Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Biol ; 436(5): 168271, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37699454

RESUMEN

Algae encode up to five different types of cryptochrome photoreceptors. So far, relatively little is known about the biological functions of the DASH (Drosophila, Arabidopsis, Synechocystis and Homo)-type cryptochromes. The green alga Chlamydomonas reinhardtii encodes two of them. CRY-DASH1 also called DCRY1 has its maximal absorption peak in the UV-A range. It is localized in the chloroplast and plays an important role in balancing the photosynthetic machinery. Here, we performed a comparative analysis of chloroplast proteins from wild type and a knockout mutant of CRY-DASH1 named cry-dash1mut, using label-free quantitative proteomics as well as immunoblotting. Our results show upregulation of enzymes involved in specific pathways in the mutant including key enzymes of chlorophyll and carotenoid biosynthesis consistent with increased levels of photosynthetic pigments in cry-dash1mut. There is also an increase in certain redox as well as photosystem I and II proteins, including D1. Strikingly, CRY-DASH1 is coregulated in a D1 deletion mutant, where its amount is increased. In contrast, key proteins of the central carbon metabolism, including glycolysis/gluconeogenesis, dark fermentation and the oxidative pentose phosphate pathway are downregulated in cry-dash1mut. Similarly, enzymes of histidine biosynthesis are downregulated in cry-dash1mut leading to a reduction in the amount of free histidine. Yet, transcripts encoding for several of these proteins are at a similar level in the wild type and cry-dash1mut or even opposite. We show that CRY-DASH1 can bind to RNA, taking the psbA RNA encoding D1 as target. These data suggest that CRY-DASH1 regulates plastidial metabolic pathways at the posttranscriptional level.


Asunto(s)
Chlamydomonas reinhardtii , Proteínas de Cloroplastos , Criptocromos , Fotosíntesis , Plastidios , Vías Biosintéticas , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/metabolismo , Proteínas de Cloroplastos/genética , Proteínas de Cloroplastos/metabolismo , Criptocromos/genética , Criptocromos/metabolismo , Regulación hacia Abajo , Histidina/biosíntesis , Histidina/genética , Plastidios/genética , Plastidios/metabolismo , Rayos Ultravioleta , Eliminación de Gen , Transcripción Genética
2.
Front Neuroendocrinol ; 70: 101068, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37061205

RESUMEN

Research in preclinical models indicates that estrogens are neuroprotective and positively impact cognitive aging. However, clinical data are equivocal as to the benefits of menopausal estrogen therapy to the brain and cognition. Pre-existing cardiometabolic disease may modulate mechanisms by which estrogens act, potentially reducing or reversing protections they provide against cognitive decline. In the current review we propose mechanisms by which cardiometabolic disease may alter estrogen effects, including both alterations in actions directly on brain memory systems and actions on cardiometabolic systems, which in turn impact brain memory systems. Consideration of mechanisms by which estrogen administration can exert differential effects dependent upon health phenotype is consistent with the move towards precision or personalized medicine, which aims to determine which treatment interventions will work for which individuals. Understanding effects of estrogens in both healthy and unhealthy models of aging is critical to optimizing the translational link between preclinical and clinical research.


Asunto(s)
Enfermedades Cardiovasculares , Estrógenos , Humanos , Encéfalo , Menopausia/psicología , Cognición , Enfermedades Cardiovasculares/tratamiento farmacológico
3.
Cereb Cortex ; 31(7): 3194-3212, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33675359

RESUMEN

Thalamocortical neurons (TCNs) play a critical role in the maintenance of thalamocortical oscillations, dysregulation of which can result in certain types of seizures. Precise control over firing rates of TCNs is foundational to these oscillations, yet the transcriptional mechanisms that constrain these firing rates remain elusive. We hypothesized that Shox2 is a transcriptional regulator of ion channels important for TCN function and that loss of Shox2 alters firing frequency and activity, ultimately perturbing thalamocortical oscillations into an epilepsy-prone state. In this study, we used RNA sequencing and quantitative PCR of control and Shox2 knockout mice to determine Shox2-affected genes and revealed a network of ion channel genes important for neuronal firing properties. Protein regulation was confirmed by Western blotting, and electrophysiological recordings showed that Shox2 KO impacted the firing properties of a subpopulation of TCNs. Computational modeling showed that disruption of these conductances in a manner similar to Shox2's effects modulated frequency of oscillations and could convert sleep spindles to near spike and wave activity, which are a hallmark for absence epilepsy. Finally, Shox2 KO mice were more susceptible to pilocarpine-induced seizures. Overall, these results reveal Shox2 as a transcription factor important for TCN function in adult mouse thalamus.


Asunto(s)
Potenciales de Acción/fisiología , Corteza Cerebral/metabolismo , Proteínas de Homeodominio/biosíntesis , Neuronas/metabolismo , Convulsiones/metabolismo , Tálamo/metabolismo , Animales , Proteínas de Homeodominio/genética , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Red Nerviosa/metabolismo , Convulsiones/genética , Convulsiones/prevención & control , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
4.
Brain Behav Immun Health ; 6: 100104, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34589865

RESUMEN

The impact of trauma on mental health is complex with poorly understood underlying mechanisms. Mitochondrial dysfunction is increasingly implicated in psychopathologies and mood disorders, including post-traumatic stress disorder (PTSD). We hypothesized that defects in mitochondrial energy metabolism in the cerebellum, an emerging region of interest in the pathobiology of mood disorders, would be associated with PTSD-like symptomatology, and that PTSD-like symptomatology would correlate with the activities of the mitochondrial electron transport chain (mtETC) and fatty acid oxidation (FAO) pathways. We assayed mitochondrial energy metabolism and fatty acid profiling using targeted metabolomics in mice exposed to a recently developed paradigm for PTSD-induction. 48 wild type male FVB.129P2 mice were exposed to a trauma, and PTSD-like and resilient animals were identified using behavioral profiling. Mice displaying PTSD-like symptomatology displayed reduced mtETC complex activities in the cerebellum, and cerebellar mtETC complex activity negatively correlated with PTSD-like symptomatology. PTSD-like animals also displayed fatty acid profiles consistent with FAO dysfunction in both cerebellum and plasma. Machine learning analysis of all biochemical measures in this cohort of animals also identified plasma acetylcarnitine, along with reduced activity of cerebellar complex I and IV as well as succinate:cytochrome c oxidoreductase as state predictive discriminators of PTSD-symptomatology. Our data also suggest that trauma-induced impaired mtETC function in the cerebellum and concomitant impaired multi-system fatty acid oxidation are candidate drivers of PTSD-like behavior in mice. These bioenergetic and metabolic changes may offer an informative window into the underlying biology and highlight novel potential targets for diagnostics and therapeutic interventions in PTSD.

5.
Commun Biol ; 1: 82, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271963

RESUMEN

Previous genomic studies in humans indicate that SIRT1, a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, is involved in anxiety and depression, but the mechanisms are unclear. We previously showed that SIRT1 is highly activated in the nuclear fraction of the dentate gyrus of the chronically stressed animals and inhibits memory formation and increases anhedonic behavior during chronic stress, but specific functional targets of cytoplasmic SIRT1 are unknown. Here, we demonstrate that SIRT1 activity rapidly modulates intrinsic and synaptic properties of the dentate gyrus granule cells and anxiety behaviors through deacetylation of BK channel α subunits in control animals. Chronic stress decreases BKα channel membrane expression, and SIRT1 activity has no rapid effects on synaptic transmission or intrinsic properties in the chronically stressed animal. These results suggest SIRT1 activity rapidly modulates the physiological function of the dentate gyrus, and this modulation participates in the maladaptive stress response.

6.
Stress ; 21(4): 292-303, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29916754

RESUMEN

Post-traumatic stress disorder (PTSD) is characterized by the development of paradoxical memory disturbances including intrusive memories and amnesia for specific details of the traumatic experience. Despite evidence that women are at higher risk to develop PTSD, most animal research has focused on the processes by which male rodents develop adaptive fear memory. As such, the mechanisms contributing to sex differences in the development of PTSD-like memory disturbances are poorly understood. In this investigation, we exposed adult male and female Wistar rats to the synthetic alarm odor 2,4,5-trimethylthiazole (TMT) to assess development of generalized fear behavior and rapid modulation of glutamate uptake and signaling cascades associated with hippocampus-dependent long-term memory. We report that female Wistar rats exposed to alarm odor exhibit context discrimination impairments relative to TMT-exposed male rats, suggesting the intriguing possibility that females are at greater risk in developing generalized fear memories. Mechanistically, alarm odor exposure rapidly modulated signaling cascades consistent with activation of the CREB shut-off cascade in the male, but not the female hippocampus. Moreover, TMT exposure dampened glutamate uptake and affected expression of the glutamate transporter, GLT-1 in the hippocampus. Taken together, these results provide evidence for rapid sex-dependent modulation of CREB signaling in the hippocampus by alarm odor exposure which may contribute to the development of generalized fear.


Asunto(s)
Miedo/fisiología , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Memoria/fisiología , Odorantes , Animales , Femenino , Masculino , Ratas , Ratas Wistar , Factores Sexuales , Trastornos por Estrés Postraumático/metabolismo
7.
Artículo en Inglés | MEDLINE | ID: mdl-29643800

RESUMEN

Spatial memory processing requires functional interaction between the hippocampus and the medial entorhinal cortex (MEC). The grid cells of the MEC are most abundant in layer II and rely on a complex network of local inhibitory interneurons to generate spatial firing properties. Stress can cause spatial memory deficits in males, but the specific underlying mechanisms affecting the known memory pathways remain unclear. Stress activates both the autonomic nervous system and the hypothalamic-pituitary-adrenal axis to release norepinephrine (NE) and glucocorticoids, respectively. Given that adrenergic receptor (AR) and glucocorticoid receptor (GR) expression is abundant in the MEC, both glucocorticoids and NE released in response to stress may have rapid effects on MEC-LII networks. We used whole-cell patch clamp electrophysiology in MEC slice preparations from male mice to test the effects of NE and glucocorticoids on inhibitory synaptic inputs of MEC-LII principal cells. Application of NE (100 µM) increased the frequency and amplitude of spontaneous inhibitory post-synaptic currents (sIPSCs) in approximately 75% of the principal cells tested. Unlike NE, bath application of dexamethasone (Dex, 1 µM), a synthetic glucocorticoid, or corticosterone (1 µM) the glucocorticoid in rodents, rapidly decreased the frequency of sIPSCs, but not miniature (mIPSCs) in MEC-LII principal cells. Interestingly, pre-treatment with Dex prior to NE application led to an NE-induced increase in sIPSC frequency in all cells tested. This effect was mediated by the α1-AR, as application of an α1-AR agonist, phenylephrine (PHE) yielded the same results, suggesting that a subset of cells in MEC-LII are unresponsive to α1-AR activation without prior activation of GR. We conclude that activation of GRs primes a subset of principal cells that were previously insensitive to NE to become responsive to α1-AR activation in a transcription-independent manner. These findings demonstrate the ability of stress hormones to markedly alter inhibitory signaling within MEC-LII circuits and suggest the intriguing possibility of modulation of network processing upstream of the hippocampus.

8.
Hippocampus ; 27(9): 1016-1029, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28599071

RESUMEN

Post-traumatic stress disorder (PTSD) is characterized by memory disturbances following trauma. Acute predator threat has emerged as an ethological model of PTSD, yet the effects of predator odor on signaling cascades associated with long-term memory remain poorly understood. In this study, we exposed male and female Wistar rats to the synthetic predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT) to assess behavioral and physiological responses as well as rapid modulation of signal transduction cascades associated with learning and memory in the male and female hippocampus. During exposure to TMT in the homecage, both male and female animals displayed robust immobility, avoidance, and altered activity as a function of time. Physiologically, TMT exposure increased circulating corticosterone and blood glucose in both male and female rodents, suggesting that TMT evokes sex-independent behavioral and physiological responses. With respect to signal transduction, TMT exposure rapidly reduced phosphorylation of cyclic-adenosine monophosphate response element binding protein (CREB) in the male, but not the female hippocampus. Furthermore, TMT exposure reduced phosphorylation of extracellular signal-regulated kinase 1/2 and increased nuclear expression of the synapto-nuclear messenger protein Jacob in the male hippocampus, consistent with activation of the CREB shut-off pathway. In a follow-up behavioral experiment, post-training exposure to TMT did not affect spatial water maze performance of male rats. However, male rats re-introduced to the context in which TMT had previously been presented displayed avoidance and hyperactivity, but not freezing behavior or elevated corticosterone responses, suggesting that TMT exposure supports a form of contextual conditioning which is not characterized by immobility. Taken together, our findings suggest that TMT evokes similar behavioral and physiological responses in male and female Wistar rats, but affects distinct signaling cascades in the male and female hippocampus which may contribute to behavioral disruptions associated with predator exposure.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Miedo/psicología , Hipocampo/metabolismo , Odorantes , Trastornos por Estrés Postraumático/metabolismo , Animales , Glucemia/efectos de los fármacos , Corticosterona/sangre , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Pérdida de Tono Postural/efectos de los fármacos , Pérdida de Tono Postural/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Fosforilación/fisiología , Ratas , Ratas Wistar , Factores Sexuales , Trastornos por Estrés Postraumático/inducido químicamente , Tiazoles/administración & dosificación
9.
J Physiol ; 594(13): 3729-44, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27146976

RESUMEN

KEY POINTS: Synaptic excitation and inhibition must be properly balanced in individual neurons and neuronal networks to allow proper brain function. Disrupting this balance may lead to autism spectral disorders and epilepsy. We show the basic helix-loop-helix transcription factor NeuroD2 promotes inhibitory synaptic drive but also decreases cell-intrinsic neuronal excitability of cortical pyramidal neurons both in vitro and in vivo. We identify two genes potentially downstream of NeuroD2-mediated transcription that regulate these parameters: gastrin-releasing peptide and the small conductance, calcium-activated potassium channel, SK2. Our results reveal an important function for NeuroD2 in balancing synaptic neurotransmission and intrinsic excitability. Our results offer insight into how synaptic innervation and intrinsic excitability are coordinated during cortical development. ABSTRACT: Synaptic excitation and inhibition must be properly balanced in individual neurons and neuronal networks for proper brain function. Disruption of this balance during development may lead to autism spectral disorders and epilepsy. Synaptic excitation is counterbalanced by synaptic inhibition but also by attenuation of cell-intrinsic neuronal excitability. To maintain proper excitation levels during development, neurons must sense activity over time and regulate the expression of genes that control these parameters. While this is a critical process, little is known about the transcription factors involved in coordinating gene expression to control excitatory/inhibitory synaptic balance. We show here that the basic helix-loop-helix transcription factor NeuroD2 promotes inhibitory synaptic drive but also decreases cell-intrinsic neuronal excitability of cortical pyramidal neurons both in vitro and in vivo as shown by ex vivo analysis of a NeuroD2 knockout mouse. Using microarray analysis and comparing wild-type and NeuroD2 knockout cortical networks, we identified two potential gene targets of NeuroD2 that contribute to these processes: gastrin-releasing peptide (GRP) and the small conductance, calcium-activated potassium channel, SK2. We found that the GRP receptor antagonist RC-3059 and the SK2 specific blocker apamin partially reversed the effects of increased NeuroD2 expression on inhibitory synaptic drive and action potential repolarization, respectively. Our results reveal an important function for NeuroD2 in balancing synaptic neurotransmission and intrinsic excitability and offer insight into how these processes are coordinated during cortical development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Neuropéptidos/fisiología , Células Piramidales/fisiología , Corteza Somatosensorial/fisiología , Sinapsis/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Cultivadas , Péptido Liberador de Gastrina/genética , Potenciales Postsinápticos Inhibidores , Ratones Noqueados , Neuropéptidos/genética , Ratas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética
10.
Development ; 142(14): 2521-32, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26138475

RESUMEN

In humans, atrial fibrillation is often triggered by ectopic pacemaking activity in the myocardium sleeves of the pulmonary vein (PV) and systemic venous return. The genetic programs that abnormally reinforce pacemaker properties at these sites and how this relates to normal sinoatrial node (SAN) development remain uncharacterized. It was noted previously that Nkx2-5, which is expressed in the PV myocardium and reinforces a chamber-like myocardial identity in the PV, is lacking in the SAN. Here we present evidence that in mice Shox2 antagonizes the transcriptional output of Nkx2-5 in the PV myocardium and in a functional Nkx2-5(+) domain within the SAN to determine cell fate. Shox2 deletion in the Nkx2-5(+) domain of the SAN caused sick sinus syndrome, associated with the loss of the pacemaker program. Explanted Shox2(+) cells from the embryonic PV myocardium exhibited pacemaker characteristics including node-like electrophysiological properties and the capability to pace surrounding Shox2(-) cells. Shox2 deletion led to Hcn4 ablation in the developing PV myocardium. Nkx2-5 hypomorphism rescued the requirement for Shox2 for the expression of genes essential for SAN development in Shox2 mutants. Similarly, the pacemaker-like phenotype induced in the PV myocardium in Nkx2-5 hypomorphs reverted back to a working myocardial phenotype when Shox2 was simultaneously deleted. A similar mechanism is also adopted in differentiated embryoid bodies. We found that Shox2 interacts with Nkx2-5 directly, and discovered a substantial genome-wide co-occupancy of Shox2, Nkx2-5 and Tbx5, further supporting a pivotal role for Shox2 in the core myogenic program orchestrating venous pole and pacemaker development.


Asunto(s)
Proteínas de Homeodominio/fisiología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Venas Pulmonares/metabolismo , Nodo Sinoatrial/metabolismo , Factores de Transcripción/fisiología , Animales , Relojes Biológicos , Diferenciación Celular , Linaje de la Célula , Separación Celular , Electrocardiografía , Cuerpos Embrioides/metabolismo , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Genoma , Corazón/embriología , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Transgénicos , Fenotipo , Estructura Terciaria de Proteína , Proteínas de Dominio T Box/metabolismo
12.
Neurobiol Learn Mem ; 118: 167-77, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25555360

RESUMEN

Females experience depression, posttraumatic stress disorder (PTSD), and anxiety disorders at approximately twice the rate of males, but the mechanisms underlying this difference remain undefined. The effect of sex hormones on neural substrates presents a possible mechanism. We investigated the effect of ovariectomy at two ages, before puberty and in adulthood, and 17ß-estradiol (E2) replacement administered chronically in drinking water on anxiety level, fear memory formation, and extinction. Based on previous studies, we hypothesized that estradiol replacement would impair fear memory formation and enhance extinction rate. Females, age 4 weeks and 10 weeks, were divided randomly into 4 groups; sham surgery, OVX, OVX+low E2 (200nM), and OVX+high E2 (1000nM). Chronic treatment with high levels of E2 significantly increased anxiety levels measured in the elevated plus maze. In both age groups, high levels of E2 significantly increased contextual fear memory but had no effect on cued fear memory. In addition, high E2 decreased the rate of extinction in both ages. Finally, catechol-O-methyltransferase (COMT) is important for regulation of catecholamine levels, which play a role in fear memory formation and extinction. COMT expression in the hippocampus was significantly reduced by high E2 replacement, implying increased catecholamine levels in the hippocampus of high E2 mice. These results suggest that estradiol enhanced fear memory formation, and inhibited fear memory extinction, possibly stabilizing the fear memory in female mice. This study has implications for a neurobiological mechanism for PTSD and anxiety disorders.


Asunto(s)
Ansiedad/fisiopatología , Catecol O-Metiltransferasa/metabolismo , Estradiol/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Memoria/fisiología , Animales , Estradiol/administración & dosificación , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Memoria/efectos de los fármacos , Ratones , Ovariectomía , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo
13.
J Biol Chem ; 290(4): 2007-23, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25488669

RESUMEN

The atrioventricular (AV) junction plays a critical role in chamber septation and transmission of cardiac conduction pulses. It consists of structures that develop from embryonic dorsal mesenchymal protrusion (DMP) and the embryonic AV canal. Despite extensive studies on AV junction development, the genetic regulation of DMP development remains poorly understood. In this study we present evidence that Shox2 is expressed in the developing DMP. Intriguingly, this Shox2-expressing domain possesses a pacemaker-specific genetic profile including Hcn4 and Tbx3. This genetic profile leads to nodal-like electrophysiological properties, which is gradually silenced as the AV node becomes matured. Phenotypic analyses of Shox2(-/-) mice revealed a hypoplastic and defectively differentiated DMP, likely attributed to increased apoptosis, accompanied by dramatically reduced expression of Bmp4 and Hcn4, ectopic activation of Cx40, and an aberrant pattern of action potentials. Interestingly, conditional deletion of Bmp4 or inhibition of BMP signaling by overexpression of Noggin using a Shox2-Cre allele led to a similar DMP hypoplasia and down-regulation of Hcn4, whereas activation of a transgenic Bmp4 allele in Shox2(-/-) background attenuated DMP defects. Moreover, the lack of Hcn4 expression in the DMP of mice carrying Smad4 conditional deletion and direct binding of pSmad1/5/8 to the Hcn4 regulatory region further confirm the Shox2-BMP genetic cascade in the regulation of DMP development. Our results reveal that Shox2 regulates DMP fate and development by controlling BMP signaling through the Smad-dependent pathway to drive tissue growth and to induce Hcn4 expression and suggest a temporal pacemaking function for the DMP during early cardiogenesis.


Asunto(s)
Relojes Biológicos , Proteínas Morfogenéticas Óseas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Proteínas de Homeodominio/metabolismo , Potenciales de Acción , Alelos , Animales , Apoptosis , Proteína Morfogenética Ósea 4/genética , Diferenciación Celular , Electrofisiología , Femenino , Tabiques Cardíacos/embriología , Proteínas de Homeodominio/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Fenotipo , Transducción de Señal
14.
Endocrinology ; 155(8): 2942-52, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24693964

RESUMEN

Evidence suggests that when presented with novel acute stress, animals previously exposed to chronic homotypic or heterotypic stressors exhibit normal or enhanced hypothalamic-pituitary-adrenal (HPA) response compared with animals exposed solely to that acute stressor. The molecular mechanisms involved in this effect remain unknown. The extracellular signal-regulated kinase (ERK) is one of the key pathways regulated in the hippocampus in both acute and chronic stress. The aim of this study was to examine the interaction of prior chronic stress, using the chronic variable stress model (CVS), with exposure to a novel acute stressor (2,5-dihydro-2,4,5-trimethyl thiazoline; TMT) on ERK activation, expression of the downstream protein BCL-2, and the glucocorticoid receptor co-chaperone BAG-1 in control and chronically stressed male rats. TMT exposure after chronic stress resulted in a significant interaction of chronic and acute stress in all 3 hippocampus subregions on ERK activation and BCL-2 expression. Significantly, acute stress increased ERK activation, BCL-2 and BAG-1 protein expression in the dentate gyrus (DG) of CVS-treated rats compared with control, CVS-treated alone, and TMT-only animals. Furthermore, CVS significantly increased ERK activation in medial prefrontal cortex, but acute stress had no significant effect. Inhibition of corticosterone synthesis with metyrapone had no significant effect on ERK activation in the hippocampus; therefore, glucocorticoids alone do not mediate the molecular effects. Finally, because post-translational modifications of histones are believed to play an important role in the stress response, we examined changes in histone acetylation. We found that, in general, chronic stress decreased K12H4 acetylation, whereas acute stress increased acetylation. These results indicate a molecular mechanism by which chronic stress-induced HPA axis plasticity can lead to neurochemical alterations in the hippocampus that influence reactivity to subsequent stress exposure. This may represent an important site of dysfunction that contributes to stress-induced pathology such as depression, anxiety disorders, and posttraumatic stress disorder.


Asunto(s)
Giro Dentado/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema de Señalización de MAP Quinasas , Sistema Hipófiso-Suprarrenal/metabolismo , Estrés Psicológico/metabolismo , Acetilación , Acetiltransferasas/metabolismo , Animales , Histonas/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Distribución Aleatoria , Ratas , Ratas Wistar , Estrés Fisiológico , Tiazoles
15.
Front Cell Neurosci ; 8: 451, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25628536

RESUMEN

Memory acquisition and synaptic plasticity are accompanied by changes in the intrinsic excitability of CA1 pyramidal neurons. These activity-dependent changes in excitability are mediated by modulation of intrinsic currents which alters the responsiveness of the cell to synaptic inputs. The afterhyperpolarization (AHP), a major contributor to the regulation of neuronal excitability, is reduced in animals that have acquired several types of hippocampus-dependent memory tasks and also following synaptic potentiation by high frequency stimulation. BK channels underlie the fast AHP and contribute to spike repolarization, and this AHP is reduced in animals that successfully acquired trace-eyeblink conditioning. This suggests that BK channel function is activity-dependent, but the mechanisms are unknown. In this study, we found that blockade of BK channels with paxilline (10 µM) decreased I AHP amplitude and increased spike half-width and instantaneous frequency in response to a +100 pA depolarization. In addition, induction of long term potentiation (LTP) by theta burst stimulation (TBS) in CA1 pyramidal neurons reduced BK channel's contribution to I AHP, spike repolarization, and instantaneous frequency. This result indicates that BK channel activity is decreased following synaptic potentiation. Interestingly, blockade of mammalian target of rapamycin (MTORC1) with rapamycin (400 nM) following synaptic potentiation restored BK channel function, suggesting a role for protein translation in signaling events which decreased postsynaptic BK channel activity following synaptic potentiation.

16.
Learn Mem ; 20(12): 686-94, 2013 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-24248121

RESUMEN

Downstream regulatory element antagonist modulator (DREAM)/calsenilin(C)/K⁺ channel interacting protein 3 (KChIP3) is a multifunctional Ca²âº-binding protein highly expressed in the hippocampus that inhibits hippocampus-sensitive memory and synaptic plasticity in male mice. Initial studies in our lab suggested opposing effects of DR/C/K3 expression in female mice. Fluctuating hormones that occur during the estrous cycle may affect these results. In this study, we hypothesized that DR/C/K3 interacts with 17ß-estradiol, the primary estrogen produced by the ovaries, to play a role in hippocampus function. We investigated the role of estradiol and DR/C/K3 in learning strategy in ovariectomized (OVX) female mice. OVX WT and DR/C/K3 knockout (KO) mice were given three injections of vehicle (sesame oil) or 17ß-estradiol benzoate (0.25 mg in 100 mL sesame oil) 48, 24, and 2 h before training and testing. DR/C/K3 and estradiol had a time-dependent effect on strategy use in the female mice. Male KO mice exhibited enhanced place strategy relative to WT 24 h after pre-exposure. Fear memory formation was significantly reduced in intact female KO mice relative to intact WT mice, and OVX reduced fear memory formation in the WT, but had no effect in the KO mice. Long-term potentiation in hippocampus slices from female mice was enhanced by circulating ovarian hormones in both WT and DR/C/K3 KO mice. Paired-pulse depression was not affected by ovarian hormones but was reduced in DR/C/K3 KO mice. These results provide the first evidence that DR/C/K3 plays a timing-dependent role in estradiol regulation of learning, memory, and plasticity.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Anticonceptivos/farmacología , Estradiol/análogos & derivados , Proteínas de Interacción con los Canales Kv/metabolismo , Proteínas Represoras/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Estradiol/farmacología , Miedo/efectos de los fármacos , Miedo/fisiología , Femenino , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Proteínas de Interacción con los Canales Kv/deficiencia , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Ovariectomía , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Proteínas Represoras/deficiencia , Factores de Tiempo
17.
Biol Psychiatry ; 74(12): 927-35, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24011821

RESUMEN

BACKGROUND: Exposure to chronic stress produces negative effects on mood and hippocampus-dependent memory formation. Alterations in signaling cascades and histone acetylation present a mechanism of modulation of transcription that may underlie stress-dependent processes in the hippocampus critical to learning and memory and development of depressive behaviors. METHODS: The rat model of chronic variable stress (CVS) was used to investigate the role of changes in protein acetylation and other molecular components of hippocampus-dependent memory formation and anhedonic behavior in response to CVS. RESULTS: Chronic variable stress treatment decreased both extracellular signal-regulated protein kinases 1 and 2 activation and Bcl-2 expression in all three regions of the hippocampus that corresponded behaviorally with a decrease in memory for the novel object location task and increased anhedonia. Extracellular signal-regulated protein kinases 1 and 2 activation was not significantly affected in the amygdala and increased in the medial prefrontal cortex by CVS. Chronic variable stress had no significant effect on activation of Akt in the hippocampus. We investigated molecular and behavioral effects of infusion of the sirtuin inhibitor, sirtinol, into the dentate gyrus (DG). Sirtinol infusion into the DG prevented the CVS-mediated decrease in extracellular signal-regulated protein kinases 1 and 2 activity and Bcl-2 expression, as well as histone acetylation in the DG previously observed following CVS. This corresponded to enhanced performance on the novel object location memory task, as well as reduced anhedonic behavior. CONCLUSIONS: These results suggest that changes in sirtuin activity contribute to changes in molecular cascades and histone acetylation within the hippocampus observed following CVS and may represent a novel therapeutic target for stress-induced depression.


Asunto(s)
Giro Dentado/metabolismo , Hipocampo/metabolismo , Sirtuina 1/metabolismo , Estrés Psicológico/patología , Animales , Inmunoprecipitación de Cromatina , Enfermedad Crónica , Corticosterona/sangre , Modelos Animales de Enfermedad , Preferencias Alimentarias/fisiología , Masculino , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Estrés Psicológico/sangre , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación
18.
Physiol Behav ; 105(5): 1168-74, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22226989

RESUMEN

Recent research investigating Pavlovian fear conditioning and fear extinction has elucidated the neurocircuitry involved in acquisition and inhibition of fear responses. Modulatory factors that may underlie individual differences in fear acquisition and inhibition, however, are not well understood. Testosterone is known to affect anxiety-like behavior and cognitive processing. In this study, we hypothesized that castration would increase anxiety and reduce memory for contextual fear conditioning in an age-dependent manner. In addition, castration would reduce the rate of extinction to context, as high levels of testosterone correlate with reduced PTSD-like symptoms. We compared behaviors in male mice that were castrated at one of two different time points, either before puberty (at 4 weeks) or after puberty (at 10 weeks) to sham-operated control mice. The behaviors investigated included: anxiety, cued and contextual fear conditioning, and extinction of the fear memory. An interaction of hormone status and age and a significant effect of age were measured in the elevated plus maze, a measure of anxiety. Castration caused a significant reduction of contextual fear memory, but no effect on cued fear memory. There was no significant effect of castration on extinction. Interestingly, a significant effect of age of the mouse at the time of testing was observed on extinction. These results suggest that endogenous androgens during puberty are important for anxiety and fear memory formation. In addition, these results define a late post-adolescent developmental time point for changes in anxiety and fear extinction.


Asunto(s)
Ansiedad/fisiopatología , Condicionamiento Clásico/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Testosterona/fisiología , Factores de Edad , Análisis de Varianza , Animales , Aprendizaje por Asociación/fisiología , Conducta Animal/fisiología , Castración , Inhibición Psicológica , Masculino , Memoria/fisiología , Ratones , Ratones de la Cepa 129 , Factores Sexuales , Maduración Sexual/fisiología
19.
J Physiol ; 589(Pt 14): 3517-32, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21606111

RESUMEN

The dentate gyrus of the hippocampus is thought to control information flow into the rest of the hippocampus. Under pathological conditions, such as epilepsy, this protective feature is circumvented and uninhibited activity flows throughout the hippocampus. Many factors can modulate excitability of the dentate gyrus and ultimately, the hippocampus. It is therefore of critical importance to understand the mechanisms involved in regulating excitability in the dentate gyrus. Dynorphin, the endogenous ligand for the kappa (κ) opioid receptor (KOR), is thought to be involved in neuromodulation in the dentate gyrus. Both dynorphin and its receptor are widely expressed in the dentate gyrus and have been implicated in epilepsy and other complex behaviours such as stress-induced deficits in learning and stress-induced depression-like behaviours. Administration of KOR agonists can prevent both the behavioural and electroencephalographic measures of seizures in several different models of epilepsy. Antagonism of the KORs also prevents stress-induced behaviours. This evidence suggests the KORs as possible therapeutic targets for various pathological conditions. In addition, KOR agonists prevent the induction of LTP. Although there are several mechanisms through which dynorphin could mediate these effects, no studies to date investigated the effects of KOR activation on intrinsic membrane properties and cell excitability. We used whole-cell, patch-clamp recordings from acute mouse hippocampus slices to investigate the effect of KOR activation on dentate gyrus granule cell excitability. The agonist U69,593 (U6, 1 µM) resulted in a lower spike threshold, a decreased latency to first spike, an increased spike half-width, and an overall increase in spike number with current injections ranging from 15 to 45 pA. There was also a reduction in the interspike interval (ISI) both early and late in the spike train, with no change in membrane potential or input resistance. Preincubation of the slice with the selective KOR antagonist, nor-binalthorphimine (BNI, 1 µM) inhibited the effect of U6 on the latency to first spike and spike half-width suggesting that these effects are mediated through KORs. The inclusion of GDP-ßS (1 mM) in the recording pipette prevented all of the U6 effects, suggesting that all effects are mediated via a G-protein-dependent mechanism. Inclusion of the A-type K+ current blocker, 4-aminopyridine (4-AP, 5 mM) in the pipette also antagonised the effects of U6. Kv4.2 is one of the channel α subunits thought to be responsible for carrying the A-type K+ current. Incubation of hippocampus slices with U6 resulted in a decrease in the Kv4.2 subunit protein at the cell surface. These results are consistent with an increase in cell excitability in response to KOR activation and may reflect new possibilities for additional opioid functions.


Asunto(s)
Giro Dentado/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores Opioides kappa/metabolismo , 4-Aminopiridina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Bencenoacetamidas/farmacología , Giro Dentado/metabolismo , Dinorfinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Masculino , Ratones , Ratones de la Cepa 129 , Naltrexona/análogos & derivados , Naltrexona/farmacología , Neuronas/metabolismo , Técnicas de Placa-Clamp/métodos , Canales de Potasio/metabolismo , Pirrolidinas/farmacología , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Tionucleótidos/farmacología
20.
Neurosci Lett ; 489(3): 154-8, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21146591

RESUMEN

Corticosterone (CORT) release from the adrenal glands in response to acutely stressful stimuli is well-characterized, however several non-experimental, environmental stressors can also engender a CORT response. The aim of this study was to investigate an acute activation of the HPA axis in pair-housed animals in response to separation. We observed a rapid significant increase in CORT in the animal remaining in the home cage following cage mate removal, that was not caused by cage opening and transient removal of cage mate. In addition, we examined this response in both control, non-stressed animals and in animals subjected to chronic variable stress (CVS) and found that although basal levels of CORT differed between control and CVS animals, there was no significant difference in the acute CORT levels between the control and CVS animals after separation, indicating that this environmental event is perceived as acutely stressful in both conditions. Furthermore, we examined the time course of CORT activation and found that CORT levels rapidly rise within minutes of separation peaking at 15 min and returning to baseline by 90 min. The results of this study demonstrate that separation can induce an acute stress response in the remaining cage mate measured by increased CORT and should be considered in molecular, behavioral, and electrophysiological studies.


Asunto(s)
Corticosterona/sangre , Aislamiento Social , Estrés Psicológico/sangre , Animales , Modelos Animales de Enfermedad , Vivienda para Animales , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...