Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain Res Bull ; 202: 110724, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37543295

RESUMEN

BACKGROUND: Ketamine, despite its efficacy in treating depression, raises concerns regarding safety due to potential abuse, cognitive impairment, and bladder toxicity. Ketamine can affect the locus coeruleus (LC) norepinephrine and attention networks. This study explored the protective effects of electroacupuncture (EA) on the LC of rats exposed to repeated administration of ketamine while investigating the potential role of the Calcium CaM-dependent protein kinase II (CAMK II)/ cAMP response element binding protein (CREB) pathway in mediating EA's impact on ketamine-induced neuronal injury in LC. METHODS: Rats were repeatedly injected intraperitoneally with ketamine hydrochloride (50 mg/kg) once daily for seven days. Subsequently, EA was performed at the acupoints "Zusanli" (ST36) and "Sanyinjiao" (SP-6) once daily following ketamine administration. The Morris water maze test was employed to assess behavioral changes in the rats. Neuronal injury was examined using Nissl staining, and the expression of CAMK II, CREB, and phospho-CREB (p-CREB) was evaluated through immunohistochemistry and western blotting. RESULTS: EA mitigated the cognitive and exploratory impairments and attenuated neuronal injury in the LC induced by repeated administration of ketamine. The expression of CAMK II and p-CREB proteins in the LC increased following 7 days of ketamine administration. However, EA treatment led to a downregulation of CAMK II and p-CREB expression. CONCLUSION: Repeated administration of ketamine in male rats can lead to neuronal injury and neurobehavioral dysfunction. However, EA was found to ameliorate neurodegeneration in the LC and enhance neurobehavioral symptoms. This therapeutic effect of EA may be attributed to its modulation of the CAMKII/CREB pathway, thereby mitigating the aforementioned adverse effects.


Asunto(s)
Electroacupuntura , Ketamina , Ratas , Masculino , Animales , Locus Coeruleus/metabolismo , Ratas Sprague-Dawley , Ketamina/toxicidad , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo
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.
Brain ; 146(6): 2259-2267, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36625030

RESUMEN

The CNS houses naturally occurring pathways that project from the brain to modulate spinal neuronal activity. The noradrenergic locus coeruleus (the A6 nucleus) originates such a descending control whose influence on pain modulation encompasses an interaction with a spinally projecting non-cerulean noradrenergic cell group. Hypothesizing the origin of an endogenous pain inhibitory pathway, our aim was to identify this cell group. A5 and A7 noradrenergic nuclei also spinally project. We probed their activity using an array of optogenetic manipulation techniques during in vivo electrophysiological experimentation. Interestingly, noxious stimulus evoked spinal neuronal firing was decreased upon opto-activation of A5 neurons (two-way ANOVA with Tukey post hoc, P < 0.0001). Hypothesizing that this may reflect activity in the noradrenergic diffuse noxious inhibitory control circuit, itself activated upon application of a conditioning stimulus, we opto-inhibited A5 neurons with concurrent conditioning stimulus application. Surprisingly, no spinal neuronal inhibition was observed; activity in the diffuse noxious inhibitory control circuit was abolished (two-way ANOVA, P < 0.0001). We propose that the A5 nucleus is a critical relay nucleus for mediation of diffuse noxious inhibitory controls. Given the plasticity of diffuse noxious inhibitory controls in disease, and its back and forward clinical translation, our data reveal a potential therapeutic target.


Asunto(s)
Control Inhibidor Nocivo Difuso , Humanos , Control Inhibidor Nocivo Difuso/fisiología , Dolor/metabolismo , Neuronas/metabolismo , Locus Coeruleus/metabolismo , Encéfalo/metabolismo , Norepinefrina/metabolismo , Médula Espinal/metabolismo
4.
Ann Neurol ; 91(2): 282-288, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34981555

RESUMEN

Narcolepsy type 1 (NT1) is a chronic sleep disorder correlated with loss of hypocretin(orexin). In NT1 post-mortem brains, we observed 88% reduction in corticotropin-releasing hormone (CRH)-positive neurons in the paraventricular nucleus (PVN) and significantly less CRH-positive fibers in the median eminence, whereas CRH-neurons in the locus coeruleus and thalamus, and other PVN neuronal populations were spared: that is, vasopressin, oxytocin, tyrosine hydroxylase, and thyrotropin releasing hormone-expressing neurons. Other hypothalamic cell groups, that is, the suprachiasmatic, ventrolateral preoptic, infundibular, and supraoptic nuclei and nucleus basalis of Meynert, were unaffected. The surprising selective decrease in CRH-neurons provide novel targets for diagnostics and therapeutic interventions. ANN NEUROL 2022;91:282-288.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Hipotálamo/metabolismo , Hipotálamo/patología , Narcolepsia/patología , Neuronas/patología , Anciano , Anciano de 80 o más Años , Recuento de Células , Femenino , Humanos , Hipotálamo/diagnóstico por imagen , Inmunohistoquímica , Locus Coeruleus/citología , Locus Coeruleus/diagnóstico por imagen , Locus Coeruleus/metabolismo , Masculino , Eminencia Media/citología , Eminencia Media/diagnóstico por imagen , Eminencia Media/metabolismo , Persona de Mediana Edad , Narcolepsia/diagnóstico por imagen , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/diagnóstico por imagen , Núcleo Hipotalámico Paraventricular/metabolismo
5.
Biomolecules ; 10(7)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32630356

RESUMEN

Noncompetitive N-methyl-D-aspartate/glutamate receptor (NMDAR) antagonists contribute to the pathophysiology of schizophrenia and mood disorders but improve monoaminergic antidepressant-resistant mood disorder and suicidal ideation. The mechanisms of the double-edged sword clinical action of NMDAR antagonists remained to be clarified. The present study determined the interaction between the NMDAR antagonist (MK801), α1 adrenoceptor antagonist (prazosin), and α2A adrenoceptor agonist (guanfacine) on mesocortical and mesothalamic catecholaminergic transmission, and thalamocortical glutamatergic transmission using multiprobe microdialysis. The inhibition of NMDAR in the locus coeruleus (LC) by local MK801 administration enhanced both the mesocortical noradrenergic and catecholaminergic coreleasing (norepinephrine and dopamine) transmissions. The mesothalamic noradrenergic transmission was also enhanced by local MK801 administration in the LC. These mesocortical and mesothalamic transmissions were activated by intra-LC disinhibition of transmission of γ-aminobutyric acid (GABA) via NMDAR inhibition. Contrastingly, activated mesothalamic noradrenergic transmission by MK801 enhanced intrathalamic GABAergic inhibition via the α1 adrenoceptor, resulting in the suppression of thalamocortical glutamatergic transmission. The thalamocortical glutamatergic terminal stimulated the presynaptically mesocortical catecholaminergic coreleasing terminal in the superficial cortical layers, but did not have contact with the mesocortical selective noradrenergic terminal (which projected terminals to deeper cortical layers). Furthermore, the α2A adrenoceptor suppressed the mesocortical and mesothalamic noradrenergic transmissions somatodendritically in the LC and presynaptically/somatodendritically in the reticular thalamic nucleus (RTN). These discrepancies between the noradrenergic and catecholaminergic transmissions in the mesocortical and mesothalamic pathways probably constitute the double-edged sword clinical action of noncompetitive NMDAR antagonists.


Asunto(s)
Maleato de Dizocilpina/administración & dosificación , Guanfacina/administración & dosificación , Locus Coeruleus/metabolismo , Prazosina/administración & dosificación , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Corteza Cerebral/metabolismo , Maleato de Dizocilpina/farmacología , Dopamina/metabolismo , Guanfacina/farmacología , Hipotálamo/metabolismo , Locus Coeruleus/efectos de los fármacos , Masculino , Microdiálisis/instrumentación , Norepinefrina/metabolismo , Prazosina/farmacología , Ratas , Transmisión Sináptica/efectos de los fármacos
6.
J Neurosci ; 40(22): 4309-4322, 2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32317389

RESUMEN

Classical studies suggest that growth hormone (GH) secretion is controlled by negative-feedback loops mediated by GH-releasing hormone (GHRH)- or somatostatin-expressing neurons. Catecholamines are known to alter GH secretion and neurons expressing TH are located in several brain areas containing GH-responsive cells. However, whether TH-expressing neurons are required to regulate GH secretion via negative-feedback mechanisms is unknown. In the present study, we showed that between 50% and 90% of TH-expressing neurons in the periventricular, paraventricular, and arcuate hypothalamic nuclei and locus ceruleus of mice exhibited STAT5 phosphorylation (pSTAT5) after an acute GH injection. Ablation of GH receptor (GHR) from TH cells or in the entire brain markedly increased GH pulse secretion and body growth in both male and female mice. In contrast, GHR ablation in cells that express the dopamine transporter (DAT) or dopamine ß-hydroxylase (DBH; marker of noradrenergic/adrenergic cells) did not affect body growth. Nevertheless, less than 50% of TH-expressing neurons in the hypothalamus were found to express DAT. Ablation of GHR in TH cells increased the hypothalamic expression of Ghrh mRNA, although very few GHRH neurons were found to coexpress TH- and GH-induced pSTAT5. In summary, TH neurons that do not express DAT or DBH are required for the autoregulation of GH secretion via a negative-feedback loop. Our findings revealed a critical and previously unidentified group of catecholaminergic interneurons that are apt to sense changes in GH levels and regulate the somatotropic axis in mice.SIGNIFICANCE STATEMENT Textbooks indicate until now that the pulsatile pattern of growth hormone (GH) secretion is primarily controlled by GH-releasing hormone and somatostatin neurons. The regulation of GH secretion relies on the ability of these cells to sense changes in circulating GH levels to adjust pituitary GH secretion within a narrow physiological range. However, our study identifies a specific population of tyrosine hydroxylase-expressing neurons that is critical to autoregulate GH secretion via a negative-feedback loop. The lack of this mechanism in transgenic mice results in aberrant GH secretion and body growth. Since GH plays a key role in cell proliferation, body growth, and metabolism, our findings provide a major advance to understand how the brain regulates the somatotropic axis.


Asunto(s)
Exocitosis , Retroalimentación Fisiológica , Hormona del Crecimiento/metabolismo , Neuronas/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Animales , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Dopamina beta-Hidroxilasa/genética , Dopamina beta-Hidroxilasa/metabolismo , Femenino , Hormona Liberadora de Hormona del Crecimiento/genética , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Tirosina 3-Monooxigenasa/genética
7.
J Neurophysiol ; 123(5): 1739-1755, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32208895

RESUMEN

Brain stimulation approaches are important to gain causal mechanistic insights into the relevance of functional brain regions and/or neurophysiological systems for human cognitive functions. In recent years, transcutaneous vagus nerve stimulation (tVNS) has attracted considerable popularity. It is a noninvasive brain stimulation technique based on the stimulation of the vagus nerve. The stimulation of this nerve activates subcortical nuclei, such as the locus coeruleus and the nucleus of the solitary tract, and from there, the activation propagates to the cortex. Since tVNS is a novel stimulation technique, this literature review outlines a brief historical background of tVNS, before detailing underlying neurophysiological mechanisms of action, stimulation parameters, cognitive effects of tVNS on healthy humans, and, lastly, current challenges and future directions of tVNS research in cognitive functions. Although more research is needed, we conclude that tVNS, by increasing norepineprine (NE) and gamma-aminobutyric acid (GABA) levels, affects NE- and GABA-related cognitive performance. The review provides detailed background information how to use tVNS as a neuromodulatory tool in cognitive neuroscience and outlines important future leads of research on tVNS.


Asunto(s)
Corteza Cerebral/fisiología , Cognición/fisiología , Locus Coeruleus/fisiología , Memoria/fisiología , Norepinefrina/metabolismo , Núcleo Solitario/fisiología , Estimulación Eléctrica Transcutánea del Nervio , Estimulación del Nervio Vago , Ácido gamma-Aminobutírico/metabolismo , Corteza Cerebral/metabolismo , Humanos , Locus Coeruleus/metabolismo , Núcleo Solitario/metabolismo
8.
Eur Neuropsychopharmacol ; 29(4): 482-492, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30878321

RESUMEN

PTSD is heterogeneous disorder that can be long lasting and often has delayed onset following exposure to a traumatic event. Therefore, it is important to take a staging approach to evaluate progression of biological mechanisms of the disease. Here, we begin to evaluate the temporal trajectory of changes following exposure to traumatic stressors in the SPS rat PTSD model. The percent of animals displaying severe anxiety on EPM increased from 17.5% at one week to 57.1% two weeks after SPS stressors, indicating delayed onset or progressive worsening of the symptoms. The LC displayed prolonged activation, and dysbalance of the CRH/NPY systems, with enhanced CRHR1 gene expression, coupled with reduced mRNAs for NPY and Y2R. In the mediobasal hypothalamus, increased CRH mRNA levels were sustained, but there was a flip in alterations of HPA regulatory molecules, GR and FKBP5 and Y5 receptor at two weeks compared to one week. Two weeks after SPS, intranasal NPY at 300 µg/rat, but not 150 µg which was effective after one week, reversed SPS triggered elevated anxiety. It also reversed SPS elicited depressive/despair symptoms and hyperarousal. Overall, the results reveal time-dependent progression in development of anxiety symptoms and molecular impairments in gene expression for CRH and NPY systems in LC and mediobasal hypothalamus by SPS. With longer time afterwards only a higher dose of NPY was effective in reversing behavioral impairments triggered by SPS, indicating that therapeutic approaches should be adjusted according to the degree of biological progression of the disorder.


Asunto(s)
Expresión Génica , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Neuropéptido Y/farmacología , Trastornos por Estrés Postraumático/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/biosíntesis , Masculino , Neuropéptido Y/biosíntesis , Ratas , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Receptores de Neuropéptido Y/biosíntesis , Proteínas de Unión a Tacrolimus/biosíntesis , Factores de Tiempo
9.
Neuron ; 102(4): 745-761.e8, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-30922875

RESUMEN

Norepinephrine (NE) is a key biogenic monoamine neurotransmitter involved in a wide range of physiological processes. However, its precise dynamics and regulation remain poorly characterized, in part due to limitations of available techniques for measuring NE in vivo. Here, we developed a family of GPCR activation-based NE (GRABNE) sensors with a 230% peak ΔF/F0 response to NE, good photostability, nanomolar-to-micromolar sensitivities, sub-second kinetics, and high specificity. Viral- or transgenic-mediated expression of GRABNE sensors was able to detect electrical-stimulation-evoked NE release in the locus coeruleus (LC) of mouse brain slices, looming-evoked NE release in the midbrain of live zebrafish, as well as optogenetically and behaviorally triggered NE release in the LC and hypothalamus of freely moving mice. Thus, GRABNE sensors are robust tools for rapid and specific monitoring of in vivo NE transmission in both physiological and pathological processes.


Asunto(s)
Proteínas Fluorescentes Verdes/genética , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Mesencéfalo/metabolismo , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 2/genética , Animales , Animales Modificados Genéticamente , Estimulación Eléctrica , Técnicas In Vitro , Microscopía Intravital , Ratones , Microscopía Fluorescente , Optogenética , Ingeniería de Proteínas , Pez Cebra
10.
EMBO J ; 37(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30209240

RESUMEN

Stress-induced cortical alertness is maintained by a heightened excitability of noradrenergic neurons innervating, notably, the prefrontal cortex. However, neither the signaling axis linking hypothalamic activation to delayed and lasting noradrenergic excitability nor the molecular cascade gating noradrenaline synthesis is defined. Here, we show that hypothalamic corticotropin-releasing hormone-releasing neurons innervate ependymal cells of the 3rd ventricle to induce ciliary neurotrophic factor (CNTF) release for transport through the brain's aqueductal system. CNTF binding to its cognate receptors on norepinephrinergic neurons in the locus coeruleus then initiates sequential phosphorylation of extracellular signal-regulated kinase 1 and tyrosine hydroxylase with the Ca2+-sensor secretagogin ensuring activity dependence in both rodent and human brains. Both CNTF and secretagogin ablation occlude stress-induced cortical norepinephrine synthesis, ensuing neuronal excitation and behavioral stereotypes. Cumulatively, we identify a multimodal pathway that is rate-limited by CNTF volume transmission and poised to directly convert hypothalamic activation into long-lasting cortical excitability following acute stress.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Estrés Fisiológico , Neuronas Adrenérgicas/patología , Animales , Factor Neurotrófico Ciliar/genética , Hipotálamo/patología , Locus Coeruleus/patología , Ratones , Ratones Noqueados , Ratas
11.
Nat Neurosci ; 21(7): 963-973, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29915192

RESUMEN

The paraventricular nucleus of the thalamus (PVT) is increasingly being recognized as a critical node linking stress detection to the emergence of adaptive behavioral responses to stress. However, despite growing evidence implicating the PVT in stress processing, the neural mechanisms by which stress impacts PVT neurocircuitry and promotes stressed states remain unknown. Here we show that stress exposure drives a rapid and persistent reduction of inhibitory transmission onto projection neurons of the posterior PVT (pPVT). This stress-induced disinhibition of the pPVT was associated with a locus coeruleus-mediated rise in the extracellular concentration of dopamine in the midline thalamus, required the function of dopamine D2 receptors on PVT neurons, and increased sensitivity to stress. Our findings define the locus coeruleus as an important modulator of PVT function: by controlling the inhibitory tone of the pPVT, it modulates the excitability of pPVT projection neurons and controls stress responsivity.


Asunto(s)
Dopamina/metabolismo , Locus Coeruleus/metabolismo , Inhibición Neural/fisiología , Neuronas/metabolismo , Receptores de Dopamina D2/metabolismo , Estrés Fisiológico/fisiología , Tálamo/metabolismo , Animales , Electrochoque , Miedo/fisiología , Femenino , Glicoproteínas de Membrana , Ratones , Vías Nerviosas , Receptores de Interleucina-1
12.
Brain Res ; 1689: 75-88, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29625116

RESUMEN

The amygdaloid nuclear complex has been linked to the regulation of emotional behavior and energy regulation in that emotional stress might cause either reduction or enhancement of eating. We examined hypothalamic neuronal origin of feeding/arousal-related peptidergic fibers containing cocaine- and amphetamine-regulated transcript (CART) and neuropeptide Y (NPY) located in the rat amygdala along with its efferent projections to the brainstem monoaminergic nuclei. First, central (CeA) as well as medial (MeA) amygdala, among several amygdaloid subdivisions, exhibited the most prominent NPY or CART immunostaining which consisted of a substantial number of somata as well as labeled fibers. When we examined hypothalamic neuronal origin of NPY or CART fibers projecting to the CeA and MeA, medial and lateral arcuate nuclei were neuronal origins of NPY and CART fibers, respectively. However, the majority (>70%) of amygdala-projecting CART neurons which co-contained melanin-concentrating hormone (MCH) originated from the lateral hypothalamus (LH), zona incerta (ZI), and dorsal hypothalamic area (DA). This observation implied that the CeA as well as the MeA might receive potent second-order (and downstream) feeding-related CART input from the lateral hypothalamic regions in addition to first-order CART or NPY input from the Arc. Second, a large number of CeA neurons projected to the locus coeruleus (LC), whereas only a small number of MeA cells projected to the dorsal raphe (DR); none of the CeA or MeA cells provided dual projections to the LC and DR. Finally, a portion of MCH cells in the LH, ZI, and DA sent divergent axon collaterals to the CeA and LC. Considering that the CeA sends substantial GABAergic input to the LC, the present observation might serve as an anatomical substrate to support the potent hypnogenic role of MCH neurons in the LH regions during cataplexy and REM sleep.


Asunto(s)
Amígdala del Cerebelo/citología , Núcleo Dorsal del Rafe/citología , Locus Coeruleus/citología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuropéptido Y/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Núcleo Dorsal del Rafe/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Masculino , Melaninas/metabolismo , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Datos Preliminares , Ratas Sprague-Dawley
13.
Neurobiol Dis ; 115: 9-16, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29522818

RESUMEN

BACKGROUND: The majority of patients diagnosed with idiopathic rapid eye movement sleep behaviour disorder (iRBD) progress over time to a Lewy-type α-synucleinopathy such as Parkinson's disease or dementia with Lewy bodies. This in vivo molecular imaging study aimed to investigate if extrastriatal monoaminergic systems are affected in iRBD patients and if this coincides with neuroinflammation. METHODS: We studied twenty-one polysomnography-confirmed iRBD patients with 18F-DOPA and 11C-PK11195 positron emission tomography (PET) to investigate extrastriatal monoaminergic function and microglial activation. Twenty-nine healthy controls (n = 9 18F-DOPA and n = 20 11C-PK11195) were also investigated. Analyses were performed within predefined regions of interest and at voxel-level with Statistical Parametric Mapping. RESULTS: Regions of interest analysis detected monoaminergic dysfunction in iRBD thalamus with a 15% mean reduction of 18F-DOPA Ki values compared to controls (mean difference = -0.00026, 95% confidence interval [-0.00050 to -0.00002], p-value = 0.03). No associated thalamic changes in 11C-PK11195 binding were observed. Other regions sampled showed no 18F-DOPA or 11C-PK11195 PET differences between groups. Voxel-level interrogation of 11C-PK11195 binding identified areas with significantly increased binding within the occipital lobe of iRBD patients. CONCLUSION: Thalamic monoaminergic dysfunction in iRBD patients may reflect terminal dysfunction of projecting neurons from the locus coeruleus and dorsal raphe nucleus, two structures that regulate REM sleep and are known to be involved in the early phase of PD. The observation of significantly raised microglial activation in the occipital lobe of these patients might suggest early local Lewy-type α-synuclein pathology and possibly an increased risk for later cognitive dysfunction.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Núcleo Dorsal del Rafe/metabolismo , Locus Coeruleus/metabolismo , Microglía/metabolismo , Trastorno de la Conducta del Sueño REM/metabolismo , Tálamo/metabolismo , Anciano , Dihidroxifenilalanina/metabolismo , Núcleo Dorsal del Rafe/diagnóstico por imagen , Femenino , Humanos , Locus Coeruleus/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Polisomnografía/métodos , Tomografía de Emisión de Positrones/métodos , Trastorno de la Conducta del Sueño REM/diagnóstico por imagen , Trastorno de la Conducta del Sueño REM/fisiopatología , Tálamo/diagnóstico por imagen
14.
J Anesth ; 32(1): 48-53, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29128909

RESUMEN

BACKGROUND: Neuropeptide S (NPS) is an endogenous neuropeptide controlling anxiolysis, wakefulness, and analgesia. NPS containing neurons exist near to the locus coeruleus (LC) involved in the descending anti-nociceptive system. NPS interacts with central noradrenergic neurons; thus brain noradrenergic signaling may be involved in NPS-induced analgesia. We tested NPS analgesia in noradrenergic neuron-lesioned rats using a selective LC noradrenergic neurotoxin, N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4). METHODS: A total 66 male Sprague-Dawley rats weighing 350-450 g were used. Analgesic effects of NPS were evaluated using hot-plate and tail-flick test with or without DSP-4. The animal allocated into 3 groups; hot-plate with NPS alone intracerebroventricular (icv) (0.0, 1.0, 3.3, and 10.0 nmol), tail-flick NPS alone icv (0.0 and 10.0 nmol), and hot-plate with NPS and DSP-4 (0 or 50 mg/kg ip). In hot-plate with NPS and DSP-4 group, noradrenaline content in the cerebral cortex, pons, hypothalamus, were measured. RESULTS: NPS 10 nmol icv prolonged hot plate (%MPE) but not tail flick latency at 30 and 40 min after administration. DSP-4 50 mg/kg decreased noradrenaline content in the all 3 regions. The NA depletion inhibited NPS analgesic effect in the hot plate test but not tail flick test. There was a significant correlation between hot plate latency (percentage of maximum possible effect: %MPE) with NPS 10 nmol and NA content in the cerebral cortex (p = 0.017, r 2 = 0.346) which noradrenergic innervation arisen mainly from the LC. No other regions had the correlation. CONCLUSIONS: NPS analgesia interacts with LC noradrenergic neuronal activity.


Asunto(s)
Analgésicos/farmacología , Bencilaminas/farmacología , Norepinefrina/metabolismo , Dolor/tratamiento farmacológico , Animales , Encéfalo/metabolismo , Hipotálamo/metabolismo , Locus Coeruleus/metabolismo , Masculino , Neuronas/metabolismo , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley
15.
Zhongguo Zhen Jiu ; 37(5): 513-520, 2017 May 12.
Artículo en Chino | MEDLINE | ID: mdl-29231613

RESUMEN

OBJECTIVE: To observe the effects of electroacupuncture (EA) on pain behavior in rats with bone cancer pain and morphine tolerance, and to explore partial action mechanism. METHODS: Forty-two SD healthy female rats were randomly divided into a sham operation group (7 rats), a bone cancer pain group (8 rats), a morphine tolerance group (9 rats), an EA group (9 rats) and a sham EA group (9 rats). The rats in the sham operation group were treated with injection of phosphate buffer saline at medullary cavity of left-side tibia, and the rats in the remaining groups were injected with MRMT-1 breast cancer cells. After operation, no treatment was given to rats in the sham operation group and bone cancer pain group. 11 days after operation, rats in the morphine tole-rance group, EA group and sham EA group were treated with intraperitoneal injection of morphine hydrochloride, once every 12 hours, for 11 days to establish the model of bone cancer pain and morphine tolerance. One day after the establishment of this bone cancer pain model, the rats in the morphine tolerance group were injected with morphine, once every 12 hours (9:00 a.m. and 9:00 p.m.) for 7 days; the rats in the EA group and sham EA group were injected with morphine at 9:00 a.m., and treated with EA (2 Hz/100 Hz) and sham EA (only injected into the subcutaneous tissue) at bilateral "Zusanli" (ST 36) and "Kunlun" (BL 60), 30 min per treatment, once a day for 7 days. One day before cancer cell injection, 6 days, 8 days, 10 days after operation, after 30 min on 1 days, 5 days, 9 days, 11 days of morphine injection, and after 30 min on 1 days, 3 days, 5 days, 7 days of EA treatment, the paw withdrawal threshold (PWT) was measured in each group. On 11 day of morphine injection, HE staining was applied to observe the morphology and structure change of tibia in the sham operation group, bone cancer pain group and morphine tolerance group, random 2 rats in each group. On 7 days of EA treatment, fluorescent immunohistochemical method was applied to observe the expression of µ-opioid receptor positive cells in nucleus ceruleus in each group, random 4 rats in each one. RESULTS: After 10 days of the cancer cells injection, the PWT of 28 rats of bone cancer pain model (8 rats in the bone cancer pain group, 8 rats in the morphine tolerance group, 6 rats in the EA group and 6 rats in the sham EA group) was significantly lower than that of 7 rats in the sham operation group (P<0.01). After one day of morphine injection, the PWT of the morphine tolerance group, EA group and sham EA group was higher than that of the bone cancer pain group (all P<0.01); on 11 d of morphine injection, the PWT of the morphine tolerance group, EA group and sham EA group was not significantly different from that of the bone cancer pain group (all P>0.05). On 11 d of morphine injection, the tumor induced by cancer cells was observed in upper 1/3 tibia in the bone cancer pain group and morphine tolerance group, and the marrow cavity was filled with MRMT-1 cancer cells; no abnormal change was observed in the sham operation group. On 1 d, 3 d, 5 d and 7 d of EA treatment, the PWT of the cancer pain group, morphine tolerance group and sham EA group was lower than that of the EA group (all P<0.01). On 7 d of EA treatment, the positive expression of MOR in nucleus ceruleus in the cancer pain group, morphine tolerance group, EA group and sham EA group was lower than that in the sham operation group (P<0.01, P<0.05), and that in the cancer pain group, morphine tolerance group and sham EA group was lower than that in the EA group (all P<0.01). CONCLUSIONS: EA can improve mechanical pain threshold in rats with bone cancer pain-morphine tolerance, and improve the abnormal pain, which is likely to be involved with improvement of the MOR positive cells expression in nucleus ceruleus by EA.


Asunto(s)
Dolor en Cáncer/terapia , Electroacupuntura , Locus Coeruleus/metabolismo , Umbral del Dolor , Receptores Opioides mu/metabolismo , Analgésicos Opioides/uso terapéutico , Animales , Dolor en Cáncer/metabolismo , Femenino , Morfina/uso terapéutico , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
16.
J Neuroendocrinol ; 29(8)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28683170

RESUMEN

Gonadotrophin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that inhibits gonadotrophin synthesis and release in birds and mammals. In Japanese quail, GnIH neurones express the noradrenergic receptor and receive noradrenergic innervation. Treatment with noradrenaline (NA) stimulates GnIH release from diencephalic tissue blocks in vitro. However, the effects of NA on hypothalamic GnIH gene expression have not been determined. We investigated noradrenergic regulation of GnIH gene expression in the brain of male quail using the selective noradrenergic neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP-4). We first showed that DSP-4 reduced the number of noradrenergic (dopamine-ß-hydroxylase immunoreactive) cells in the locus coeruleus (LoC) and specifically lowered the NA concentration in the hypothalamus of male quail. Other monoamines, such as dopamine and serotonin, were not affected by drug treatment. DSP-4 did not decrease the numbers of noradrenergic cells of the lateral tegmental cell group, nor the plasma NA concentration. Decreased hypothalamic NA levels after DSP-4 treatment did not change GnIH gene expression in the brains of quail during their interaction with conspecifics. On the other hand, GnIH gene expression increased in the brains of quail socially isolated for 1 hour after DSP-4 treatment. These results suggest that some noradrenergic neurones have inhibitory effects on GnIH gene expression of the hypothalamus in solitary quail.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Proteínas Aviares/genética , Coturnix , Hormonas Hipotalámicas/genética , Hipotálamo/metabolismo , Animales , Bencilaminas/administración & dosificación , Glucemia/metabolismo , Diencéfalo/efectos de los fármacos , Diencéfalo/metabolismo , Expresión Génica , Hipotálamo/efectos de los fármacos , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Masculino , Norepinefrina/sangre , ARN Mensajero/metabolismo , Aislamiento Social
17.
Brain Struct Funct ; 222(8): 3491-3508, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28382577

RESUMEN

The noradrenergic locus coeruleus (LC) plays an important role in the promotion and maintenance of arousal and alertness. Our group recently described coerulean projections to cochlear root neurons (CRNs), the first relay of the primary acoustic startle reflex (ASR) circuit. However, the role of the LC in the ASR and its modulation, prepulse inhibition (PPI), is not clear. In this study, we damaged LC neurons and fibers using a highly selective neurotoxin, DSP-4, and then assessed ASR and PPI in male and female rats. Our results showed that ASR amplitude was higher in males at 14 days after DSP-4 injection when compared to pre-administration values and those in the male control group. Such modifications in ASR amplitude did not occur in DSP-4-injected females, which exhibited ASR amplitude within the range of control values. PPI differences between males and females seen in controls were not observed in DSP-4-injected rats for any interstimulus interval tested. DSP-4 injection did not affect ASR and PPI latencies in either the male or the female groups, showing values that were consistent with the sex-related variability observed in control rats. Furthermore, we studied the noradrenergic receptor system in the cochlear nerve root using gene expression analysis. When compared to controls, DSP-4-injected males showed higher levels of expression in all adrenoceptor subtypes; however, DSP-4-injected females showed varied effects depending on the receptor type, with either up-, downregulations, or maintenance of expression levels. Lastly, we determined noradrenaline levels in CRNs and other LC-targeted areas using HPLC assays, and these results correlated with behavioral and adrenoceptor expression changes post DSP-4 injection. Our study supports the participation of LC in ASR and PPI, and contributes toward a better understanding of sex-related differences observed in somatosensory gating paradigms.


Asunto(s)
Núcleo Coclear/fisiología , Locus Coeruleus/fisiología , Neuronas/fisiología , Inhibición Prepulso/fisiología , Reflejo de Sobresalto , Caracteres Sexuales , Estimulación Acústica , Animales , Núcleo Coclear/citología , Núcleo Coclear/metabolismo , Dopamina beta-Hidroxilasa/metabolismo , Femenino , Locus Coeruleus/citología , Locus Coeruleus/metabolismo , Masculino , Vías Nerviosas/fisiología , Neuronas/citología , Neuronas/metabolismo , Norepinefrina/metabolismo , Ratas Wistar , Receptores Adrenérgicos/metabolismo
18.
ASN Neuro ; 9(2): 1759091417696161, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28266222

RESUMEN

Damage occurring to noradrenergic neurons in the locus coeruleus (LC) contributes to the evolution of neuroinflammation and neurodegeneration in a variety of conditions and diseases. One cause of LC damage may be loss of neurotrophic support from LC target regions. We tested this hypothesis by conditional unilateral knockout of brain-derived neurotrophic factor (BDNF) in adult mice. To evaluate the consequences of BDNF loss in the context of neurodegeneration, the mice harbored familial mutations for human amyloid precursor protein and presenilin-1. In these mice, BDNF depletion reduced tyrosine hydroxylase staining, a marker of noradrenergic neurons, in the rostral LC. BDNF depletion also reduced noradrenergic innervation in the hippocampus, the frontal cortex, and molecular layer of the cerebellum, assessed by staining for dopamine beta hydroxylase. BDNF depletion led to an increase in cortical amyloid plaque numbers and size but was without effect on plaque numbers in the striatum, a site with minimal innervation from the LC. Interestingly, cortical Iba1 staining for microglia was reduced by BDNF depletion and was correlated with reduced dopamine beta hydroxylase staining. These data demonstrate that reduction of BDNF levels in an LC target region can cause retrograde damage to LC neurons, leading to exacerbation of neuropathology in distinct LC target areas. Methods to reduce BDNF loss or supplement BDNF levels may be of value to reduce neurodegenerative processes normally limited by LC noradrenergic activities.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Hipocampo/metabolismo , Hipocampo/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Humanos , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Placa Amiloide/metabolismo , Placa Amiloide/patología , Presenilina-1/genética , Presenilina-1/metabolismo
19.
Int J Eat Disord ; 50(2): 152-156, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27611116

RESUMEN

OBJECTIVE: Emotional eating (EE) has been linked to norepinephrine dysfunction. Therefore, we aimed to investigate the relationship between EE and norepinephrine transporter (NET) availability. METHOD: Ten severely obese individuals (body mass index (BMI) 42.4 ± 3.7 kg/m2 ) and ten non-obese, healthy controls (BMI 23.9 ± 2.5 kg/m2 ) matched for age and sex were studied using (S,S)-[11 C]-O-methylreboxetine ([11 C]MRB) positron emission tomography (PET). Kinetic modeling of regional tissue time activity curves was performed using multilinear reference tissue model 2 (MRTM2, with the occipital cortex as a reference region) to estimate binding potential based on individual PET-MR coregistration. To test for associations of EE and NET availability, participants completed the EE subscale of the Dutch Eating Behavior Questionnaire before scanning. RESULTS: Obese individuals and non-obese, healthy controls did not significantly differ regarding EE scores and regional NET availability. For obese individuals only, correlative data analyses pointed to a sinoidal distribution pattern as a higher degree of EE related to lower NET availability in the locus coeruleus and to higher NET availability in the left thalamus. DISCUSSION: These results indicate that central in vivo NET availability is altered in EE of individuals with obesity. © 2016 Wiley Periodicals, Inc.(Int J Eat Disord 2017; 50:152-156).


Asunto(s)
Emociones , Trastornos de Alimentación y de la Ingestión de Alimentos/psicología , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Obesidad Mórbida/metabolismo , Adulto , Trastornos de Alimentación y de la Ingestión de Alimentos/metabolismo , Femenino , Humanos , Locus Coeruleus/metabolismo , Masculino , Morfolinas , Proyectos Piloto , Tomografía de Emisión de Positrones , Cintigrafía , Radiofármacos , Reboxetina , Tálamo/metabolismo
20.
J Nat Med ; 70(4): 749-59, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27417451

RESUMEN

Exposure to severe stress can lead to the development of neuropsychiatric disorders such as depression and post-traumatic stress disorder (PTSD) in at-risk individuals. Gastrodin (GAS), a primary constituent of an Oriental herbal medicine, has been shown to effectively treat various mood disorders. Thus, the present study aimed to determine whether GAS would ameliorate stress-associated depression-like behaviors in a rat model of single prolonged stress (SPS)-induced PTSD. Following the SPS procedure, rats received intraperitoneal administration of GAS (20, 50, or 100 mg/kg) once daily for 2 weeks. Subsequently, the rats performed the forced swimming test, and norepinephrine (NE) levels in the hippocampus were measured. Daily GAS (100 mg/kg) significantly reversed depression-like behaviors and restored SPS-induced increases in hippocampal NE concentrations as well as tyrosine hydroxylase expression in the locus coeruleus. Furthermore, the administration of GAS attenuated SPS-induced decreases in the hypothalamic expression of neuropeptide Y and the hippocampal mRNA expression of brain-derived neurotrophic factor. These findings indicate that GAS possesses antidepressant effects in the PTSD and may be an effective herbal preparation for the treatment of PTSD.


Asunto(s)
Antidepresivos/uso terapéutico , Alcoholes Bencílicos/uso terapéutico , Depresión/tratamiento farmacológico , Gastrodia/química , Glucósidos/uso terapéutico , Fitoterapia , Extractos Vegetales/uso terapéutico , Trastornos por Estrés Postraumático/tratamiento farmacológico , Animales , Antidepresivos/farmacología , Alcoholes Bencílicos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Depresión/metabolismo , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Glucósidos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Masculino , Neuropéptido Y/metabolismo , Norepinefrina/metabolismo , Extractos Vegetales/farmacología , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/metabolismo , Natación/psicología , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA