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










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38612743

RESUMEN

Only one-third of patients with advanced MSS/pMMR endometrial cancer exhibit a lasting response to the combination treatment of Pembrolizumab and Lenvatinib. The combined administration of these two drugs is based on Lenvatinib's ability to modulate the tumor microenvironment, enabling Pembrolizumab to exert its effect. These findings underscore the importance of exploring tumor microenvironment parameters to identify markers that can accurately select candidates for this type of therapy. An open non-randomized observational association study was conducted at six clinical centers, involving a total of 28 patients with advanced MSS/pMMR endometrial cancer who received Pembrolizumab and Lenvatinib therapy. Using TSA-associated multiplex immunofluorescence, we analyzed the proportion of CD8+ T lymphocytes, CD20+ B lymphocytes, FoxP3+ T regulatory lymphocytes, and CD163+ macrophages in tumor samples prior to immunotargeted therapy. The percentage of CD20+ B lymphocytes and the CD8-to-CD20 lymphocytes ratio was significantly higher in patients who responded to treatment compared to non-responders (responders vs. non-responders: 0.24 (0.1-1.24)% vs. 0.08 (0.00-0.15)%, p = 0.0114; 1.44 (0.58-2.70) arb. unit vs. 19.00 (3.80-34.78) arb. unit, p = 0.0031). The sensitivity and specificity of these biomarkers were 85.71% and 70.59%, and 85.71% and 85.71%, respectively. The proportion of CD20+ B lymphocytes and the CD8-to-CD20 lymphocytes ratio in the stroma of endometrial cancer serves as both a prognostic marker of response to immunotargeted therapy and a prognostic factor for progression-free survival in patients.


Asunto(s)
Antagonistas de Receptores de Angiotensina , Neoplasias Endometriales , Quinolinas , Femenino , Humanos , Inhibidores de la Enzima Convertidora de Angiotensina , Neoplasias Endometriales/tratamiento farmacológico , Compuestos de Fenilurea , Microambiente Tumoral
2.
Int J Mol Sci ; 24(2)2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36674951

RESUMEN

The problem of finding more precise stratification criteria for identifying the cohort of patients who would obtain the maximum benefit from immunotherapy is acute in modern times. In our study were enrolled 18 triple-negative breast cancer patients. The Ventana SP142 test was used for PD-L1 detection. Spatial transcriptomic analysis by 10x Genomics was used to compare PD-L1-positive and PD-L1-negative tumors. The seven-color multiplex immunofluorescence (by Akoya) was used for the detection of the type of cells that carried the PD1 receptor and the PD-L1 ligand. Using pathway analysis, we showed that PD-L1-positive tumors demonstrate signatures of a cell response to cytokines, among others, and PD-L1-negative tumors demonstrate signatures of antigen presentation. PD-L1-positive and PD-L1-negative tumors have different tumor microenvironment (TME) compositions according to CIBERSORT analysis. Multiplex immunohistochemistry (IHC) confirmed the prevalence of PD1-negative M2 macrophages and PD1-negative T lymphocytes in PD-L1-positive tumors. PD-L1-positive tumors are not characterized by direct contact between cells carrying the PD1 receptor and the PD-L1 ligand. So, the absence of specific immune reactions against the tumor, predominance of pro-tumor microenvironment, and rare contact between PDL1 and PD1-positive cells may be the potential reasons for the lack of an immune checkpoint inhibitor (ICI) effect in triple-negative breast cancer patients.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Antígeno B7-H1 , Microambiente Tumoral/genética , Receptor de Muerte Celular Programada 1/metabolismo , Ligandos , Biomarcadores de Tumor/metabolismo
3.
Front Oncol ; 12: 909505, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35814376

RESUMEN

Triple-negative breast cancer has no specific treatment and unfavorable prognosis. Eribulin is one of the drugs widely used in this cohort of patients. In addition to its antimitotic effect, eribulin has an immunomodulant effect on the tumor microenvironment. In this study, we discover immunological markers, such as tumor-infiltrating lymphocytes, CD8+, CD4+, FoxP3+, CD20+ lymphocytes, and their PD1 positivity or negativity, with the ability to predict benefits from eribulin within locally advanced or metastatic triple-negative breast cancer. The primary objective was to explore the association of composition of immune cells in the microenvironment with response to eribulin. The key secondary objective was overall survival. Seven-color multiplex immunofluorescence was used to phenotype lymphocytes in the primary tumor. It has been shown that the PD1-negative-to-PD1-positive B cells ratio in primary tumors more than 3 is an independent predictor of the short-term effectiveness of eribulin [OR (95%CI) 14.09 (1.29-153.35), p=0.0029] and worse overall survival [HR (95%CI) 11.25 (1.37-70.25), p=0.0009] in patients with locally advanced or metastatic triple-negative breast cancer.

4.
Sci Rep ; 8(1): 10730, 2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30013200

RESUMEN

The functions of purinergic P2 receptors (P2Rs) for extracellular adenosine triphosphate (ATP) are poorly understood. Here, for the first time, we show that activation of P2Rs in an important arousal region, the basal forebrain (BF), promotes wakefulness, whereas inhibition of P2Rs promotes sleep. Infusion of a non-hydrolysable P2R agonist, ATP-γ-S, into mouse BF increased wakefulness following sleep deprivation. ATP-γ-S depolarized BF cholinergic and cortically-projecting GABAergic neurons in vitro, an effect blocked by antagonists of ionotropic P2Rs (P2XRs) or glutamate receptors. In vivo, ATP-γ-S infusion increased BF glutamate release. Thus, activation of BF P2XRs promotes glutamate release and excitation of wake-active neurons. Conversely, pharmacological antagonism of BF P2XRs decreased spontaneous wakefulness during the dark (active) period. Together with previous findings, our results suggest sleep-wake regulation by BF extracellular ATP involves a balance between excitatory, wakefulness-promoting effects mediated by direct activation of P2XRs and inhibitory, sleep-promoting effects mediated by degradation to adenosine.


Asunto(s)
Prosencéfalo Basal/fisiología , Receptores Purinérgicos P2/metabolismo , Vigilia/fisiología , Adenosina/metabolismo , Adenosina Trifosfato/administración & dosificación , Adenosina Trifosfato/análogos & derivados , Animales , Prosencéfalo Basal/citología , Prosencéfalo Basal/efectos de los fármacos , Electrodos Implantados , Electroencefalografía/instrumentación , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Ácido Glutámico/metabolismo , Masculino , Ratones , Modelos Animales , Técnicas de Placa-Clamp , Agonistas del Receptor Purinérgico P2/administración & dosificación , Antagonistas del Receptor Purinérgico P2/administración & dosificación , Receptores de Glutamato/metabolismo , Receptores Purinérgicos P2/efectos de los fármacos , Sueño/efectos de los fármacos , Sueño/fisiología , Vigilia/efectos de los fármacos
5.
J Neurophysiol ; 117(1): 327-335, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27784808

RESUMEN

Sleep homeostasis in rats undergoes significant maturational changes during postweaning development, but the underlying mechanisms of this process are unknown. In the present study we tested the hypothesis that the maturation of sleep is related to the functional emergence of adenosine (AD) signaling in the brain. We assessed postweaning changes in 1) wake-related elevation of extracellular AD in the basal forebrain (BF) and adjacent lateral preoptic area (LPO), and 2) the responsiveness of median preoptic nucleus (MnPO) sleep-active cells to increasing homeostatic sleep drive. We tested the ability of exogenous AD to augment homeostatic responses to sleep deprivation (SD) in newly weaned rats. In groups of postnatal day (P)22 and P30 rats, we collected dialysate from the BF/LPO during baseline (BSL) wake-sleep, SD, and recovery sleep (RS). HPLC analysis of microdialysis samples revealed that SD in P30 rats results in significant increases in AD levels compared with BSL. P22 rats do not exhibit changes in AD levels in response to SD. We recorded neuronal activity in the MnPO during BSL, SD, and RS at P22/P30. MnPO neurons exhibited adult-like increases in waking neuronal discharge across SD on both P22 and P30, but discharge rates during enforced wake were higher on P30 vs. P22. Central administration of AD (1 nmol) during SD on P22 resulted in increased sleep time and EEG slow-wave activity during RS compared with saline control. Collectively, these findings support the hypothesis that functional reorganization of an adenosinergic mechanism of sleep regulation contributes to the maturation of sleep homeostasis. NEW & NOTEWORTHY: Brain mechanisms that regulate the maturation of sleep are understudied. The present study generated first evidence about a potential mechanistic role for adenosine in the maturation of sleep homeostasis. Specifically, we demonstrate that early postweaning development in rats, when homeostatic response to sleep loss become adult like, is characterized by maturational changes in wake-related production/release of adenosine in the brain. Pharmacologically increased adenosine signaling in developing brain facilitates homeostatic responses to sleep deprivation.


Asunto(s)
Adenosina/metabolismo , Homeostasis/fisiología , Área Preóptica/crecimiento & desarrollo , Área Preóptica/metabolismo , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Sueño/fisiología , Adenosina/farmacología , Factores de Edad , Envejecimiento/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Cromatografía Líquida de Alta Presión , Electroencefalografía , Electromiografía , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/fisiología , Homeostasis/efectos de los fármacos , Área Preóptica/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sueño/efectos de los fármacos , Privación de Sueño/fisiopatología , Vigilia
6.
J Neurosci ; 36(6): 2057-67, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26865627

RESUMEN

Understanding the control of sleep-wake states by the basal forebrain (BF) poses a challenge due to the intermingled presence of cholinergic, GABAergic, and glutamatergic neurons. All three BF neuronal subtypes project to the cortex and are implicated in cortical arousal and sleep-wake control. Thus, nonspecific stimulation or inhibition studies do not reveal the roles of these different neuronal types. Recent studies using optogenetics have shown that "selective" stimulation of BF cholinergic neurons increases transitions between NREM sleep and wakefulness, implicating cholinergic projections to cortex in wake promotion. However, the interpretation of these optogenetic experiments is complicated by interactions that may occur within the BF. For instance, a recent in vitro study from our group found that cholinergic neurons strongly excite neighboring GABAergic neurons, including the subset of cortically projecting neurons, which contain the calcium-binding protein, parvalbumin (PV) (Yang et al., 2014). Thus, the wake-promoting effect of "selective" optogenetic stimulation of BF cholinergic neurons could be mediated by local excitation of GABA/PV or other non-cholinergic BF neurons. In this study, using a newly designed opto-dialysis probe to couple selective optical stimulation with simultaneous in vivo microdialysis, we demonstrated that optical stimulation of cholinergic neurons locally increased acetylcholine levels and increased wakefulness in mice. Surprisingly, the enhanced wakefulness caused by cholinergic stimulation was abolished by simultaneous reverse microdialysis of cholinergic receptor antagonists into BF. Thus, our data suggest that the wake-promoting effect of cholinergic stimulation requires local release of acetylcholine in the basal forebrain and activation of cortically projecting, non-cholinergic neurons, including the GABAergic/PV neurons. SIGNIFICANCE STATEMENT: Optogenetics is a revolutionary tool to assess the roles of particular groups of neurons in behavioral functions, such as control of sleep and wakefulness. However, the interpretation of optogenetic experiments requires knowledge of the effects of stimulation on local neurotransmitter levels and effects on neighboring neurons. Here, using a novel "opto-dialysis" probe to couple optogenetics and in vivo microdialysis, we report that optical stimulation of basal forebrain (BF) cholinergic neurons in mice increases local acetylcholine levels and wakefulness. Reverse microdialysis of cholinergic antagonists within BF prevents the wake-promoting effect. This important result challenges the prevailing dictum that BF cholinergic projections to cortex directly control wakefulness and illustrates the utility of "opto-dialysis" for dissecting the complex brain circuitry underlying behavior.


Asunto(s)
Neuronas Colinérgicas/fisiología , Prosencéfalo/fisiología , Vigilia/fisiología , Acetilcolina/metabolismo , Animales , Antagonistas Colinérgicos/administración & dosificación , Antagonistas Colinérgicos/farmacología , Neuronas Colinérgicas/efectos de los fármacos , Electroencefalografía , Electromiografía , Femenino , Masculino , Ratones , Microdiálisis , Optogenética , Parvalbúminas/metabolismo , Estimulación Luminosa , Prosencéfalo/efectos de los fármacos , Fases del Sueño/fisiología , Vigilia/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología
7.
Eur J Neurosci ; 41(2): 182-95, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25369989

RESUMEN

The tight coordination of biochemical and electrophysiological mechanisms underlies the homeostatic sleep pressure (HSP) produced by sleep deprivation (SD). We have reported that during SD the levels of inducible nitric oxide synthase (iNOS), extracellular nitric oxide (NO), adenosine [AD]ex , lactate [Lac]ex and pyruvate [Pyr]ex increase in the basal forebrain (BF). However, it is not clear whether all of them contribute to HSP leading to increased electroencephalogram (EEG) delta activity during non-rapid eye movement (NREM) recovery sleep (RS) following SD. Previously, we showed that NREM delta increase evident during RS depends on the presence of BF cholinergic (ChBF) neurons. Here, we investigated the role of ChBF cells in coordination of biochemical and EEG changes seen during SD and RS in the rat. Increases in low-theta power (5-7 Hz), but not high-theta (7-9 Hz), during SD correlated with the increase in NREM delta power during RS, and with the changes in nitrate/nitrite [NOx ]ex and [AD]ex . Lesions of ChBF cells using IgG 192-saporin prevented increases in [NOx ]ex , [AD]ex and low-theta activity, during SD, but did not prevent increases in [Lac]ex and [Pyr]ex . Infusion of NO donor DETA NONOate into the saporin-treated BF failed to increase NREM RS and delta power, suggesting ChBF cells are important for mediating NO homeostatic effects. Finally, SD-induced iNOS was mostly expressed in ChBF cells, and the intensity of iNOS induction correlated with the increase in low-theta activity. Together, our data indicate ChBF cells are important in regulating the biochemical and EEG mechanisms that contribute to HSP.


Asunto(s)
Prosencéfalo Basal/fisiología , Neuronas Colinérgicas/fisiología , Homeostasis/fisiología , Sueño/fisiología , Adenosina/metabolismo , Animales , Anticuerpos Monoclonales , Prosencéfalo Basal/efectos de los fármacos , Prosencéfalo Basal/fisiopatología , Neuronas Colinérgicas/efectos de los fármacos , Ritmo Delta/efectos de los fármacos , Ritmo Delta/fisiología , Homeostasis/efectos de los fármacos , Ácido Láctico/metabolismo , Masculino , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Nitritos/metabolismo , Compuestos Nitrosos/farmacología , Ácido Pirúvico/metabolismo , Ratas Wistar , Proteínas Inactivadoras de Ribosomas Tipo 1 , Saporinas , Sueño/efectos de los fármacos , Privación de Sueño/fisiopatología , Ritmo Teta/efectos de los fármacos , Ritmo Teta/fisiología
9.
Sleep Med Rev ; 15(2): 123-35, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20970361

RESUMEN

Adenosine is directly linked to the energy metabolism of cells. In the central nervous system (CNS) an increase in neuronal activity enhances energy consumption as well as extracellular adenosine concentrations. In most brain areas high extracellular adenosine concentrations, through A1 adenosine receptors, decrease neuronal activity and thus the need for energy. Adenosine may be a final common pathway for various sleep factors. We have identified a relatively specific area, the basal forebrain (BF), which appears to be central in the regulation/execution of recovery sleep after sleep deprivation (SD), or prolonged wakefulness. Adenosine concentration increases in this area during SD, and this increase induces sleep while prevention of the increase during SD abolishes recovery sleep. The increase in adenosine is associated with local changes in energy metabolism as indicated by increases in levels of pyruvate and lactate and increased phosphorylation of AMP-activated protein kinase. The increases in adenosine and sleep are associated with intact cholinergic system since specific lesion of the BF cholinergic cells abolishes both. Whether adenosine during SD is produced by the cholinergic neurons or astrocytes associated with them remains to be explored. An interesting, but so far unexplored question regards the relationship between the local, cortical regulation of sleep homeostasis and the global regulation of the state of sleep as executed by lower brain mechanisms, including the BF. The increase in adenosine concentration during SD also in cortical areas suggests that adenosine may have a role in the local regulation of sleep homeostasis. The core of sleep need is probably related to primitive functions of life, like energy metabolism. It can be noted that this assumption in no way excludes the possibility that later in evolution additional functions may have developed, e.g., related to complex neuronal network functions like memory and learning.


Asunto(s)
Adenosina/metabolismo , Metabolismo Energético , Homeostasis , Sueño/fisiología , Animales , Ritmo Circadiano/fisiología , Humanos , Ratones , Prosencéfalo/metabolismo , Vigilia/fisiología
10.
J Neurochem ; 116(2): 260-72, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21062286

RESUMEN

Both adenosine and nitric oxide (NO) are known for their role in sleep homeostasis, with the basal forebrain (BF) wakefulness center as an important site of action. Previously, we reported a cascade of homeostatic events, wherein sleep deprivation (SD) induces the production of inducible nitric oxide synthase (iNOS)-dependent NO in BF, leading to enhanced release of extracellular adenosine. In turn, increased BF adenosine leads to enhanced sleep intensity, as measured by increased non-rapid eye movement sleep EEG delta activity. However, the presence and time course of similar events in cortex has not been studied, although a frontal cortical role for the increase in non-rapid eye movement recovery sleep EEG delta power is known. Accordingly, we performed simultaneous hourly microdialysis sample collection from BF and frontal cortex (FC) during 11 h SD. We observed that both areas showed sequential increases in iNOS and NO, followed by increases in adenosine. BF increases began at 1 h SD, whereas FC increases began at 5 h SD. iNOS and Fos-double labeling indicated that iNOS induction occurred in BF and FC wake-active neurons. These data support the role of BF adenosine and NO in sleep homeostasis and indicate the temporal and spatial sequence of sleep homeostatic cascade for NO and adenosine.


Asunto(s)
Adenosina/metabolismo , Corteza Cerebral/metabolismo , Homeostasis/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/fisiología , Prosencéfalo/metabolismo , Privación de Sueño/metabolismo , Fases del Sueño/fisiología , Adenosina/fisiología , Animales , Corteza Cerebral/fisiología , Electroencefalografía/métodos , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/fisiología , Prosencéfalo/fisiología , Ratas , Ratas Wistar , Factores de Tiempo
11.
J Neurosci ; 30(40): 13254-64, 2010 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-20926651

RESUMEN

Sleep loss negatively impacts performance, mood, memory, and immune function, but the homeostatic factors that impel sleep after sleep loss are imperfectly understood. Pharmacological studies had implicated the basal forebrain (BF) inducible nitric oxide (NO) synthase (iNOS)-dependent NO as a key homeostatic factor, but its cellular source was obscure. To obtain direct evidence about the cellular source of iNOS-generated NO during sleep deprivation (SD), we used intracerebroventricular perfusion in rats of the cell membrane-permeable dye diaminofluorescein-2/diacetate (DAF-2/DA) that, once intracellular, bound NO and fluoresced. To circumvent the effects of neuronal NOS (nNOS), DAF-2/DA was perfused in the presence of an nNOS inhibitor. SD led to DAF-positive fluorescence only in the BF neurons, not glia. SD increased expression of iNOS, which colocalized with NO in neurons and, more specifically, in prolonged wakefulness-active neurons labeled by Fos. SD-induced iNOS expression in wakefulness-active neurons positively correlated with sleep pressure, as measured by the number of attempts to enter sleep. Importantly, SD did not induce Fos or iNOS in stress-responsive central amygdala and paraventricular hypothalamic neurons, nor did SD elevate corticosterone, suggesting that the SD protocol did not provoke iNOS expression through stress. We conclude that iNOS-produced neuronal NO is an important homeostatic factor promoting recovery sleep after SD.


Asunto(s)
Núcleo Basal de Meynert/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/biosíntesis , Privación de Sueño/metabolismo , Vigilia/fisiología , Animales , Núcleo Basal de Meynert/enzimología , Inyecciones Intraventriculares/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/enzimología , Neuronas/fisiología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Ratas , Ratas Wistar , Sueño/fisiología , Privación de Sueño/enzimología , Privación de Sueño/fisiopatología
12.
J Neurosci ; 30(26): 9007-16, 2010 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-20592221

RESUMEN

Sleep is one of the most pervasive biological phenomena, but one whose function remains elusive. Although many theories of function, indirect evidence, and even common sense suggest sleep is needed for an increase in brain energy, brain energy levels have not been directly measured with modern technology. We here report that ATP levels, the energy currency of brain cells, show a surge in the initial hours of spontaneous sleep in wake-active but not in sleep-active brain regions of rat. The surge is dependent on sleep but not time of day, since preventing sleep by gentle handling of rats for 3 or 6 h also prevents the surge in ATP. A significant positive correlation was observed between the surge in ATP and EEG non-rapid eye movement delta activity (0.5-4.5 Hz) during spontaneous sleep. Inducing sleep and delta activity by adenosine infusion into basal forebrain during the normally active dark period also increases ATP. Together, these observations suggest that the surge in ATP occurs when the neuronal activity is reduced, as occurs during sleep. The levels of phosphorylated AMP-activated protein kinase (P-AMPK), well known for its role in cellular energy sensing and regulation, and ATP show reciprocal changes. P-AMPK levels are lower during the sleep-induced ATP surge than during wake or sleep deprivation. Together, these results suggest that sleep-induced surge in ATP and the decrease in P-AMPK levels set the stage for increased anabolic processes during sleep and provide insight into the molecular events leading to the restorative biosynthetic processes occurring during sleep.


Asunto(s)
Adenosina Trifosfato/metabolismo , Encéfalo/fisiología , Sueño/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina/farmacología , Animales , Encéfalo/efectos de los fármacos , Fármacos del Sistema Nervioso Central/farmacología , Creatina/metabolismo , Ritmo Delta , Electroencefalografía , Masculino , Fosfocreatina/metabolismo , Fosforilación , Fotoperiodo , Ratas , Ratas Sprague-Dawley , Privación de Sueño/metabolismo , Fases del Sueño/efectos de los fármacos , Fases del Sueño/fisiología , Factores de Tiempo , Vigilia/fisiología
13.
Neurobiol Aging ; 31(11): 2011-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19058880

RESUMEN

Nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS) in the cholinergic basal forebrain (BF) during sleep deprivation (SD) is implicated in adenosine (AD) release and induction of recovery sleep. Aging is associated with impairments in sleep homeostasis, such as decrease in non-rapid eye movement sleep (NREM) intensity following SD. We hypothesized that age related changes in sleep homeostasis may be induced by impairments in NO-mediated sleep induction. To test this hypothesis we measured levels of NO and iNOS in the BF during SD as well as recovery sleep after SD and NO-donor (DETA/NO) infusion into the BF in three age groups of rats (young, 4 months; middle-aged, 14 months; old, 24 months). We found that in aged rats as compared to young (1) recovery NREM sleep intensity was significantly decreased, (2) neither iNOS nor NO increased in the BF during SD, and (3) DETA/NO infusion failed to induce sleep. Together, these results support our hypothesis that aging impairs the mechanism through which NO in the BF induces sleep.


Asunto(s)
Envejecimiento/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Masculino , Microdiálisis , Prosencéfalo/metabolismo , Ratas , Ratas Wistar , Fases del Sueño/fisiología
14.
Neuroreport ; 20(11): 1013-8, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19491711

RESUMEN

Multiple studies indicate that adenosine released in the basal forebrain during prolonged wakefulness could affect recovery sleep. It is still unclear which of adenosine receptors provide its sleep-modulating effects in the basal forebrain. We infused adenosine A1 and A2A receptors antagonists into the rat basal forebrain during sleep deprivation and compared characteristics of recovery non-rapid eye movement (non-REM) sleep (its amount and non-REM sleep delta power) after sleep deprivation, and after sleep deprivation combined with perfusion of antagonists. A1 receptor antagonist significantly reduced recovery sleep amount and delta power, whereas A2A receptor antagonist had no effect on recovery sleep. We conclude that adenosine can promote recovery non-REM sleep when acting through A1 receptors in the basal forebrain.


Asunto(s)
Homeostasis/fisiología , Prosencéfalo/fisiología , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Sueño/fisiología , Antagonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A2 , Análisis de Varianza , Animales , Cateterismo , Electrodos Implantados , Electroencefalografía , Homeostasis/efectos de los fármacos , Masculino , Prosencéfalo/efectos de los fármacos , Ratas , Ratas Wistar , Sueño/efectos de los fármacos , Privación de Sueño/tratamiento farmacológico , Teobromina/análogos & derivados , Teobromina/farmacología , Teofilina/análogos & derivados , Teofilina/farmacología
15.
Psychopharmacology (Berl) ; 201(1): 147-60, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18661122

RESUMEN

RATIONALE: During prolonged wakefulness, the concentrations of nitric oxide (NO) and adenosine (AD) increase in the basal forebrain (BF). AD inhibits neuronal activity via adenosine (A1) receptors, thus providing a potential mechanism for sleep facilitation. Although NO in the BF increases adenosine and promotes sleep, it is not clear whether the sleep promotion by NO is mediated through adenosine increase, or NO independently of adenosine could modulate sleep. OBJECTIVE: The objective of the study was to clarify whether NO modulates the discharge rate of BF neurons and whether this effect is mediated via AD. MATERIALS AND METHODS: We measured the discharge rates of BF neurons in anesthetized rats during microdialysis infusion of NO donor alone or in combination with A1 receptor antagonist, 8-cyclopentyl-1,3-dimethylxanthine. RESULTS: NO dose dependently modulated the discharge rate of BF neurons. NO donor (0.5 mM) increased the discharge rates in 48% of neurons and decreased it in 22%. A 1-mM dose decreased it in 55% and increased in 18%. Tactile stimulus affected the discharge rates of most neurons: 60% increased (stimulus-on) it and 14% decreased it (stimulus-off). A 1-mM NO donor predominantly inhibited neurons of both stimulus related types. A small proportion of stimulus-on (23%) neurons but none of the stimulus-off neurons were activated by NO donor. The blockade of A1 receptors partly prevented the inhibitory effect of NO on most of the neurons. This response was more prominent in stimulus-on than in stimulus-off neurons. CONCLUSION: NO modulates the BF neuronal discharge rates in a dose-dependent manner. The inhibitory effect is partly mediated via adenosine A1 receptors.


Asunto(s)
Neuronas/efectos de los fármacos , Neuronas/fisiología , Óxido Nítrico/metabolismo , Prosencéfalo/citología , Prosencéfalo/efectos de los fármacos , Adenosina/metabolismo , Antagonistas del Receptor de Adenosina A1 , Animales , Banda Diagonal de Broca/efectos de los fármacos , Banda Diagonal de Broca/fisiología , Relación Dosis-Respuesta a Droga , Electroencefalografía , Electrofisiología/instrumentación , Electrofisiología/métodos , Espacio Extracelular/efectos de los fármacos , Masculino , Microdiálisis , Neuronas/citología , Donantes de Óxido Nítrico/farmacología , Compuestos Nitrosos/farmacología , Estimulación Física , Área Preóptica/efectos de los fármacos , Área Preóptica/fisiología , Prosencéfalo/fisiología , Ratas , Ratas Wistar , Sustancia Innominada/efectos de los fármacos , Sustancia Innominada/fisiología , Teofilina/análogos & derivados , Teofilina/farmacología , Percepción del Tacto , Uretano/farmacología
17.
Neurobiol Aging ; 25(2): 231-8, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14749141

RESUMEN

Orexin A and B (hypocretin 1 and 2) are hypothalamic peptides, which are synthesized in the lateral hypothalamus. Orexins participate in the regulation energy balance, food intake, vigilance and several endocrine and autonomic functions. The widespread projections of the orexin neurons suggest that they may have a role in coordination of different brain activities. The effects of ageing on the orexin system have not been studied previously. Prepro-orexin gene expression in the lateral hypothalamus, and the contents of orexin A and B peptides in the lateral hypothalamus and hypothalamus were measured in young, middle-aged and old (3, 12 and 24 months) rats. In the course of ageing, the expression of the prepro-orexin gene and the levels of orexin A and B decreased; the main decrease occurred by 12 months. Sleep deprivation for 6h increased slightly the expression of prepro-orexin gene in young rats. Deterioration of the orexin system may play a role in the phenomenon associated with aging, e.g. decreased consolidation of vigilance states, endocrine changes and dysfunctions of autonomic nervous system.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Neuropéptidos/metabolismo , Precursores de Proteínas/metabolismo , Animales , Encéfalo/anatomía & histología , Recuento de Células/métodos , Hibridación in Situ/métodos , Masculino , Neuronas/metabolismo , Neuropéptidos/genética , Orexinas , Precursores de Proteínas/genética , Radioinmunoensayo/métodos , Ratas , Ratas Wistar , Privación de Sueño/metabolismo , Vigilia/fisiología
18.
ScientificWorldJournal ; 3: 790-8, 2003 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12941976

RESUMEN

While the exact function of sleep remains unknown, it is evident that sleep was developed early in phylogenesis and represents an ancient and vital strategy for survival. Several pieces of evidence suggest that the function of sleep is associated with energy metabolism, saving of energy, and replenishment of energy stores. Prolonged wakefulness induces signs of energy depletion in the brain, while experimentally induced, local energy depletion induces increase in sleep, similarly as would a period of prolonged wakefulness. The key molecule in the induction of sleep appears to be adenosine, which induces sleep locally in the basal forebrain.


Asunto(s)
Adenosina/fisiología , Metabolismo Energético/fisiología , Sueño/fisiología , Animales , Humanos
19.
Eur J Neurosci ; 17(4): 863-9, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12603276

RESUMEN

Sleep saves energy, but can brain energy depletion induce sleep? We used 2,4-dinitrophenol (DNP), a molecule which prevents the synthesis of ATP, to induce local energy depletion in the basal forebrain of rats. Three-hour DNP infusions induced elevations in extracellular concentrations of lactate, pyruvate and adenosine, as well as increases in non-REM sleep during the following night. Sleep was not affected when DNP was administered to adjacent brain areas, although the metabolic changes were similar. The amount and the timing of the increase in non-REM sleep, as well as in the concentrations of lactate, pyruvate and adenosine with 0.5-1.0 mM DNP infusion, were comparable to those induced by 3 h of sleep deprivation. Here we show that energy depletion in localized brain areas can generate sleep. The energy depletion model of sleep induction could be applied to in vitro research into the cellular mechanisms of prolonged wakefulness.


Asunto(s)
Metabolismo Energético/fisiología , Prosencéfalo/metabolismo , Sueño/fisiología , 2,4-Dinitrofenol/farmacología , Adenosina/metabolismo , Análisis de Varianza , Animales , Cromatografía Líquida de Alta Presión/métodos , Ritmo Circadiano/fisiología , Relación Dosis-Respuesta a Droga , Electroencefalografía/métodos , Metabolismo Energético/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ácido Láctico/metabolismo , Masculino , Microdiálisis/métodos , Cianuro de Potasio/farmacología , Prosencéfalo/efectos de los fármacos , Ácido Pirúvico/metabolismo , Ratas , Sueño/efectos de los fármacos , Privación de Sueño/metabolismo , Fases del Sueño/efectos de los fármacos , Fases del Sueño/fisiología , Desacopladores/farmacología
20.
Sleep Med Rev ; 6(4): 321-32, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12531135

RESUMEN

Adenosine is directly linked to the energy metabolism of cells. In the central nervous system an increase in neuronal activity enhances energy consumption as well as extracellular adenosine concentrations. In most brain areas high extracellular adenosine concentrations, through A(1) adenosine receptors, decrease neuronal activity and thus the need for energy. Adenosine seems to act as a direct negative feed-back inhibitor of neuronal activity. Hypoxia and ischemia induce very high extracellular adenosine levels, which may limit further brain damage. In brain areas that regulate cortical vigilance, particularly in the basal forebrain, high extracellular adenosine concentrations, induced by prolonged wakefulness, decrease the activity of presumably cholinergic cells and via this mechanism promote sleep. Our hypothesis is that in the cholinergic basal forebrain prolonged wakefulness induces local energy depletion that generates increases in extracellular adenosine concentrations in this area. In addition to the immediate effects, high extracellular adenosine concentrations also induce intracellular changes in signal transduction and transcription, e.g. increase in A(1) receptor expression and NF-kappaB binding activity. These changes may at least partially mediate the long term effects of prolonged wakefulness. Adenosine may also be a common mediator of the effects of several other sleep-inducing factors.


Asunto(s)
Adenosina/metabolismo , Sueño/fisiología , Encéfalo/metabolismo , Metabolismo Energético/fisiología , Humanos , Privación de Sueño/metabolismo , Vigilia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...