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.
Front Neurosci ; 16: 852010, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35527817

RESUMEN

(R,S)-ketamine is an N-methyl-D-aspartate (NMDA) receptor antagonist that was originally developed as an anesthetic. Most recently, (R,S)-ketamine has been used as a rapid-acting antidepressant, and we have reported that (R,S)-ketamine can also be a prophylactic against stress in adult mice. However, most pre-clinical studies have been performed in adult mice. It is still unknown how an acute (R,S)-ketamine injection influences behavior across the lifespan (e.g., to adolescent or aged populations). Here, we administered saline or (R,S)-ketamine at varying doses to adolescent (5-week-old) and aged (24-month-old) 129S6/SvEv mice of both sexes. One hour later, behavioral despair, avoidance, locomotion, perseverative behavior, or contextual fear discrimination (CFD) was assessed. A separate cohort of mice was sacrificed 1 h following saline or (R,S)-ketamine administration. Brains were processed to quantify the marker of inflammation Cyclooxygenase 2 (Cox-2) expression to determine whether the acute effects of (R,S)-ketamine were partially mediated by changes in brain inflammation. Our findings show that (R,S)-ketamine reduced behavioral despair and perseverative behavior in adolescent female, but not male, mice and facilitated CFD in both sexes at specific doses. (R,S)-ketamine reduced Cox-2 expression specifically in ventral CA3 (vCA3) of male mice. Notably, (R,S)-ketamine was not effective in aged mice. These results underscore the need for sex- and age-specific approaches to test (R,S)-ketamine efficacy across the lifespan.

2.
Int J Neuropsychopharmacol ; 25(6): 512-523, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35229871

RESUMEN

BACKGROUND: (R,S)-ketamine, an N-methyl-D-aspartate receptor antagonist, is frequently used as an anesthetic and as a rapid-acting antidepressant. We and others have reported that (R,S)-ketamine is prophylactic against stress in adult mice but have yet to test its efficacy in adolescent or aged populations. METHODS: Here, we administered saline or (R,S)-ketamine as a prophylactic at varying doses to adolescent (5-week-old) and aged (24-month-old) 129S6/SvEv mice of both sexes 1 week before a 3-shock contextual fear-conditioning (CFC) stressor. Following CFC, we assessed behavioral despair, avoidance, perseverative behavior, locomotion, and contextual fear discrimination. To assess whether the prophylactic effect could persist into adulthood, adolescent mice were injected with saline or varying doses of (R,S)-ketamine and administered a 3-shock CFC as a stressor 1 month later. Mice were then re-exposed to the aversive context 5 days later and administered behavioral tests as aforementioned. Brains were also processed to quantify Cyclooxygenase 2 expression as a proxy for inflammation to determine whether the prophylactic effects of (R,S)-ketamine were partially due to changes in brain inflammation. RESULTS: Our data indicate that (R,S)-ketamine is prophylactic at sex-specific doses in adolescent but not aged mice. (R,S)-ketamine attenuated learned fear and perseverative behavior in females, reduced behavioral despair in males, and facilitated contextual fear discrimination in both sexes. (R,S)-ketamine reduced Cyclooxygenase 2 expression specifically in ventral Cornu Ammonis region 3 of male mice. CONCLUSIONS: These findings demonstrate that prophylactic (R,S)-ketamine efficacy is sex, dose, and age dependent and will inform future studies investigating (R,S)-ketamine efficacy across the lifespan.


Asunto(s)
Ketamina , Animales , Antidepresivos/farmacología , Ciclooxigenasa 2/farmacología , Miedo , Femenino , Ketamina/farmacología , Masculino , Ratones , Estrés Psicológico
3.
Biol Psychiatry ; 90(7): 458-472, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34274107

RESUMEN

BACKGROUND: Major depressive disorder is a common, recurrent illness. Recent studies have implicated the NMDA receptor in the pathophysiology of major depressive disorder. (R,S)-ketamine, an NMDA receptor antagonist, is an effective antidepressant but has numerous side effects. Here, we characterized a novel NMDA receptor antagonist, fluoroethylnormemantine (FENM), to determine its effectiveness as a prophylactic and/or antidepressant against stress-induced maladaptive behavior. METHODS: Saline, memantine (10 mg/kg), (R,S)-ketamine (30 mg/kg), or FENM (10, 20, or 30 mg/kg) was administered before or after contextual fear conditioning in 129S6/SvEv mice. Drug efficacy was assayed using various behavioral tests. Protein expression in the hippocampus was quantified with immunohistochemistry or Western blotting. In vitro radioligand binding was used to assay drug binding affinity. Patch clamp electrophysiology was used to determine the effect of drug administration on glutamatergic activity in ventral hippocampal cornu ammonis 3 (vCA3) 1 week after injection. RESULTS: Given after stress, FENM decreased behavioral despair and reduced perseverative behavior. When administered after re-exposure, FENM facilitated extinction learning. As a prophylactic, FENM attenuated learned fear and decreased stress-induced behavioral despair. FENM was behaviorally effective in both male and female mice. (R,S)-ketamine, but not FENM, increased expression of c-fos in vCA3. Both (R,S)-ketamine and FENM attenuated large-amplitude AMPA receptor-mediated bursts in vCA3, indicating a common neurobiological mechanism for further study. CONCLUSIONS: Our results indicate that FENM is a novel drug that is efficacious when administered at various times before or after stress. Future work will further characterize FENM's mechanism of action with the goal of clinical development.


Asunto(s)
Trastorno Depresivo Mayor , Ketamina , Memantina/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Femenino , Ketamina/farmacología , Masculino , Memantina/análogos & derivados , Ratones , Estrés Psicológico
4.
Biol Psychiatry ; 89(12): 1150-1161, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33766406

RESUMEN

BACKGROUND: Posttraumatic stress disorder can develop after a traumatic event and results in heightened, inappropriate fear and anxiety. Although approximately 8% of the U.S. population is affected by posttraumatic stress disorder, only two drugs have been approved by the Food and Drug Administration to treat it, both with limited efficacy. Propranolol, a nonselective ß-adrenergic antagonist, has shown efficacy in decreasing exaggerated fear, and there has been renewed interest in using it to treat fear disorders. METHODS: Here, we sought to determine the mechanisms by which propranolol attenuates fear by utilizing an activity-dependent tagging system, ArcCreERT2 x eYFP mice. 129S6/SvEv mice were administered a 4-shock contextual fear conditioning paradigm followed by immediate or delayed context reexposures. Saline or propranolol was administered either before or after the first context reexposure. To quantify hippocampal, prefrontal, and amygdalar memory traces, ArcCreERT2 x eYFP mice were administered a delayed context reexposure with either a saline or propranolol injection before context reexposure. RESULTS: Propranolol decreased fear expression only when administered before a delayed context reexposure. Fear memory traces were affected in the dorsal dentate gyrus and basolateral amygdala after propranolol administration in the ArcCreERT2 x eYFP mice. Propranolol acutely altered functional connectivity between the hippocampal, cortical, and amygdalar regions. CONCLUSIONS: These data indicate that propranolol may decrease fear expression by altering network-correlated activity and by weakening the reactivation of the initial traumatic memory trace. This work contributes to the understanding of noradrenergic drugs as therapeutic aids for patients with posttraumatic stress disorder.


Asunto(s)
Complejo Nuclear Basolateral , Propranolol , Amígdala del Cerebelo , Animales , Miedo , Humanos , Memoria , Ratones , Propranolol/farmacología
5.
Neuropsychopharmacology ; 46(5): 882-890, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32919399

RESUMEN

In the United States, ~1.4 million individuals identify as transgender. Many transgender adolescents experience gender dysphoria related to incongruence between their gender identity and sex assigned at birth. This dysphoria may worsen as puberty progresses. Puberty suppression by gonadotropin-releasing hormone agonists (GnRHa), such as leuprolide, can help alleviate gender dysphoria and provide additional time before irreversible changes in secondary sex characteristics may be initiated through feminizing or masculinizing hormone therapy congruent with the adolescent's gender experience. However, the effects of GnRH agonists on brain function and mental health are not well understood. Here, we investigated the effects of leuprolide on reproductive function, social and affective behavior, cognition, and brain activity in a rodent model. Six-week-old male and female C57BL/6J mice were injected daily with saline or leuprolide (20 µg) for 6 weeks and tested in several behavioral assays. We found that leuprolide increases hyperlocomotion, changes social preference, and increases neuroendocrine stress responses in male mice, while the same treatment increases hyponeophagia and despair-like behavior in females. Neuronal hyperactivity was found in the dentate gyrus (DG) of leuprolide-treated females, but not males, consistent with the elevation in hyponeophagia and despair-like behavior in females. These data show for the first time that GnRH agonist treatment after puberty onset exerts sex-specific effects on social- and affective behavior, stress regulation, and neural activity. Investigating the behavioral and neurobiological effects of GnRH agonists in mice will be important to better guide the investigation of potential consequences of this treatment for youth experiencing gender dysphoria.


Asunto(s)
Personas Transgénero , Adolescente , Animales , Femenino , Identidad de Género , Hormona Liberadora de Gonadotropina , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Pubertad , Estados Unidos
6.
Front Cell Dev Biol ; 8: 541, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32719795

RESUMEN

Early diagnosis of Alzheimer's disease (AD) supposedly increases the effectiveness of therapeutic interventions. However, presently available diagnostic procedures are either invasive or require complex and expensive technologies, which cannot be applied at a larger scale to screen populations at risk of AD. We were looking for a biomarker allowing to unveil a dysfunction of molecular mechanisms, which underly synaptic plasticity and memory, before the AD phenotype is manifested and investigated the effects of transcranial direct current stimulation (tDCS) in 3×Tg-AD mice, an experimental model of AD which does not exhibit any long-term potentiation (LTP) and memory deficits at the age of 3 months (3×Tg-AD-3M). Our results demonstrated that tDCS differentially affected 3×Tg-AD-3M and age-matched wild-type (WT) mice. While tDCS increased LTP at CA3-CA1 synapses and memory in WT mice, it failed to elicit these effects in 3×Tg-AD-3M mice. Remarkably, 3×Tg-AD-3M mice did not show the tDCS-dependent increases in pCREB Ser133 and pCaMKII Thr286 , which were found in WT mice. Of relevance, tDCS induced a significant increase of plasma BDNF levels in WT mice, which was not found in 3×Tg-AD-3M mice. Collectively, our results showed that plasticity mechanisms are resistant to tDCS effects in the pre-AD stage. In particular, the lack of BDNF responsiveness to tDCS in 3×Tg-AD-3M mice suggests that combining tDCS with dosages of plasma BDNF levels may provide an easy-to-detect and low-cost biomarker of covert impairment of synaptic plasticity mechanisms underlying memory, which could be clinically applicable. Testing proposed here might be useful to identify AD in its preclinical stage, allowing timely and, hopefully, more effective disease-modifying interventions.

7.
Behav Brain Res ; 378: 112238, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31563463

RESUMEN

Individuals with peripheral inflammation are a particularly vulnerable population for developing depression and are also more resistant towards traditional antidepressants. This signals the need for novel drugs that can effectively treat this patient population. Recently, we have demonstrated that (R,S)-ketamine is a prophylactic against a variety of stressors, but have yet to test if it is protective against inflammatory-induced vulnerability to a stressor. Here, male 129S6/SvEv mice were administered saline or (R,S)-ketamine (30 mg/kg) 6 days before an injection of vehicle (VEH) or lipopolysaccharide (LPS) (0.83 or 1.0 mg/kg, serotypes O111:B4 or O127:B8). Twenty-four hours after LPS administration, mice were administered a contextual fear conditioning (CFC) paradigm, followed by a context re-exposure and the forced swim test (FST). In a separate cohort, we tested if (R,S)-ketamine was effective as a prophylactic against polyinosinic-polycytidylic acid (PIC), a viral mimetic. (R,S)-ketamine was effective as a prophylactic for attenuating learned fear in the O111:B4 and O127:B8 strains of LPS. (R,S)-ketamine was also effective as a prophylactic for decreasing stress-induced depressive-like behavior in the O111:B4 and O127:B8 strains of LPS. Both of these effects were limited to administration of 1.0, but not 0.83 mg/kg of the O111:B4 and O127:B8 strains of LPS. (R,S)-ketamine was not effective against either stress phenotype following PIC administration. These data suggest that prophylactic (R,S)-ketamine may protect against selective inflammation-induced stress phenotypes following an inflammatory challenge. Future studies will be necessary to determine if (R,S)-ketamine can be useful in patient populations with peripheral inflammation.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Depresión/prevención & control , Inflamación/complicaciones , Ketamina/farmacología , Estrés Psicológico/prevención & control , Animales , Antidepresivos/administración & dosificación , Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Depresión/etiología , Miedo/fisiología , Inflamación/inducido químicamente , Ketamina/administración & dosificación , Lipopolisacáridos/administración & dosificación , Masculino , Ratones , Ratones de la Cepa 129 , Estrés Psicológico/etiología
8.
Nat Commun ; 10(1): 4799, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31641124

RESUMEN

Metabolic diseases harm brain health and cognitive functions, but whether maternal metabolic unbalance may affect brain plasticity of next generations is still unclear. Here, we demonstrate that maternal high fat diet (HFD)-dependent insulin resistance multigenerationally impairs synaptic plasticity, learning and memory. HFD downregulates BDNF and insulin signaling in maternal tissues and epigenetically inhibits BDNF expression in both germline and hippocampus of progeny. Notably, exposure of the HFD offspring to novel enriched environment restores Bdnf epigenetic activation in the male germline and counteracts the transmission of cognitive impairment to the next generations. BDNF administration to HFD-fed mothers or preserved insulin sensitivity in HFD-fed p66Shc KO mice also prevents the intergenerational transmission of brain damage to the progeny. Collectively, our data suggest that maternal diet multigenerationally impacts on descendants' brain health via gametic mechanisms susceptible to lifestyle.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Aprendizaje/fisiología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Epigénesis Genética , Femenino , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica , Hipocampo/fisiopatología , Histona Desacetilasa 2/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/metabolismo , Sirtuina 2/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética
9.
Science ; 364(6440): 578-583, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-31073064

RESUMEN

Young adult-born granule cells (abGCs) in the dentate gyrus (DG) have a profound impact on cognition and mood. However, it remains unclear how abGCs distinctively contribute to local DG information processing. We found that the actions of abGCs in the DG depend on the origin of incoming afferents. In response to lateral entorhinal cortex (LEC) inputs, abGCs exert monosynaptic inhibition of mature granule cells (mGCs) through group II metabotropic glutamate receptors. By contrast, in response to medial entorhinal cortex (MEC) inputs, abGCs directly excite mGCs through N-methyl-d-aspartate receptors. Thus, a critical function of abGCs may be to regulate the relative synaptic strengths of LEC-driven contextual information versus MEC-driven spatial information to shape distinct neural representations in the DG.


Asunto(s)
Giro Dentado/fisiología , Corteza Entorrinal/fisiología , Hipocampo/fisiología , Neuronas/fisiología , Animales , Células Cultivadas , Potenciales Evocados , Humanos , Ratones , Ratones Transgénicos , Receptores de N-Metil-D-Aspartato/fisiología , Sinapsis/fisiología
10.
PLoS Pathog ; 15(3): e1007617, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30870531

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a DNA neurotropic virus, usually establishing latent infections in the trigeminal ganglia followed by periodic reactivations. Although numerous findings suggested potential links between HSV-1 and Alzheimer's disease (AD), a causal relation has not been demonstrated yet. Hence, we set up a model of recurrent HSV-1 infection in mice undergoing repeated cycles of viral reactivation. By virological and molecular analyses we found: i) HSV-1 spreading and replication in different brain regions after thermal stress-induced virus reactivations; ii) accumulation of AD hallmarks including amyloid-ß protein, tau hyperphosphorylation, and neuroinflammation markers (astrogliosis, IL-1ß and IL-6). Remarkably, the progressive accumulation of AD molecular biomarkers in neocortex and hippocampus of HSV-1 infected mice, triggered by repeated virus reactivations, correlated with increasing cognitive deficits becoming irreversible after seven cycles of reactivation. Collectively, our findings provide evidence that mild and recurrent HSV-1 infections in the central nervous system produce an AD-like phenotype and suggest that they are a risk factor for AD.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/virología , Herpesvirus Humano 1/patogenicidad , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides , Animales , Encéfalo/virología , Cognición/fisiología , Trastornos del Conocimiento/etiología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/virología , Modelos Animales de Enfermedad , Femenino , Herpesvirus Humano 1/metabolismo , Ratones , Ratones Endogámicos BALB C , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/virología , Ganglio del Trigémino/virología , Activación Viral/fisiología , Replicación Viral/fisiología
11.
Biol Psychiatry ; 84(11): 846-856, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29615190

RESUMEN

BACKGROUND: We previously reported that a single injection of ketamine prior to stress protects against the onset of depressive-like behavior and attenuates learned fear. However, the molecular pathways and brain circuits underlying ketamine-induced stress resilience are still largely unknown. METHODS: Here, we tested whether prophylactic ketamine administration altered neural activity in the prefrontal cortex and/or hippocampus. Mice were injected with saline or ketamine (30 mg/kg) 1 week before social defeat. Following behavioral tests assessing depressive-like behavior, mice were sacrificed and brains were processed to quantify ΔFosB expression. In a second set of experiments, mice were stereotaxically injected with viral vectors into ventral CA3 (vCA3) in order to silence or overexpress ΔFosB prior to prophylactic ketamine administration. In a third set of experiments, ArcCreERT2 mice, a line that allows for the indelible labeling of neural ensembles activated by a single experience, were used to quantify memory traces representing a contextual fear conditioning experience following prophylactic ketamine administration. RESULTS: Prophylactic ketamine administration increased ΔFosB expression in the ventral dentate gyrus and vCA3 of social defeat mice but not of control mice. Transcriptional silencing of ΔFosB activity in vCA3 inhibited prophylactic ketamine efficacy, while overexpression of ΔFosB mimicked and occluded ketamine's prophylactic effects. In ArcCreERT2 mice, ketamine administration altered memory traces representing the contextual fear conditioning experience in vCA3 but not in the ventral dentate gyrus. CONCLUSIONS: Our data indicate that prophylactic ketamine may be protective against a stressor by altering neural activity, specifically the neural ensembles representing an individual stressor in vCA3.


Asunto(s)
Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/fisiología , Depresión/tratamiento farmacológico , Ketamina/farmacología , Estrés Psicológico/tratamiento farmacológico , Animales , Escala de Evaluación de la Conducta , Condicionamiento Clásico/efectos de los fármacos , Miedo , Femenino , Masculino , Ratones , Proteínas Proto-Oncogénicas c-fos/metabolismo
12.
Neuropsychopharmacology ; 43(9): 1813-1821, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29599484

RESUMEN

Recently, we have shown that ketamine given prior to stress exposure protects against the development of depressive-like behavior in mice. These data suggest that it may be possible to prevent the induction of affective disorders before they develop by administering prophylactic pharmaceuticals, a relatively nascent and unexplored strategy for psychiatry. Here, we performed metabolomics analysis of brain and plasma following prophylactic ketamine treatment in order to identify markers of stress resilience enhancement. We administered prophylactic ketamine in mice to buffer against fear expression. Following behavioral analyses, untargeted metabolomic profiling was performed on both hemispheres of the prefrontal cortex (PFC) and the hippocampus (HPC), and plasma. We found that prophylactic ketamine attenuated learned fear. Eight metabolites were changed in the PFC and HPC upon ketamine treatment. Purine and pyrimidine metabolism were most significantly changed in the HPC, PFC, and, interestingly, plasma of mice two weeks after prophylactic administration. Moreover, most precursors to inhibitory neurotransmitters were increased whereas precursors to excitatory neurotransmitters were decreased. Strikingly, these long-term metabolomic changes were not observed when no stressor was administered. Our results suggest that prophylactic treatment differentially affects purine and pyrimidine metabolism and neurotransmission in brain and plasma following stress, which may underlie the long-lasting resilience to stress induced by a single injection of ketamine. These data may provide novel targets for prophylactic development, and indicate an interaction effect of prophylactic ketamine and stress. To our knowledge, this is the first study that identifies metabolomic alterations and biomarker candidates for prophylactic ketamine efficacy in mice.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ketamina/farmacología , Psicotrópicos/farmacología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Animales , Miedo/efectos de los fármacos , Miedo/fisiología , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Metaboloma/efectos de los fármacos , Ratones de la Cepa 129 , Neurotransmisores/metabolismo , Purinas/metabolismo , Pirimidinas/metabolismo
13.
Sci Rep ; 8(1): 262, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321633

RESUMEN

Exposure to extremely low-frequency electromagnetic fields (ELFEF) influences the expression of key target genes controlling adult neurogenesis and modulates hippocampus-dependent memory. Here, we assayed whether ELFEF stimulation affects olfactory memory by modulating neurogenesis in the subventricular zone (SVZ) of the lateral ventricle, and investigated the underlying molecular mechanisms. We found that 30 days after the completion of an ELFEF stimulation protocol (1 mT; 50 Hz; 3.5 h/day for 12 days), mice showed enhanced olfactory memory and increased SVZ neurogenesis. These effects were associated with upregulated expression of mRNAs encoding for key regulators of adult neurogenesis and were mainly dependent on the activation of the Wnt pathway. Indeed, ELFEF stimulation increased Wnt3 mRNA expression and nuclear localization of its downstream target ß-catenin. Conversely, inhibition of Wnt3 by Dkk-1 prevented ELFEF-induced upregulation of neurogenic genes and abolished ELFEF's effects on olfactory memory. Collectively, our findings suggest that ELFEF stimulation increases olfactory memory via enhanced Wnt/ß-catenin signaling in the SVZ and point to ELFEF as a promising tool for enhancing SVZ neurogenesis and olfactory function.


Asunto(s)
Campos Electromagnéticos , Ventrículos Laterales/fisiología , Memoria , Neurogénesis , Corteza Olfatoria/fisiología , Corteza Olfatoria/efectos de la radiación , Vía de Señalización Wnt , Animales , Discriminación en Psicología , Femenino , Masculino , Ratones , Odorantes , Percepción Olfatoria , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
14.
Sci Rep ; 6: 22180, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26908001

RESUMEN

The effects of transcranial direct current stimulation (tDCS) on brain functions and the underlying molecular mechanisms are yet largely unknown. Here we report that mice subjected to 20-min anodal tDCS exhibited one-week lasting increases in hippocampal LTP, learning and memory. These effects were associated with enhanced: i) acetylation of brain-derived neurotrophic factor (Bdnf) promoter I; ii) expression of Bdnf exons I and IX; iii) Bdnf protein levels. The hippocampi of stimulated mice also exhibited enhanced CREB phosphorylation, pCREB binding to Bdnf promoter I and recruitment of CBP on the same regulatory sequence. Inhibition of acetylation and blockade of TrkB receptors hindered tDCS effects at molecular, electrophysiological and behavioral levels. Collectively, our findings suggest that anodal tDCS increases hippocampal LTP and memory via chromatin remodeling of Bdnf regulatory sequences leading to increased expression of this gene, and support the therapeutic potential of tDCS for brain diseases associated with impaired neuroplasticity.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Epigénesis Genética , Potenciación a Largo Plazo , Memoria , Estimulación Transcraneal de Corriente Directa , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Ensamble y Desensamble de Cromatina , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor trkB/metabolismo
15.
J Neurosci ; 34(38): 12893-903, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25232124

RESUMEN

Intracellular accumulation of amyloid-ß (Aß) protein has been proposed as an early event in AD pathogenesis. In patients with mild cognitive impairment, intraneuronal Aß immunoreactivity was found especially in brain regions critically involved in the cognitive deficits of AD. Although a large body of evidence demonstrates that Aß42 accumulates intraneuronally ((in)Aß), the action and the role of Aß42 buildup on synaptic function have been poorly investigated. Here, we demonstrate that basal synaptic transmission and LTP were markedly depressed following Aß42 injection into the neuron through the patch pipette. Control experiments performed with the reverse peptide (Aß42-1) allowed us to exclude that the effects of (in)Aß depended on changes in oncotic pressure. To further investigate (in)Aß synaptotoxicity we used an Aß variant harboring oxidized methionine in position 35 that does not cross the neuronal plasma membrane and is not uploaded from the extracellular space. This Aß42 variant had no effects on synaptic transmission and plasticity when applied extracellularly, but induced synaptic depression and LTP inhibition after patch-pipette dialysis. Finally, the injection of an antibody raised against human Aß42 (6E10) in CA1 pyramidal neurons of mouse hippocampal brain slices and autaptic microcultures did not, per se, significantly affect LTP and basal synaptic transmission, but it protected against the toxic effects of extracellular Aß42. Collectively, these findings suggest that Aß42-induced impairment of glutamatergic synaptic function depends on its internalization and intracellular accumulation thus paving the way to a systemic proteomic analysis of intracellular targets/partners of Aß42.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Ácido Glutámico/fisiología , Hipocampo/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Transmisión Sináptica/efectos de los fármacos , Péptidos beta-Amiloides/administración & dosificación , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/fisiología , Espacio Intracelular/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Microinyecciones , Plasticidad Neuronal/fisiología , Fragmentos de Péptidos/administración & dosificación , Cultivo Primario de Células , Transmisión Sináptica/fisiología
16.
Mol Neurobiol ; 49(3): 1472-86, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24532268

RESUMEN

Throughout life, adult neurogenesis generates new neurons in the dentate gyrus of hippocampus that have a critical role in memory formation. Strategies able to stimulate this endogenous process have raised considerable interest because of their potential use to treat neurological disorders entailing cognitive impairment. We previously reported that mice exposed to extremely low-frequency electromagnetic fields (ELFEFs) showed increased hippocampal neurogenesis. Here, we demonstrate that the ELFEF-dependent enhancement of hippocampal neurogenesis improves spatial learning and memory. To gain insights on the molecular mechanisms underlying ELFEFs' effects, we extended our studies to an in vitro model of neural stem cells (NSCs) isolated from the hippocampi of newborn mice. We found that ELFEFs enhanced proliferation and neuronal differentiation of hippocampal NSCs by regulation of epigenetic mechanisms leading to pro-neuronal gene expression. Upon ELFEF stimulation of NSCs, we observed a significant enhancement of expression of the pro-proliferative gene hairy enhancer of split 1 and the neuronal determination genes NeuroD1 and Neurogenin1. These events were preceded by increased acetylation of H3K9 and binding of the phosphorylated transcription factor cAMP response element-binding protein (CREB) on the regulatory sequence of these genes. Such ELFEF-dependent epigenetic modifications were prevented by the Cav1-channel blocker nifedipine, and were associated with increased occupancy of CREB-binding protein (CBP) to the same loci within the analyzed promoters. Our results unravel the molecular mechanisms underlying the ELFEFs' ability to improve endogenous neurogenesis, pointing to histone acetylation-related chromatin remodeling as a critical determinant. These findings could pave the way to the development of novel therapeutic approaches in regenerative medicine.


Asunto(s)
Campos Electromagnéticos , Epigénesis Genética/fisiología , Hipocampo/citología , Hipocampo/fisiología , Neurogénesis/fisiología , Animales , Proliferación Celular/fisiología , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
17.
Behav Brain Res ; 265: 61-8, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24525423

RESUMEN

Epigenetic modifications such as histone acetylation in cortical or allocortical regions have been shown to be necessary for the formation of long-term memories. Here we investigated whether similar changes were occurring also in the ventral striatum and whether they are necessary for the consolidation of aversive memory. To this purpose we performed immediate post-training focal administrations of the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA, 5, 10 or 15 µg/side) or the DNA methyltransferase (DNMT) inhibitor, 5-aza-2'-deoxycytidine (5-AZA, 0.0625 or 0.125 µg/side) in the ventral striatum of mice trained in one-trial inhibitory avoidance task. Intra-ventral striatal SAHA administrations, immediately after training, improved memory retention. Opposite effects were found with 5-AZA. We also found that training in the one-trial inhibitory avoidance is accompanied by increased acetylation of specific residues that can be further increased by intra-VS SAHA administrations. Intra-VS 5-AZA administrations on the other hand reduced training-induced histones acetylation at the same residues. These findings imply the occurrence of histone acetylation in the ventral striatum in order to store aversive memory. Moreover, they suggest that the effects induced by the DNMT inhibitor 5-AZA may at least partially due to blockade of H3 and H4 acetylation. These results suggest that the contemporary activation of similar molecular mechanisms might be needed in different brain regions to enable the formation of long-term memories.


Asunto(s)
Reacción de Prevención/fisiología , Cuerpo Estriado/metabolismo , Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Memoria/fisiología , Análisis de Varianza , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Cuerpo Estriado/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Decitabina , Relación Dosis-Respuesta a Droga , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Masculino , Memoria/efectos de los fármacos , Ratones , Tiempo de Reacción/efectos de los fármacos , Vorinostat
18.
Eur J Neurosci ; 39(6): 893-903, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24382162

RESUMEN

In recent years, much effort has been devoted to identifying stimuli capable of enhancing adult neurogenesis, a process that generates new neurons throughout life, and that appears to be dysfunctional in the senescent brain and in several neuropsychiatric and neurodegenerative diseases. We previously reported that in vivo exposure to extremely low-frequency electromagnetic fields (ELFEFs) promotes the proliferation and neuronal differentiation of hippocampal neural stem cells (NSCs) that functionally integrate in the dentate gyrus. Here, we extended our studies to specifically assess the influence of ELFEFs on hippocampal newborn cell survival, which is a very critical issue in adult neurogenesis regulation. Mice were injected with 5-bromo-2'-deoxyuridine (BrdU) to label newborn cells, and were exposed to ELFEFs 9 days later, when the most dramatic decrease in the number of newly generated neurons occurs. The results showed that ELFEF exposure (3.5 h/day for 6 days) enhanced newborn neuron survival as documented by double staining for BrdU and doublecortin, to identify immature neurons, or NeuN labeling of mature neurons. The effects of ELFEFs were associated with enhanced spatial learning and memory. In an in vitro model of hippocampal NSCs, ELFEFs exerted their pro-survival action by rescuing differentiating neurons from apoptotic cell death. Western immunoblot assay revealed reduced expression of the pro-apoptotic protein Bax, and increased levels of the anti-apoptotic protein Bcl-2, in the hippocampi of ELFEF-exposed mice as well as in ELFEF-exposed NSC cultures, as compared with their sham-exposed counterparts. Our results may have clinical implications for the treatment of impaired neurogenesis associated with brain aging and neurodegenerative diseases.


Asunto(s)
Apoptosis , Campos Electromagnéticos , Hipocampo/efectos de la radiación , Neuronas/efectos de la radiación , Animales , Supervivencia Celular/efectos de la radiación , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Hipocampo/fisiología , Masculino , Aprendizaje por Laberinto , Memoria , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
19.
PLoS One ; 8(8): e73246, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23991183

RESUMEN

Neural stem cells generate neurons in the hippocampal dentate gyrus in mammals, including humans, throughout adulthood. Adult hippocampal neurogenesis has been the focus of many studies due to its relevance in processes such as learning and memory and its documented impairment in some neurodegenerative diseases. However, we are still far from having a complete picture of the mechanism regulating this process. Our study focused on the possible role of cyclic nucleotide-gated (CNG) channels. These voltage-independent channels activated by cyclic nucleotides, first described in retinal and olfactory receptors, have been receiving increasing attention for their involvement in several brain functions. Here we show that the rod-type, CNGA1, and olfactory-type, CNGA2, subunits are expressed in hippocampal neural stem cells in culture and in situ in the hippocampal neurogenic niche of adult mice. Pharmacological blockade of CNG channels did not affect cultured neural stem cell proliferation but reduced their differentiation towards the neuronal phenotype. The membrane permeant cGMP analogue, 8-Br-cGMP, enhanced neural stem cell differentiation to neurons and this effect was prevented by CNG channel blockade. In addition, patch-clamp recording from neuron-like differentiating neural stem cells revealed cGMP-activated currents attributable to ion flow through CNG channels. The current work provides novel insights into the role of CNG channels in promoting hippocampal neurogenesis, which may prove to be relevant for stem cell-based treatment of cognitive impairment and brain damage.


Asunto(s)
GMP Cíclico/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Hipocampo/citología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Animales , Western Blotting , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Hipocampo/efectos de los fármacos , Ratones , Técnicas de Placa-Clamp
20.
Brain ; 136(Pt 4): 1216-30, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23518710

RESUMEN

Cocaine seeking behaviour and relapse have been linked to impaired potentiation and depression at excitatory synapses in the nucleus accumbens, but the mechanism underlying this process is poorly understood. We show that, in the rat nucleus accumbens core, D-serine is the endogenous coagonist of N-methyl-D-aspartate receptors, and its presence is essential for N-methyl-D-aspartate receptor-dependent potentiation and depression of synaptic transmission. Nucleus accumbens core slices obtained from cocaine-treated rats after 1 day of abstinence presented significantly reduced D-serine concentrations, increased expression of the D-serine degrading enzyme, D-amino acid oxidase, and downregulated expression of serine racemase, the enzyme responsible for D-serine synthesis. The D-serine deficit was associated with impairment of potentiation and depression of glutamatergic synaptic transmission, which was restored by slice perfusion with exogenous D-serine. Furthermore, in vivo administration of D-serine directly into the nucleus accumbens core blocked behavioural sensitization to cocaine. These results provide evidence for a critical role of D-serine signalling in synaptic plasticity relevant to cocaine addiction.


Asunto(s)
Cocaína/farmacología , Plasticidad Neuronal/efectos de los fármacos , Núcleo Accumbens/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/antagonistas & inhibidores , Transmisión Sináptica/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Equidae , Masculino , Ratones , Núcleo Accumbens/patología , Núcleo Accumbens/ultraestructura , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/agonistas , Serina/metabolismo , Serina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...