Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Synapse ; 73(9): e22101, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30964565

RESUMEN

Chronic stress induces maladaptive neural responses in several brain areas including hippocampus. It has been demonstrated that chronic stress exposure induced a downregulation of the putative presynaptic type 2 metabotropic glutamate (mGlu2) receptors, which would reduce the negative feedback role exerted by these receptors. The reduced availability of these receptors would enhance glutamate overflow in the hippocampus, supporting the hypothesis that hippocampal glutamatergic neurotransmission plays a key etiopathological determinant in stress-induced neuropsychiatric disorders. Since modulation of glutamatergic neurotransmission has been shown to represent an interesting pharmacological tool to treat psychiatric disorders, in the present study we have investigated the effects of the mGlu2 receptor positive allosteric modulator (PAM) LY487379. The rational bases of our study were: (a) chronic restraint stress (CRS) application in C57/BALB6 mouse induced a loss of resilience at the behavioral, biochemical, and electrophysiological level; (b) a superimposed familiar stressor (restraint) but not unfamiliar (i.e., forced swim stress) completely reversed the effects of CRS. Using the CRS model, in the present study we have investigated the effects of LY487379, an mGlu2 PAM, as well as a superimposed familiar stressor (acute restraint stress-ARS), on the immobility time at the tail suspension test and electrophysiological profile of glutamatergic transmission in the dentate gyrus (DG).


Asunto(s)
Giro Dentado/efectos de los fármacos , Distrés Psicológico , Piridinas/farmacología , Receptores de Glutamato Metabotrópico/agonistas , Sulfonamidas/farmacología , Transmisión Sináptica/efectos de los fármacos , Animales , Giro Dentado/metabolismo , Masculino , Trastornos Mentales/etiología , Trastornos Mentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Restricción Física/efectos adversos , Transmisión Sináptica/fisiología
2.
Pharmacol Res ; 132: 130-134, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29689315

RESUMEN

Prolonged stress predisposes susceptible individuals to a number of physiological disorders including cardiovascular disease, obesity and gastrointestinal disorders, as well as psychiatric and neurodegenerative disorders. Preclinical studies have suggested that manipulation of the glucocorticoid milieu can trigger cellular, molecular and behavioral derangement resembling the hallmarks of Alzheimer's Disease (AD). For example, stress or glucocorticoid administration can increase amyloid ß precursor protein and tau phosphorylation which are involved in synaptic dysfunction and neuronal death associated with AD. Although since AD was first described in 1906 at a conference in Tubingen, Germany by Alois Alzheimer our knowledge of neuropathological and neurochemical alterations of AD has been impressively increased, at present, pharmacotherapy is symptomatic at best and has no influence on the progression of the disorder. It is generally believed that most of the drugs developed as disease modifiers have failed in clinical trials because treatment started too late, i.e., after the clinical onset of AD. Because AD pathology begins several years prior to the clinical diagnosis, it is imperative to identify subjects at high risk to develop the disorder. Consequently, the search for putative risk factors has gained importance. ApoE4, diabetes/metabolic syndrome, cardiovascular disorders, and a low cognitive reserve are established risk factors for AD. The focus of this review is on stress and glucocorticoids as potential factors increasing the risk to develop AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Estrés Psicológico/complicaciones , Animales , Glucocorticoides , Humanos , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Factores de Riesgo
3.
Proc Natl Acad Sci U S A ; 112(48): 14960-5, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26627246

RESUMEN

Excitatory amino acids play a key role in both adaptive and deleterious effects of stressors on the brain, and dysregulated glutamate homeostasis has been associated with psychiatric and neurological disorders. Here, we elucidate mechanisms of epigenetic plasticity in the hippocampus in the interactions between a history of chronic stress and familiar and novel acute stressors that alter expression of anxiety- and depressive-like behaviors. We demonstrate that acute restraint and acute forced swim stressors induce differential effects on these behaviors in naive mice and in mice with a history of chronic-restraint stress (CRS). They reveal a key role for epigenetic up- and down-regulation of the putative presynaptic type 2 metabotropic glutamate (mGlu2) receptors and the postsynaptic NR1/NMDA receptors in the hippocampus and particularly in the dentate gyrus (DG), a region of active neurogenesis and a target of antidepressant treatment. We show changes in DG long-term potentiation (LTP) that parallel behavioral responses, with habituation to the same acute restraint stressor and sensitization to a novel forced-swim stressor. In WT mice after CRS and in unstressed mice with a BDNF loss-of-function allele (BDNF Val66Met), we show that the epigenetic activator of histone acetylation, P300, plays a pivotal role in the dynamic up- and down-regulation of mGlu2 in hippocampus via histone-3-lysine-27-acetylation (H3K27Ac) when acute stressors are applied. These hippocampal responses reveal a window of epigenetic plasticity that may be useful for treatment of disorders in which glutamatergic transmission is dysregulated.


Asunto(s)
Conducta Animal , Giro Dentado/metabolismo , Epigénesis Genética , Ácido Glutámico/metabolismo , Estrés Psicológico/metabolismo , Transmisión Sináptica , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ansiedad/patología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Giro Dentado/patología , Depresión/genética , Depresión/metabolismo , Depresión/patología , Ácido Glutámico/genética , Histonas/genética , Histonas/metabolismo , Masculino , Ratones , Ratones Mutantes , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/patología , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
4.
Proc Natl Acad Sci U S A ; 110(12): 4804-9, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382250

RESUMEN

Epigenetic mechanisms are involved in the pathophysiology of depressive disorders and are unique potential targets for therapeutic intervention. The acetylating agent L-acetylcarnitine (LAC), a well-tolerated drug, behaves as an antidepressant by the epigenetic regulation of type 2 metabotropic glutamate (mGlu2) receptors. It caused a rapid and long-lasting antidepressant effect in Flinders Sensitive Line rats and in mice exposed to chronic unpredictable stress, which, respectively, model genetic and environmentally induced depression. In both models, LAC increased levels of acetylated H3K27 bound to the Grm2 promoter and also increased acetylation of NF-ĸB-p65 subunit, thereby enhancing the transcription of Grm2 gene encoding for the mGlu2 receptor in hippocampus and prefrontal cortex. Importantly, LAC reduced the immobility time in the forced swim test and increased sucrose preference as early as 3 d of treatment, whereas 14 d of treatment were needed for the antidepressant effect of chlorimipramine. Moreover, there was no tolerance to the action of LAC, and the antidepressant effect was still seen 2 wk after drug withdrawal. Conversely, NF-ĸB inhibition prevented the increase in mGlu2 expression induced by LAC, whereas the use of a histone deacetylase inhibitor supported the epigenetic control of mGlu2 expression. Finally, LAC had no effect on mGlu2 knockout mice exposed to chronic unpredictable stress, and a single injection of the mGlu2/3 receptor antagonist LY341495 partially blocked LAC action. The rapid and long-lasting antidepressant action of LAC strongly suggests a unique approach to examine the epigenetic hypothesis of depressive disorders in humans, paving the way for more efficient antidepressants with faster onset of action.


Asunto(s)
Acetilcarnitina/farmacología , Antidepresivos/farmacología , Epigénesis Genética/efectos de los fármacos , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Corteza Prefrontal/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Acetilación/efectos de los fármacos , Aminoácidos , Animales , Clomipramina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hipocampo/patología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/genética , Nootrópicos/farmacología , Corteza Prefrontal/patología , Ratas , Receptores de Glutamato Metabotrópico/genética , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Factores de Tiempo , Xantenos
5.
Hell J Nucl Med ; 18(3): 233-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26574693

RESUMEN

OBJECTIVE: Human beta-defensin-3 (HBD-3) is an antimicrobial peptide which is up-regulated during inflammation. Based on the previously demonstrated capacity of technetium-99m ((99m)Tc) labelled HBD-3 of distinguishing infection from inflammation in rats, we have decided to collect information on the potential toxicity of the tracer in view of its possible use for imaging in humans. MATERIALS AND METHODS: Recombinant HBD-3 underwent labeling with (99m)Tc. The CD1 mice were selected as standard rodent species. Ten mice, 5 male and 5 female, were subjected to physical examination and housed in a dedicated room in 5 per cage. After 9 days pre-test period, all mice were weighted for dose adjustment and received intravenously 6mcg/mouse of (99m)Tc-HBD-3. Mortality was recorded daily, while body weight was registered once a week. Clinical observation of animals was performed daily for sickness symptoms due to the drug treatment. At day 19 a second dose of 6mcg/mouse (99m)Tc-HBD-3, was administered. Twenty-four hours after the second dose (day 20) the animals were euthanized. A piece of liver, kidneys, heart and lungs was collected for histopathological analysis. RESULTS: Our results showed that the labelled-HBD-3 dose did not induce significant toxicity in mice. Of course these parameters were not sufficient to authorize use in humans. This non-toxic dose of HBD-3 when translated from animals to humans resulted in an equivalent dose of approximately 25 times higher than that needed for imaging. CONCLUSION: Our non toxicity data of using (99m)Tc-beta-defensin-3 in mice offer a further indication in favour of the clinical use of this radiopharmaceutical in all cases where discrimination between infection and inflammation is needed.


Asunto(s)
Radiofármacos/toxicidad , Tecnecio/administración & dosificación , Tecnecio/toxicidad , beta-Defensinas/administración & dosificación , beta-Defensinas/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Femenino , Marcaje Isotópico , Dosificación Letal Mediana , Ratones , Radiofármacos/administración & dosificación , Tasa de Supervivencia , Tecnecio/química , beta-Defensinas/química
6.
Aging Clin Exp Res ; 26(4): 461-4, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24435333

RESUMEN

BACKGROUND AND AIMS: Increasing evidence in the literature suggests a link between the brain-derived neurotrophic factor (BDNF) system and adult depression, supporting a role in the pathophysiology of the disease and response to therapy. Few studies have reported BDNF serum levels in elderly depressed subjects and their relationship with antidepressant therapy. The aim of the study was to evaluate BDNF serum levels in naive elderly depressed patients, before and after antidepressant treatment. METHODS: We enrolled n = 5 elderly naive patients affected by depression, according to the Diagnostic and Statistical Manual of Mental Disorders-IV Text Revision criteria for major depressive episode. BDNF serum levels were evaluated through ELISA method. Cognitive functions were examined by Mini Mental State Examination (MMSE) and severity of depression was assessed by Geriatric Depression Scale (GDS). BDNF levels were measured at baseline (T0) and after 2 months (T2) of escitalopram. Ten healthy elderly subjects were enrolled as a control group. RESULTS: The serum BDNF levels in patients (T0) and controls were 11.5 ± 0.6 and 13.6 ± 3.4 ng/ml (m ± SD), respectively. At T2, the patients showed a significant improvement of depressive symptoms (p < 0.05), with a not significant increase of MMSE. The serum BDNF concentrations increased to 16.0 ± 2.7 ng/ml at T2 (p < 0.05), beyond the levels of BDNF in controls. The increase in BDNF levels was significantly related to the improvement in GDS scores of the patients (r = 0.9, p < 0.05). CONCLUSIONS: Serum BDNF levels may be considered as a marker of response to antidepressant treatment for depression in the elderly.


Asunto(s)
Antidepresivos/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/sangre , Citalopram/uso terapéutico , Depresión/sangre , Depresión/tratamiento farmacológico , Trastorno Depresivo Mayor/sangre , Trastorno Depresivo Mayor/tratamiento farmacológico , Anciano , Estudios de Casos y Controles , Cognición/fisiología , Manual Diagnóstico y Estadístico de los Trastornos Mentales , Femenino , Humanos , Masculino , Índice de Severidad de la Enfermedad
7.
J Neurochem ; 125(5): 649-56, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22849384

RESUMEN

The α2 δ subunit of voltage-sensitive calcium channels (VSCCs) is the molecular target of pregabalin and gabapentin, two drugs marked for the treatment of focal epilepsy, neuropathic pain, and anxiety disorders. Expression of the α2 δ subunit is up-regulated in the dorsal horns of the spinal cord in models of neuropathic pain, suggesting that plastic changes in the α2 δ subunit are associated with pathological states. Here, we examined the expression of the α2 δ-1 subunit in the amygdala, hippocampus, and frontal cortex in the trimethyltiazoline (TMT) mouse model of innate anxiety. TMT is a volatile molecule present in the feces of the rodent predator, red fox. Mice that show a high defensive behavior during TMT exposure developed anxiety-like behavior in the following 72 h, as shown by the light-dark test. Anxiety was associated with an increased expression of the α2 δ-1 subunit of VSCCs in the amygdaloid complex at all times following TMT exposure (4, 24, and 72 h). No changes in the α2 δ-1 protein levels were seen in the hippocampus and frontal cortex of mice exposed to TMT. Pregabalin (30 mg/kg, i.p.) reduced anxiety-like behavior in TMT-exposed mice, but not in control mice. These data offer the first demonstration that the α2 δ-1 subunit of VSCCs undergoes plastic changes in a model of innate anxiety, and supports the use of pregabalin as a disease-dependent drug in the treatment of anxiety disorders.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Canales de Calcio/biosíntesis , Regulación de la Expresión Génica , Odorantes , Conducta Predatoria/fisiología , Animales , Ansiedad/psicología , Canales de Calcio/genética , Zorros , Masculino , Ratones , Regulación hacia Arriba/genética
8.
Rev Neurosci ; 23(5-6): 723-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23159867

RESUMEN

Although costly in energy and time, social play is present and evolutionarily conserved in nearly all young mammals. Ontogenetic factors responsible for this particular form of supposed rewarding behavior are incompletely understood. Here, we have focused our attention on maternal glucocorticoid hormone. We used a model in which neonate rats are fed by mothers in which drinking water has been supplemented with 0.2 mg/ml corticosterone. The control groups were lactated by water-drinking mothers. Both male and female adolescent offspring of corticosterone (CORT) supplemented dams (CORT-nursed) showed an increase in social play behavior (i.e., pinning, pouncing, wrestling/boxing and social exploration) when compared to controls. No differences were observed between CORT-nursed progeny of both sexes and controls in the exploration of the arena during the social encounter. Finally, no differences were found in CORT plasma levels in basal conditions and following a social play session in both male and female CORT-nursed rats. These results indicate that variations in the maternal glucocorticoid status are able, directly or indirectly, to influence social play behavior in the offspring, although there is no direct relationship between the level of social play behavior and the intensity of adrenocortical activation.


Asunto(s)
Corticosterona/efectos adversos , Lactancia/efectos de los fármacos , Exposición Materna/efectos adversos , Conducta Social , Análisis de Varianza , Animales , Animales Recién Nacidos , Corticosterona/sangre , Femenino , Sistema Hipotálamo-Hipofisario/fisiología , Masculino , Juego e Implementos de Juego/psicología , Ratas , Ratas Wistar , Factores Sexuales
9.
Stress ; 15(4): 378-84, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22074385

RESUMEN

Nociceptin/orphanin FQ (N/OFQ) peptide and its receptor are not only ubiquitously expressed in mammalian brain and spinal cord but are also abundant in limbic structures, particularly in the hippocampus. The widespread distribution of N/OFQ reflects the broad spectrum of its biological actions such as nociception, food intake, spontaneous locomotor activity, and learning and memory processes. Since the hippocampus is involved in the control of adrenocortical activity, its role in stress-related phenomena is well characterized. In male Wistar rats, we first examined the effects of acute restraint stress (120 min) on the brain immunohistochemical localization of N/OFQ. The analysis carried out on sections obtained at the onset of stress revealed enhanced expression of N/OFQ in CA1, CA3, and the dentate gyrus as well as increased plasma corticosterone concentrations. Next, we examined whether endogenous glucocorticoid hormone plays a role in the modulation of hippocampal N/OFQ expression in response to stress. To this end, rats were injected with corticosterone (1 mg/kg) or subjected to restraint stress 1 week after adrenalectomy. Two hours after corticosterone administration, plasma glucocorticoid concentrations were comparable to those observed after restraint stress, while N/OFQ expression had significantly increased in all the hippocampal subfields examined. By contrast, in adrenalectomized rats, stress did not modify protein expression. These results confirm that stress can affect N/OFQ expression and that glucocorticoids may constitute hormonal mediators of this complex interplay.


Asunto(s)
Péptidos Opioides/biosíntesis , Estrés Psicológico/fisiopatología , Adrenalectomía , Animales , Corticosterona/fisiología , Hipocampo , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar , Restricción Física , Nociceptina
11.
Biology (Basel) ; 11(12)2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36552294

RESUMEN

In 1981, Wylie Vale, Joachim Spiess, Catherine Rivier, and Jean Rivier reported on the characterization of a 41-amino-acid peptide from ovine hypothalamic extracts with high potency and intrinsic activity stimulating the secretion of adrenocorticotropic hormone and ß-endorphin by cultured anterior pituitary cells. With its sequence known, this neuropeptide was determined to be a hormone and consequently named corticotropin-releasing hormone (CRH), although the term corticotropin-releasing factor (CRF) is still used and preferred in some circumstances. Several decades have passed since this seminal contribution that opened a new research era, expanding the understanding of the coding of stress-related processes. The characterization of CRH receptors, the availability of CRH agonists and antagonists, and advanced immunocytochemical staining techniques have provided evidence that CRH plays a role in the regulation of several biological systems. The purpose of this review is to summarize the present knowledge of this 41-amino-acid peptide.

12.
Behav Brain Res ; 428: 113891, 2022 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-35421428

RESUMEN

Adolescence is a critical period for brain development. In most mammalian species, disturbances experienced during adolescence constitute a risk factor for several neuropsychiatric disorders. In this study, we compared the biochemical and behavioral profile induced by postweaning social isolation (PWSI) in inbred C57BL/6 N mice with that of BTBR mice, a rodent model of autism spectrum disorders. Male C57BL/6 N mice were either housed in groups of four or isolated from weaning (postnatal day 21) for four weeks before experimental analyses. After weaning, male BTBR mice were housed four per cage and analyzed at 48 days of age. PWSI reduced hippocampal levels of type 2 metabotropic glutamate (mGlu2) receptors, and glucocorticoid and mineralocorticoid receptors. A similar reduction was seen in group-housed BTBR mice. Plasma corticosterone levels in basal conditions were not influenced by PWSI, but were increased in BTBR mice. Social investigation (total and head sniffing) and the number of ultrasonic vocalizations were reduced in both PWSI mice and age-matched group-housed BTBR mice, indicating a lower social responsiveness in both groups of mice. These results suggest that absence of social stimuli during adolescence induces an endophenotype with social deficit features, which mimics the phenotype of a mouse model of autism spectrum disorders.


Asunto(s)
Trastorno Autístico , Receptores de Glutamato Metabotrópico , Animales , Trastorno Autístico/psicología , Modelos Animales de Enfermedad , Masculino , Mamíferos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Fenotipo , Receptores de Glutamato Metabotrópico/genética , Conducta Social , Aislamiento Social
13.
J Neurosci ; 30(24): 8096-101, 2010 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-20554860

RESUMEN

Oleoylethanolamide (OEA) is a biologically active lipid amide that is released by small-intestinal enterocytes during the absorption of dietary fat and inhibits feeding by engaging the nuclear receptor, peroxisome proliferator-activated receptor-alpha (PPAR-alpha). Previous studies have shown that the anorexic effects of systemically administered OEA require the activation of sensory afferents of the vagus nerve. The central circuits involved in mediating OEA-induced hypophagia remain unknown. In the present study, we report the results of in situ hybridization and immunohistochemistry experiments in rats and mice, which show that systemic injections of OEA (5-10 mg kg(-1), intraperitoneal) enhance expression of the neuropeptide oxytocin in magnocellular neurons of the paraventricular nucleus (PVN) and supraoptic nucleus (SON) of the hypothalamus. No such effect is observed with other hypothalamic neuropeptides, including vasopressin, thyrotropin-releasing hormone and pro-opiomelanocortin. The increase in oxytocin expression elicited by OEA was absent in mutant PPAR-alpha-null mice. Pharmacological blockade of oxytocin receptors in the brain by intracerebroventricular infusion of the selective oxytocin antagonist, L-368,899, prevented the anorexic effects of OEA. The results suggest that OEA suppresses feeding by activating central oxytocin transmission.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Ácidos Oléicos/farmacología , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Supraóptico/efectos de los fármacos , Animales , Canfanos/farmacología , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Endocannabinoides , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxitocina/antagonistas & inhibidores , Oxitocina/sangre , Oxitocina/genética , PPAR alfa/deficiencia , Núcleo Hipotalámico Paraventricular/metabolismo , Piperazinas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Núcleo Supraóptico/metabolismo
14.
Anesthesiology ; 114(6): 1380-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21532463

RESUMEN

BACKGROUND: Propofol is associated with postoperative mood alterations and induces a higher incidence of dreaming compared with other general anesthetics. These effects might be mediated by propofol's inhibitory action on fatty acid amide hydrolase, the enzyme that degrades the endocannabinoid anandamide. Because propofol is also associated with a higher incidence of traumatic memories from perioperative awareness and intensive care unit treatment and the endocannabinoid system is involved in regulating memory consolidation of emotional experiences, the authors investigated whether propofol, at anesthetic doses, modulates memory consolidation via an activation of the endocannabinoid system. METHODS: Male Sprague-Dawley rats were trained on an inhibitory avoidance task in which they received an inescapable foot shock upon entering the dark compartment of the apparatus. Drugs were administered intraperitoneally immediately or 30, 90, or 180 min after training. On the retention test 48 h later, the latency to reenter the dark compartment was recorded and taken as a measure of memory retention. RESULTS: The anesthetic doses of propofol administered after training significantly increased latencies of 48-h inhibitory avoidance performance (483.4 ± 181.3, 432.89 ± 214.06, 300 and 350 mg/kg, respectively; mean ± SD) compared with the corresponding vehicle group (325.33 ± 221.22, mean ± SD), which is indicative of stronger memory consolidation in propofol treated rats. Administration of a nonimpairing dose of the cannabinoid receptor antagonist rimonabant blocked the memory enhancement induced by propofol (123.39 ± 133.10, mean ± SD). Delayed administration of propofol 90 and 180 min after training or immediate posttraining administration of the benzodiazepine midazolam or the barbiturate pentobarbital did not significantly alter retention. CONCLUSIONS: These findings indicate that propofol, in contrast to other commonly used sedatives, enhances emotional memory consolidation when administered immediately after a stressful event by enhancing endocannabinoid signaling.


Asunto(s)
Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Memoria/efectos de los fármacos , Propofol/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Masculino , Memoria/fisiología , Piperidinas/metabolismo , Piperidinas/farmacología , Propofol/farmacología , Pirazoles/metabolismo , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Rimonabant
15.
Front Pharmacol ; 12: 644521, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33716754

RESUMEN

Amphetamine is a potent psychostimulant that increases brain monoamine levels. Extensive evidence demonstrated that norepinephrine is crucially involved in the regulation of memory consolidation for stressful experiences. Here, we investigated amphetamine effects on the consolidation of long-term recognition memory in rats exposed to different intensities of forced swim stress immediately after training. Furthermore, we evaluated whether such effects are dependent on the activation of the peripheral adrenergic system. To this aim, male adult Sprague Dawley rats were subjected to an object recognition task and intraperitoneally administered soon after training with amphetamine (0.5 or 1 mg/kg), or its corresponding vehicle. Rats were thereafter exposed to a mild (1 min, 25 ± 1°C) or strong (5 min, 19 ± 1°C) forced swim stress procedure. Recognition memory retention was assessed 24-h after training. Our findings showed that amphetamine enhances the consolidation of memory in rats subjected to mild stress condition, while it impairs long-term memory performance in rats exposed to strong stress. These dichotomic effects is dependent on stress-induced activation of the peripheral adrenergic response.

16.
J Nucl Med ; 50(5): 823-6, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19372483

RESUMEN

UNLABELLED: Human beta-defensin-3 (HBD-3) is an antimicrobial peptide with bactericidal effects on many gram-positive and gram-negative bacteria and some yeast species and, if radiolabeled, might be used to distinguish bacterial infection from sterile inflammation. The goals of the present study were to develop methods for radiolabeling HBD-3 with (99m)Tc and to perform preliminary investigations on (99m)Tc-labeled HBD-3 as a means to evaluate induced infection in an animal model. To this purpose, Staphylococcus aureus-induced infection was used to evaluate the capability of (99m)Tc-HBD-3 to distinguish infection from aseptic inflammation in rats. METHODS: Twenty to 40 microg of recombinant HBD-3 were labeled with (99m)Tc(+) hexa-coordinated with 3 molecules of CO and H(2)O and separated by a column from free (99m)Tc. (99m)Tc-HBD-3 was added to cultures of a bacterial suspension of S. aureus and Escherichia coli to evaluate in vitro antibacterial activity. A bacterial suspension of S. aureus and a carrageenan solution were used to induce infection and sterile inflammation, respectively, in opposite thighs of 9 adult rats. Three separate experiments were performed on groups of 3 rats each. The animals received different doses of (99m)Tc-HBD-3 injected through a cannula into the jugular vein. After sacrifice of the animals, tissue samples were obtained from sites of infection, inflammation, and control muscle (left foreleg) at 1, 3, and 5 h after (99m)Tc-HBD-3 administration. Tissue samples were weighed and then counted in a well-counter. Simultaneously, 1 mL of a standard solution of (99m)Tc-HBD-3 corresponding to each administered dose was counted. RESULTS: (99m)Tc-HBD-3 retained antibacterial activity. Radioactivity in tissue samples from the infected sites was significantly higher than that in samples of either induced inflammation or normal control muscle (ratio, approximately 3:1) at 3 and 5 h after injection, whereas similar radioactivity counts were observed for tissue samples from aseptic inflammation sites and normal control muscle. CONCLUSION: In this investigation, (99m)Tc-HBD-3 retained antibacterial activity and successfully distinguished infection from aseptic inflammation in adult rats.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones Estafilocócicas/diagnóstico por imagen , Infecciones Estafilocócicas/metabolismo , Tecnecio/farmacocinética , beta-Defensinas/farmacocinética , Animales , Sistemas de Liberación de Medicamentos/métodos , Estudios de Factibilidad , Humanos , Masculino , Proyectos Piloto , Cintigrafía , Radiofármacos/farmacocinética , Ratas , Ratas Wistar , Proteínas Recombinantes/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , beta-Defensinas/genética
17.
J Psychiatr Res ; 43(3): 247-54, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18511076

RESUMEN

Recent studies have implicated brain-derived neurotrophic factor (BDNF) in the pathophysiology of depression and the activity of antidepressant drugs. Serum BDNF levels are lower in depressed patients, and increase in response to antidepressant medication. However, how BDNF responds to different classes of antidepressant drugs is unknown. We assessed serum BDNF levels in 21 patients with major depressive episode treated with sertraline, escitalopram, or venlafaxine and 20 healthy controls. Serum samples were collected between 10 a.m. and 12 p.m. at baseline, 5 weeks, and 6 months of treatment. BDNF levels were measured via immunoassay. The severity of symptoms and response to treatment were assessed by the Hamilton rating scales for depression (HRSD). Baseline serum BDNF levels were significantly lower in depressed patients compared to controls. Sertraline increased BDNF levels after 5 weeks and 6 months of treatment. Venlafaxine increased BDNF levels only after 6 months. Escitalopram did not affect BDNF levels at either time point. A significant negative association was found between percentage increase in BDNF levels and percentage decreased in HRSD scores after 6 months of treatment. In conclusion, these results suggest that different antidepressant drugs have variable effects on serum BDNF levels. This is true even though the three different drugs were equally effective in relieving symptoms of depression and anxiety.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/sangre , Citalopram/uso terapéutico , Ciclohexanoles/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Sertralina/uso terapéutico , Adulto , Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/sangre , Trastorno Depresivo Mayor/diagnóstico , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Escalas de Valoración Psiquiátrica , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Clorhidrato de Venlafaxina
18.
Regul Pept ; 154(1-3): 32-8, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19046996

RESUMEN

The participation of hypothalamus-pituitary-adrenal axis in the gastroprotective effects of nociceptin/orphanin FQ (N/OFQ) has been investigated. Gastric mucosal lesions were induced by intragastric administration of 50% ethanol, 1 ml/rat. Rats received N/OFQ either by the intracerebroventricular (icv) route, at 3 microg/rat, or by the intraperitoneal (ip) route, at 10 microg/kg, 30 min before ethanol administration. The protective effect of icv and ip administered N/OFQ was assessed in adrenalectomized rats and in rats pretreated with the glucocorticoid receptor antagonist, mifepristone, or with the CRF receptor antagonist, alpha-helical CRF(9-41). The damaging effect of ethanol was apparently not influenced by adrenalectomy. N/OFQ markedly reduced macroscopically and histologically assessed gastric mucosal damage. The extent of reduction by N/OFQ was comparable in adrenalectomized and in sham-operated rats, with either icv or ip route of administration. Pretreatment with mifepristone, both icv (80 microg/rat) and ip (10 mg/kg) injected, did not modify the response to icv and ip N/OFQ. Pretreatment with alpha-helical CRF(9-41) (25 microg/rat icv or 250 microg/kg ip), had no effect on the reduction of gastric damage produced by icv or ip N/OFQ. Present findings suggest that the gastroprotective effects of N/OFQ on ethanol-induced damage do not involve the endocrine pathway through the HPA axis.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Péptidos Opioides/farmacología , Sistema Hipófiso-Suprarrenal/metabolismo , Gastropatías/patología , Gastropatías/prevención & control , Adrenalectomía , Animales , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Etanol/toxicidad , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Masculino , Mifepristona/administración & dosificación , Péptidos Opioides/administración & dosificación , Fragmentos de Péptidos/antagonistas & inhibidores , Ratas , Ratas Wistar , Receptores de Glucocorticoides/antagonistas & inhibidores , Gastropatías/inducido químicamente , Factores de Tiempo , Nociceptina
20.
Front Pharmacol ; 10: 976, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31551781

RESUMEN

In vulnerable individuals, chronic and persistent stress is an established risk factor for disorders that are comorbid with Alzheimer's disease (AD), such as hypertension, obesity and metabolic syndrome, and psychiatric disorders. There are no disease-modifying drugs in the treatment of AD, and all phase-3 clinical trials with anti-amyloid drugs (e.g., ß- or γ-secretase inhibitors and monoclonal antibodies) did not meet the primary endpoints. There are many reasons for the lack of efficacy of anti-amyloid drugs in AD, the most likely being a late start of treatment, considering that pathophysiological mechanisms underlying synaptic dysfunction and neuronal death begin several decades before the clinical onset of AD. The identification of risk factors is, therefore, an essential step for early treatment of AD with candidate disease-modifying drugs. Preclinical studies suggest that stress, and the resulting activation of the hypothalamic-pituitary-adrenal axis, can induce biochemical abnormalities reminiscent to those found in autoptic brain samples from individuals affected by AD (e.g., increases amyloid precursor protein and tau hyperphosphorylation). In this review, we will critically analyze the current knowledge supporting stress as a potential risk factor for AD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA