (PRSx8) was also injected into the C1 region of male Wistar rats for the functional experiments. Anatomical results demonstrated that preBötC neurons receive projections from C1 cells, and these projections express tyrosine hydroxylase and vesicular glutamate transporter 2. Functional connection between C1 cells and preBötC was evaluated by photostimulation of ChR2-transduced C1 neurons before and after unilateral injection of the ionotropic glutamate antagonist, kynurenic acid (kyn), or cocktail of adrenergic antagonists in the preBötC. Kyn injection into preBötC blocked the increase in DiaEMG frequency without changing the MAP increase elicited by photostimulation of C1 neurons, while the injection of adrenergic antagonists into the preBötC did not change DiaEMG frequency and MAP increase induced by photostimulation of C1 cells. Our results suggest that the increase in breathing produced by photostimulation of C1 neurons can be caused by a direct glutamatergic activation of preBötC neurons.


Asunto(s)
Neuronas Adrenérgicas/fisiología , Respiración , Centro Respiratorio/fisiología , Antagonistas Adrenérgicos/farmacología , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Animales , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/fisiología , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Quinurénico/farmacología , Masculino , Optogenética , Ratas , Ratas Wistar , Centro Respiratorio/citología , Centro Respiratorio/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA