Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Circulation ; 144(23): 1856-1875, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34694145

RESUMEN

BACKGROUND: Vascular homeostasis is maintained by the differentiated phenotype of vascular smooth muscle cells (VSMCs). The landscape of protein coding genes comprising the transcriptome of differentiated VSMCs has been intensively investigated but many gaps remain including the emerging roles of noncoding genes. METHODS: We reanalyzed large-scale, publicly available bulk and single-cell RNA sequencing datasets from multiple tissues and cell types to identify VSMC-enriched long noncoding RNAs. The in vivo expression pattern of a novel smooth muscle cell (SMC)-expressed long noncoding RNA, Carmn (cardiac mesoderm enhancer-associated noncoding RNA), was investigated using a novel Carmn green fluorescent protein knock-in reporter mouse model. Bioinformatics and quantitative real-time polymerase chain reaction analysis were used to assess CARMN expression changes during VSMC phenotypic modulation in human and murine vascular disease models. In vitro, functional assays were performed by knocking down CARMN with antisense oligonucleotides and overexpressing Carmn by adenovirus in human coronary artery SMCs. Carotid artery injury was performed in SMC-specific Carmn knockout mice to assess neointima formation and the therapeutic potential of reversing CARMN loss was tested in a rat carotid artery balloon injury model. The molecular mechanisms underlying CARMN function were investigated using RNA pull-down, RNA immunoprecipitation, and luciferase reporter assays. RESULTS: We identified CARMN, which was initially annotated as the host gene of the MIR143/145 cluster and recently reported to play a role in cardiac differentiation, as a highly abundant and conserved, SMC-specific long noncoding RNA. Analysis of the Carmn GFP knock-in mouse model confirmed that Carmn is transiently expressed in embryonic cardiomyocytes and thereafter becomes restricted to SMCs. We also found that Carmn is transcribed independently of Mir143/145. CARMN expression is dramatically decreased by vascular disease in humans and murine models and regulates the contractile phenotype of VSMCs in vitro. In vivo, SMC-specific deletion of Carmn significantly exacerbated, whereas overexpression of Carmn markedly attenuated, injury-induced neointima formation in mouse and rat, respectively. Mechanistically, we found that Carmn physically binds to the key transcriptional cofactor myocardin, facilitating its activity and thereby maintaining the contractile phenotype of VSMCs. CONCLUSIONS: CARMN is an evolutionarily conserved SMC-specific long noncoding RNA with a previously unappreciated role in maintaining the contractile phenotype of VSMCs and is the first noncoding RNA discovered to interact with myocardin.


Asunto(s)
Contracción Muscular , Músculo Liso Vascular/metabolismo , Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/metabolismo , Transactivadores/metabolismo , Animales , Humanos , Ratones , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , Ratas , Transactivadores/genética
2.
Neurobiol Learn Mem ; 187: 107562, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34848328

RESUMEN

The neurobiology of emotion and episodic memory are well-researched subjects, as is their intersection: memory of emotional events (i.e. emotional memory). We and others have previously demonstrated that the emotional valence of stimuli is encoded in the dorsal hippocampus, a structure integral to the acquisition, consolidation and retrieval of long-term episodic memories. Such findings are consistent with the idea that the emotional valence of stimuli contributes to the "what" component of episodic memories ("where" and "when" being the other components). We hypothesized that being in a heightened emotional state by itself does not contribute to the "what" component of episodic memories. We tested an inference of this hypothesis - that negative emotional state does not alter re-encoding of a spatial episodic event. Rats from the experimental group explored a novel place at their baseline emotional state (Event 1) and 20 min later re-explored the same place (Event 2) in a negative emotional state induced by a state-altering event prior to Event 2. We examined neuronal ensembles that induced expression of Arc and Homer1a, two immediate-early genes (IEGs) necessary for synaptic plasticity and consolidation of long-term memories, during both events. We found that in dorsal CA1 and dorsal CA3, Event 1 and Event 2 induced IEG expression in different neuronal ensembles. This finding was reflected in a low Fidelity score, which assesses the percentage of the Event 1 IEG-expressing ensemble re-activated during Event 2. The Fidelity score was significantly higher in a control group which was at a baseline emotional state during Event 2. Groups which were matched for non-specific disruptions from the state-altering event had intermediate Fidelity scores in dorsal CA1. The Fidelity scores of the dorsal CA3 in the latter groups were similar to those of the control group. Combined, the findings reject the tested hypothesis and suggest that a negative emotional state is encoded in the hippocampus as part of the long-term memory of episodic events that lack explicit emotion-inducing stimuli. These findings also suggest that individuals who often experience strong negative emotional states incorporate these states into ongoing non-emotional episodic memories.


Asunto(s)
Emociones/fisiología , Hipocampo/fisiología , Memoria Episódica , Memoria a Largo Plazo/fisiología , Plasticidad Neuronal , Animales , Genes Inmediatos-Precoces , Masculino , Neuronas , Ratas
3.
Mol Psychiatry ; 26(11): 6666-6679, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33859360

RESUMEN

A precise fear memory encoding a traumatic event enables an individual to avoid danger and identify safety. An impaired fear memory (contextual amnesia), however, puts the individual at risk of developing posttraumatic stress disorder (PTSD) due to the inability to identify a safe context when encountering trauma-associated cues later in life. Although it is gaining attention that contextual amnesia is a critical etiologic factor for PTSD, there is no treatment currently available that can reverse contextual amnesia, and whether such treatment can prevent the development of PTSD is unknown. Here, we report that (I) a single dose of transcranial photobiomodulation (PBM) applied immediately after tone fear conditioning can reverse contextual amnesia. PBM treatment preserved an appropriately high level of contextual fear memory in rats revisiting the "dangerous" context, while control rats displayed memory impairment. (II) A single dose of PBM applied after memory recall can reduce contextual fear during both contextual and cued memory testing. (III) In a model of complex PTSD with repeated trauma, rats given early PBM interventions efficiently discriminated safety from danger during cued memory testing and, importantly, these rats did not develop PTSD-like symptoms and comorbidities. (IV) Finally, we report that fear extinction was facilitated when PBM was applied in the early intervention window of memory consolidation. Our results demonstrate that PBM treatment applied immediately after a traumatic event or its memory recall can protect contextual fear memory and prevent the development of PTSD-like psychopathological fear in rats.


Asunto(s)
Trastornos por Estrés Postraumático , Animales , Extinción Psicológica , Miedo , Memoria , Trastornos de la Memoria/etiología , Ratas , Trastornos por Estrés Postraumático/patología
4.
Proc Natl Acad Sci U S A ; 115(37): E8660-E8667, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30139920

RESUMEN

In response to vascular injury, vascular smooth muscle cells (VSMCs) may switch from a contractile to a proliferative phenotype thereby contributing to neointima formation. Previous studies showed that the long noncoding RNA (lncRNA) NEAT1 is critical for paraspeckle formation and tumorigenesis by promoting cell proliferation and migration. However, the role of NEAT1 in VSMC phenotypic modulation is unknown. Herein we showed that NEAT1 expression was induced in VSMCs during phenotypic switching in vivo and in vitro. Silencing NEAT1 in VSMCs resulted in enhanced expression of SM-specific genes while attenuating VSMC proliferation and migration. Conversely, overexpression of NEAT1 in VSMCs had opposite effects. These in vitro findings were further supported by in vivo studies in which NEAT1 knockout mice exhibited significantly decreased neointima formation following vascular injury, due to attenuated VSMC proliferation. Mechanistic studies demonstrated that NEAT1 sequesters the key chromatin modifier WDR5 (WD Repeat Domain 5) from SM-specific gene loci, thereby initiating an epigenetic "off" state, resulting in down-regulation of SM-specific gene expression. Taken together, we demonstrated an unexpected role of the lncRNA NEAT1 in regulating phenotypic switching by repressing SM-contractile gene expression through an epigenetic regulatory mechanism. Our data suggest that NEAT1 is a therapeutic target for treating occlusive vascular diseases.


Asunto(s)
Regulación de la Expresión Génica , Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/genética , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Neointima/genética , Neointima/metabolismo , Fenotipo , Interferencia de ARN , Ratas , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
5.
Int J Mol Sci ; 20(16)2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31443163

RESUMEN

Retinal neurodegeneration, an early characteristic of several blinding diseases, triggers glial activation, resulting in inflammation, secondary damage and visual impairment. Treatments that aim only at neuroprotection have failed clinically. Here, we examine the impact of modulating thioredoxin interacting protein (TXNIP) to the inflammatory secondary damage and visual impairment in a model of ischemia/reperfusion (IR). Wild type (WT) and TXNIP knockout (TKO) mice underwent IR injury by increasing intraocular pressure for 40 min, followed by reperfusion. An additional group of WT mice received intravitreal TXNIP-antisense oligomers (ASO, 100 µg/2 µL) 2 days post IR injury. Activation of Müller glial cells, apoptosis and expression of inflammasome markers and visual function were assessed. IR injury triggered early TXNIP mRNA expression that persisted for 14 days and was localized within activated Müller cells in WT-IR, compared to sham controls. Exposure of Müller cells to hypoxia-reoxygenation injury triggered endoplasmic reticulum (ER) stress markers and inflammasome activation in WT cells, but not from TKO cells. Secondary damage was evident by the significant increase in the number of occluded acellular capillaries and visual impairment in IR-WT mice but not in IR-TKO. Intervention with TXNIP-ASO prevented ischemia-induced glial activation and neuro-vascular degeneration, and improved visual function compared to untreated WT. Targeting TXNIP expression may offer an effective approach in the prevention of secondary damage associated with retinal neurodegenerative diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Daño por Reperfusión/metabolismo , Tiorredoxinas/metabolismo , Animales , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Gliosis/metabolismo , Hipoxia/metabolismo , Inflamasomas/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Daño por Reperfusión/genética , Tiorredoxinas/genética
6.
Hippocampus ; 27(3): 274-284, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28121049

RESUMEN

Evidence suggests that the memory of a recently ingested meal limits subsequent intake. Given that ventral hippocampal (vHC) neurons are involved in memory and energy intake, the present experiment tested the hypothesis that vHC neurons contribute to the formation of a memory of a meal and inhibit energy intake during the postprandial period. We tested (1) whether pharmacological inactivation of vHC neurons during the period following a sucrose meal, when the memory of the meal would be undergoing consolidation, accelerates the onset of the next sucrose meal and increases intake and (2) whether sucrose intake increases vHC expression of the synaptic plasticity marker activity-regulated cytoskeletal-associated protein (Arc). Adult male Sprague-Dawley rats were trained to consume a 32% sucrose solution daily at the same time and location. On the experimental day, the rats were given intra-vHC infusions of the GABAA receptor agonist muscimol or vehicle after they finished their first sucrose meal. Compared to vehicle infusions, postmeal intra-vHC muscimol infusions decreased the latency to the next sucrose meal, increased the amount of sucrose consumed during that meal, increased the total number of sucrose meals and the total amount of sucrose ingested. In addition, rats that consumed sucrose had higher levels of Arc expression in both vHC CA1 and CA3 subfields than cage control rats. Collectively, these findings are the first to show that vHC neurons inhibit energy intake during the postprandial period and support the hypothesis that vHC neurons form a memory of a meal and inhibit subsequent intake. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Ingestión de Energía/fisiología , Conducta Alimentaria/fisiología , Hipocampo/fisiología , Memoria/fisiología , Neuronas/fisiología , Periodo Posprandial/fisiología , Animales , Catéteres de Permanencia , Proteínas del Citoesqueleto/metabolismo , Sacarosa en la Dieta , Agonistas de Receptores de GABA-A/farmacología , Hipocampo/efectos de los fármacos , Masculino , Muscimol/farmacología , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley
7.
Eur J Neurosci ; 46(2): 1779-1789, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28544049

RESUMEN

Learning to associate a stimulus with reinforcement causes plasticity in primary sensory cortex. Neural activity caused by the associated stimulus is paired with reinforcement, but population analyses have not found a selective increase in response to that stimulus. Responses to other stimuli increase as much as, or more than, responses to the associated stimulus. Here, we applied population analysis at a new time point and additionally evaluated whether cholinergic receptor blockers interacted with the plastic changes in cortex. Three days of tone identification behavior caused responsiveness to increase broadly across primary auditory cortex, and target responses strengthened less than overall responsiveness. In pharmacology studies, behaviorally impairing doses of selective acetylcholine receptor blockers were administered during behavior. Neural responses were evaluated on the following day, while the blockers were absent. The muscarinic group, blocked by scopolamine, showed lower responsiveness and an increased response to the tone identification target that exceeded both the 3-day control group and task-naïve controls. Also, a selective increase in the late phase of the response to the tone identification stimulus emerged. Nicotinic receptor antagonism, with mecamylamine, more modestly lowered responses the following day and lowered target responses more than overall responses. Control acute studies demonstrated the muscarinic block did not acutely alter response rates, but the nicotinic block did. These results lead to the hypothesis that the decrease in the proportion of the population spiking response that is selective for the target may be explained by the balance between effects modulated by muscarinic and nicotinic receptors.


Asunto(s)
Corteza Auditiva/metabolismo , Percepción Auditiva/fisiología , Neuronas/metabolismo , Patrones de Reconocimiento Fisiológico/fisiología , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo , Estimulación Acústica , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Corteza Auditiva/efectos de los fármacos , Percepción Auditiva/efectos de los fármacos , Mapeo Encefálico , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Masculino , Mecamilamina/farmacología , Microelectrodos , Antagonistas Muscarínicos/farmacología , Neuronas/efectos de los fármacos , Antagonistas Nicotínicos/farmacología , Patrones de Reconocimiento Fisiológico/efectos de los fármacos , Ratas Sprague-Dawley , Escopolamina/farmacología
8.
J Neurosci ; 35(11): 4571-81, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25788674

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) is clinically targeted for type II diabetes treatment; however, rosiglitazone (ROSI), a PPARγ agonist, increases food intake and body/fat mass as side-effects. Mechanisms for these effects and the role of PPARγ in feeding are not understood. Therefore, we tested this role in Siberian hamsters, a model of human energy balance, and C57BL/6 mice. We tested the following: (1) how ROSI and/or GW9662 (2-chloro-5-nitro-N-phenylbenzamide; PPARγ antagonist) injected intraperitoneally or into the third ventricle (3V) affected Siberian hamster feeding behaviors; (2) whether food deprivation (FD) co-increases agouti-related protein (AgRP) and PPARγ mRNA expression in Siberian hamsters and mice; (3) whether intraperitoneally administered ROSI increases AgRP and NPY in ad libitum-fed animals; (4) whether intraperitoneally administered PPARγ antagonism blocks FD-induced increases in AgRP and NPY; and finally, (5) whether intraperitoneally administered PPARγ modulation affects plasma ghrelin. Third ventricular and intraperitoneally administered ROSI increased food hoarding and intake for 7 d, an effect attenuated by 3V GW9662, and also prevented (intraperitoneal) FD-induced feeding. FD hamsters and mice increased AgRP within the arcuate hypothalamic nucleus with concomitant increases in PPARγ exclusively within AgRP/NPY neurons. ROSI increased AgRP and NPY similarly to FD, and GW9662 prevented FD-induced increases in AgRP and NPY in both species. Neither ROSI nor GW9662 affected plasma ghrelin. Thus, we demonstrated that PPARγ activation is sufficient to trigger food hoarding/intake, increase AgRP/NPY, and possibly is necessary for FD-induced increases in feeding and AgRP/NPY. These findings provide initial evidence that FD-induced increases in AgRP/NPY may be a direct PPARγ-dependent process that controls ingestive behaviors.


Asunto(s)
Proteína Relacionada con Agouti/biosíntesis , Núcleo Arqueado del Hipotálamo/metabolismo , Conducta Alimentaria/fisiología , Neuropéptido Y/biosíntesis , PPAR gamma/biosíntesis , ARN Mensajero/biosíntesis , Animales , Cricetinae , Conducta Alimentaria/psicología , Ratones , Ratones Endogámicos C57BL , Phodopus
9.
Hippocampus ; 26(3): 405-13, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26386270

RESUMEN

There is limited knowledge regarding how the brain controls the timing of meals. Similarly, there is a large gap in our understanding of how top-down cognitive processes, such as memory influence energy intake. We hypothesize that dorsal hippocampal (dHC) neurons, which are critical for episodic memory, form a memory of a meal and inhibit meal onset during the postprandial period. In support, we showed previously that reversible inactivation of these neurons during the period following a sucrose meal accelerates the onset of the next meal. If dHC neurons form a memory of a meal, then consumption should induce synaptic plasticity in dHC neurons. To test this, we determined (1) whether a sucrose meal increases the expression of the synaptic plasticity marker activity-regulated cytoskeleton-associated protein (Arc) in dHC CA1 neurons, (2) whether previous experience with sucrose influences sucrose-induced Arc expression, and (3) whether the orosensory stimulation produced by the noncaloric sweetener saccharin is sufficient to induce Arc expression. Male Sprague-Dawley rats were trained to consume a sweetened solution at a scheduled time daily. On the experimental day, they were given a solution for 7 min, euthanized, and then fluorescence in situ hybridization procedures were used to measure meal-induced Arc mRNA. Compared to caged control rats, Arc expression was significantly higher in rats that consumed sucrose or saccharin. Interestingly, rats given additional experience with sucrose had less Arc expression than rats with less sucrose experience, even though both groups consumed similar amounts on the experimental day. Thus, this study is the first to suggest that orosensory stimulation produced by consuming a sweetened solution and possibly the hedonic value of that sweet stimulation induces synaptic plasticity in dHC CA1 neurons in an experience-dependent manner. Collectively, these findings are consistent with our hypothesis that dHC neurons form a memory of a meal.


Asunto(s)
Complejo Relacionado con el SIDA/metabolismo , Región CA1 Hipocampal/citología , Neuronas/efectos de los fármacos , Edulcorantes/farmacología , Gusto/fisiología , Complejo Relacionado con el SIDA/genética , Vías Aferentes/efectos de los fármacos , Vías Aferentes/fisiología , Análisis de Varianza , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Masculino , Neuronas/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Sacarina/farmacología , Sacarosa/farmacología , Gusto/efectos de los fármacos , Enseñanza , Factores de Tiempo
10.
Neurobiol Learn Mem ; 112: 158-67, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24583372

RESUMEN

Posttraumatic stress disorder (PTSD) is an anxiety disorder that occurs after experiencing a traumatic event. Susceptibility to PTSD exists, as only some trauma-exposed individuals develop this condition. Investigating susceptibilities in animal models can contribute to understanding the etiology of the disorder. We previously reported an animal model which allows reliable pre-classification of rats as susceptible (Sus) or resistant (Res) to developing a PTSD-like phenotype after a later trauma. Here we report that Sus, compared to Res, rats have altered hippocampal function, along the septo-temporal axis, prior to experiencing a traumatic event. In Experiment I, Res and Sus rats explored a novel box twice. Using a cellular imaging method for assessing plasticity-related immediate-early gene expression in large neuronal ensembles, Arc/Homer1a catFISH, we show that Sus rats have smaller vCA3 ensembles during the second exploration. This suppressed vCA3 activation in Sus rats was not due to a difference in exploratory behavior, or to a difference in Arc/Homer1a expression in the basolateral amygdala (BLA). BLA is a main source of inputs to vCA3, but both the ensemble size and overlap of BLA ensembles activated during the two explorations was similar to that of Res rats. Additionally, Sus rats had significant 'infidelity' in their dorsal hippocampal representations of the second event: a lower overlap, compared to Res rats, of Arc/Homer1a-expressing ensembles activated during the two explorations (the size of the ensembles were similar to those of Res rats). These differences were revealed only in conditions of relatively low stress, because they were not observed when Sus and Res rats experienced fear conditioning (Experiment II). Combined, the findings show that altered hippocampal function exists before experiencing emotional trauma in susceptible rats and suggest that this is a risk factor for PTSD.


Asunto(s)
Conducta Animal/fisiología , Genes Inmediatos-Precoces/fisiología , Hipocampo/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Estrés Psicológico/metabolismo , Animales , Complejo Nuclear Basolateral/metabolismo , Conducta Animal/clasificación , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Expresión Génica/fisiología , Masculino , Fenotipo , Ratas , Ratas Sprague-Dawley , Factores de Riesgo , Trastornos por Estrés Postraumático/etiología , Estrés Psicológico/complicaciones
11.
Proc Natl Acad Sci U S A ; 107(33): 14828-32, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20675582

RESUMEN

Models of learning-dependent sensory cortex plasticity require local activity and reinforcement. An alternative proposes that neural activity involved in anticipation of a sensory stimulus, or the preparatory set, can direct plasticity so that changes could occur in regions of sensory cortex lacking activity. To test the necessity of target-induced activity for initial sensory learning, we trained rats to detect a low-frequency sound. After learning, Arc expression and physiologically measured neuroplasticity were strong in a high-frequency auditory cortex region with very weak target-induced activity in control animals. After 14 sessions, Arc and neuroplasticity were aligned with target-induced activity. The temporal and topographic correspondence between Arc and neuroplasticity suggests Arc may be intrinsic to the neuroplasticity underlying perceptual learning. Furthermore, not all neuroplasticity could be explained by activity-dependent models but can be explained if the neural activity involved in the preparatory set directs plasticity.


Asunto(s)
Corteza Auditiva/fisiología , Proteínas del Citoesqueleto/fisiología , Aprendizaje/fisiología , Proteínas del Tejido Nervioso/fisiología , Plasticidad Neuronal/fisiología , Estimulación Acústica , Análisis de Varianza , Animales , Corteza Auditiva/metabolismo , Percepción Auditiva/fisiología , Mapeo Encefálico , Proteínas del Citoesqueleto/genética , Potenciales Evocados Auditivos/fisiología , Expresión Génica , Hibridación Fluorescente in Situ , Masculino , Proteínas del Tejido Nervioso/genética , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/fisiología , Factores de Tiempo
12.
Proc Natl Acad Sci U S A ; 107(3): 1211-6, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20080551

RESUMEN

Neuregulin 1 (NRG1) is a trophic factor thought to play a role in neural development. Recent studies suggest that it may regulate neurotransmission, mechanisms of which remain elusive. Here we show that NRG1, via stimulating GABA release from interneurons, inhibits pyramidal neurons in the prefrontal cortex (PFC). Ablation of the NRG1 receptor ErbB4 in parvalbumin (PV)-positive interneurons prevented NRG1 from stimulating GABA release and from inhibiting pyramidal neurons. PV-ErbB4(-/-) mice exhibited schizophrenia-relevant phenotypes similar to those observed in NRG1 or ErbB4 null mutant mice, including hyperactivity, impaired working memory, and deficit in prepulse inhibition (PPI) that was ameliorated by diazepam, a GABA enhancer. These results indicate that NRG1 regulates the activity of pyramidal neurons by promoting GABA release from PV-positive interneurons, identifying a critical function of NRG1 in balancing brain activity. Because both NRG1 and ErbB4 are susceptibility genes of schizophrenia, our study provides insight into potential pathogenic mechanisms of schizophrenia and suggests that PV-ErbB4(-/-) mice may serve as a model in the study of this and relevant brain disorders.


Asunto(s)
Receptores ErbB/fisiología , Interneuronas/metabolismo , Neurregulina-1/fisiología , Parvalbúminas/metabolismo , Células Piramidales/metabolismo , Animales , Receptores ErbB/genética , Interneuronas/citología , Memoria , Ratones , Ratones Noqueados , Células Piramidales/citología , Receptor ErbB-4 , Ácido gamma-Aminobutírico/metabolismo
13.
Front Behav Neurosci ; 17: 1231563, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37732174

RESUMEN

Introduction: Anxiety and anxiety-influenced disorders are sexually dimorphic with women being disproportionately affected compared to men. Given the increased prevalence in women and the documented differences in anxiety and trauma behavior between male and female rats this paper sought to examine the link between stress, anxiety, and fear learning and extinction in female rats. We tested the hypothesis that a mild stressor will induce short-and long-term increases in anxiety and produce long term effects on subsequent fear learning and extinction behavior. Methods: We induced anxiety in female Sprague- Dawley rats with a short (3 min) exposure to a ball of cat hair infused with 150 µl of cat urine (mild stressor) that elicits innate fear but does not cause fear conditioning. The control group was exposed to fake cat hair. Anxiety was assessed in the Light-Dark Open Field (LDOF) or Elevated Plus Maze (EPM) before, immediately after and 4 days after stimulus exposure. Two weeks later, all animals were subject to Contextual Fear Conditioning (CFC) in the Shock Arm of a Y-maze, blocked off from the rest of the maze. Memory and fear extinction (learning of safety) was assessed in the following four days by placing each rat in one of the Safe Arms and measuring avoidance extinction (time spent and number of entries in the Shock Arm). Results: Cat hair exposure induced changes in anxiety-like behavior in the short-term that appeared resolved 4 days later. However, the cat-hair exposed rats had long-term (2 weeks) phenotypic changes expressed as altered exploratory behavior in an emotionally neutral novel place. Fear learning and extinction were not impaired. Yet, using avoidance extinction, we demonstrated that the phenotypic difference induced by the mild stressor could be documented and dissociated from learning and memory. Discussion: These findings demonstrate that the history of stress, even mild stress, has subtle long-term effects on behavior even when short-term anxiety appears resolved.

14.
Front Behav Neurosci ; 17: 1112837, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064304

RESUMEN

Introduction: Post-Traumatic Stress Disorder (PTSD) is a psychological disorder that occurs after a traumatic event in a subset of exposed individuals. This implies the existence of susceptibility factors that foster the development of PTSD. Susceptibility factors are present before trauma and can contribute to the development and maintenance of PTSD after trauma. Manipulation of susceptibility factors may decrease the probability of developing PTSD. A putative susceptibility factor is inflammation. Patients with PTSD have been documented to have a higher pro-inflammatory profile compared to non-PTSD subjects. In addition, they are more likely to develop and die from cardiovascular disease which has a strong inflammation component. It is not known, however, whether inflammation plays a role in developing PTSD or whether reducing inflammation can prevent PTSD. Methods: We used the Revealing Individual Susceptibility to a PTSD-like phenotype (RISP) model to behaviorally classify male rats as resilient or susceptible before trauma and tested their serum and prefrontal cortical (mPFC) levels of IL-1ß, IL-6, TNFα, IL-10, IFN IFNγ, and KC/GRO to determine whether inflammation represents a putative susceptibility factor for PTSD. Results: We found elevated IL-6 levels in the mPFC, but not serum, of susceptible rats compared to resilient animals before trauma. Serum and mPFC levels were not correlated in any of the cytokines/chemokines. Rats with high anxiety-like behavior had elevated IL-6 and IL-10 mPFC levels. Acoustic startle responses were not associated with cytokine/chemokine levels. Discussion: Neuroinflammation, rather than systemic inflammation exists in susceptible male rats before trauma and is thus a putative susceptibility factor for PTSD. Thus, susceptibility appears neurogenic in its pathogenesis. The lack of differences between susceptible and resilient rats in serum cytokine/chemokine levels infers that peripheral markers will not be useful in determining susceptibility. Chronic neuroinflammation appears more broadly associated with anxiety rather than startle responses.

15.
Neuroscience ; 497: 146-156, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35764190

RESUMEN

Fear memories are important for survival and are implicated in the etiology of fear disorders such as Post Traumatic Stress Disorder (PTSD). Fear memories are well studied pre-clinically and sex differences in rodent fear expression have been reported: females tend to freeze less than males. Whether this is a difference in fear learning or expression is debated. We aimed to differentiate between these possibilities with a task that allowed female rats to express fear memory by moving, rather than freezing. We assessed fear extinction after contextual fear conditioning in the isolated Shock Arm of a Y-maze in female and male rats by either placing them back in the isolated Shock Arm (Fear Extinction in the Shock Context) or allowing them to move freely in the Y-maze during extinction training and enter/avoid the Shock Arm (Avoidance Extinction). We confirmed that female rats freeze less than males during fear extinction in both settings. During Avoidance Extinction, however, both sexes had similar avoidance of the Shock Context, showing comparable fear memory and extinction. Additionally, female rats made more entries into the non-shock arms. Thus, female and male rats have similar fear learning but females express it with an active motor response. Furthermore, female rats also exhibited an active motor response under other anxiogenic conditions (Elevated Plus Maze) and had higher reactivity (Acoustic Startle Response) but not when fear-eliciting stimuli were present: cat hair and foot-shock. In summary, female rats have an active motor response to anxiogenic stimuli which we termed 'Anxioescapic' behavior strategy.


Asunto(s)
Extinción Psicológica , Miedo , Animales , Extinción Psicológica/fisiología , Miedo/fisiología , Femenino , Reacción Cataléptica de Congelación/fisiología , Masculino , Reflejo de Sobresalto/fisiología , Caracteres Sexuales
16.
J Neurosci Methods ; 363: 109325, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34418444

RESUMEN

BACKGROUND: Pre-clinical studies of psychiatric disorders often include a measure of anxiety-like behavior. Several tasks exist that serve this purpose, but because anxiety is complex with a myriad of anxiogenic stimuli, researchers are often compelled to use multiple tasks. The Light-Dark Open Field (LDOF) combines concepts from two such tasks, Light-Dark Box and Open Field, into one task with the synergistic effect of enhanced discrimination of anxiety-like behavior. NEW METHODS: Our goal was to increase the sensitivity of the Open Field task with the addition of a shadow, conceptually similar to the Light-Dark Box, to detect concealed differences even under bright light, which is highly anxiogenic. The resulting LDOF allows assessment of anxiety due to bright light and open space simultaneously, while retaining the ability to assess the impact of each with custom indices. In addition, it maintains all the advantages and measures of the Open Field. RESULTS: Using custom created indices from measures collected in the LDOF one can assess anxiety induced by light, open space, or light and open space combined and thus elucidate anxiety-inducing factors. Using two strains of rats: an outbred strain, Sprague-Dawley (SD), and a strain that exhibits high trait anxiety, Lewis rats, we found that increased discrimination for anxiety-like behavior can be achieved with the Light-Dark Open Field. COMPARISON WITH EXISTING MODELS: The LDOF allows researchers to extract the traditional measures of an Open Field, including valuable information about locomotion and habituation while adding a further layer of discrimination with the light-dark component. Because the LDOF is a combination of two different tests, it saves time compared to running multiple experiments in series that then need to be counterbalanced to reduce artefacts and task order effects. In addition, it detects differences even when traditional tasks of anxiety have reached their ceiling sensitivity (i.e. EPM under bright light conditions). CONCLUSION: We present the Light-Dark Open Field: a simple modification of an existing Open Field apparatus that incorporates aspects of the Light-Dark Box with the addition of a shadow. The shadow (Dark Perimeter) allows for increased discrimination in detecting anxiety-like behaviors. Comparison of anxiety-like behavior between Lewis and SD rats allowed us to develop the construct and face validity of the LDOF as well as demonstrate its sensitivity even under bright light conditions. In addition, we developed 3 indices that allow one to parse out, from one set of data, the effect of two anxiogenic stimuli- bright light and open space.


Asunto(s)
Conducta Animal , Actividad Motora , Animales , Ansiedad , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley
17.
Cell Rep ; 36(5): 109469, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34348147

RESUMEN

Acetylcholine plays a critical role in the neocortex. Cholinergic agonists and acetylcholinesterase inhibitors can enhance cognitive functioning, as does intermittent electrical stimulation of the cortical source of acetylcholine, the nucleus basalis (NB) of Meynert. Here we show in two male monkeys how NB stimulation affects working memory and alters its neural code. NB stimulation increases dorsolateral prefrontal activity during the delay period of spatial working memory tasks and broadens selectivity for stimuli but does not strengthen phasic responses to each neuron's optimal visual stimulus. Paradoxically, despite this decrease in neuronal selectivity, performance improves in many task conditions, likely indicating increased delay period stability. Performance under NB stimulation does decline if distractors similar to the target are presented, consistent with reduced prefrontal selectivity. Our results indicate that stimulation of the cholinergic forebrain increases prefrontal neural activity, and this neuromodulatory tone can improve cognitive performance, subject to a stability-accuracy tradeoff.


Asunto(s)
Núcleo Basal de Meynert/fisiología , Memoria a Corto Plazo/fisiología , Corteza Prefrontal/fisiología , Potenciales de Acción/fisiología , Animales , Conducta Animal , Estimulación Eléctrica , Macaca mulatta , Masculino , Neuronas/fisiología , Análisis y Desempeño de Tareas
18.
Front Syst Neurosci ; 13: 85, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31992972

RESUMEN

Post-Traumatic Stress Disorder (PTSD) is a complex condition that develops after experiencing a severe emotional trauma, with or without physical trauma. There is no known cure and evidence-based treatments, which are effective in reducing symptoms, have low retention rates. It is therefore important, in addition to seeking new therapeutics, to identify ways to reduce the likelihood of developing PTSD. The fact that some, but not all, individuals exposed to the same traumatic event develop PTSD suggests that there is individual susceptibility. Investigating susceptibility and underlying factors will be better guided if there is a coherent framework for such investigations. In this review, we propose that susceptibility is a dynamic state that is comprised of susceptibility factors (before trauma) and sequalae factors (during or after trauma, but before PTSD diagnosis). We define key features of susceptibility and sequalae factors as: (1) they are detectable before trauma (susceptibility factors) or during/shortly after trauma (sequalae factors), (2) they can be manipulated, and (3) manipulation of these factors alters the likelihood of developing PTSD, thus affecting resilience. In this review we stress the importance of investigating susceptibility to PTSD with appropriate animal models, because prospective human studies are expensive and manipulation of susceptibility and sequalae factors for study purposes may not always be feasible. This review also provides a brief overview of a subset of animal models that study PTSD-related behaviors and related alterations in endocrine and brain systems that focus on individual differences, peri- and post-trauma. Attention is drawn to the RISP model (Revealing Individual Susceptibility to a PTSD-like Phenotype) which assesses susceptibility before trauma. Using the RISP model and expression of plasticity-associated immediate early genes, Arc and Homer1a, we have identified impaired hippocampal function as a potential susceptibility factor. We further discuss other putative susceptibility factors and approaches to mitigate them. We assert that this knowledge will guide successful strategies for interventions before, during or shortly after trauma that can decrease the probability of developing PTSD.

19.
Behav Brain Res ; 359: 560-569, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30296528

RESUMEN

Post stroke cognitive impairment (PSCI) is an understudied, long-term complication of stroke, impacting nearly 30-40% of all stroke survivors. No cure is available once the cognitive deterioration manifests. To our knowledge, this is the first study to investigate the long-term effects of C21 treatment on the development of PSCI in aged animals. Treatments with C21 or vehicle were administered orally, 24 h post-stroke, and continued for 30 days. Outcome measures for sensorimotor and cognitive function were performed using a sequence of tests, all blindly conducted and assessed at baseline as well as at different time points post-stroke. Our findings demonstrate that the angiotensin receptor (AT2R) agonist C21 effectively prevents the development of PSCI in aged animals.


Asunto(s)
Envejecimiento/efectos de los fármacos , Disfunción Cognitiva/prevención & control , Nootrópicos/farmacología , Receptor de Angiotensina Tipo 2/agonistas , Accidente Cerebrovascular/tratamiento farmacológico , Sulfonamidas/farmacología , Tiofenos/farmacología , Administración Oral , Envejecimiento/fisiología , Envejecimiento/psicología , Animales , Peso Corporal/efectos de los fármacos , Disfunción Cognitiva/etiología , Disfunción Cognitiva/fisiopatología , Modelos Animales de Enfermedad , Método Doble Ciego , Evaluación Preclínica de Medicamentos , Masculino , Actividad Motora/efectos de los fármacos , Distribución Aleatoria , Ratas Wistar , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología , Accidente Cerebrovascular/psicología , Factores de Tiempo
20.
Behav Brain Res ; 189(2): 244-9, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18295356

RESUMEN

Gene linkage and association studies have implicated the region of chromosome 10q containing the calcyon locus with attention deficit hyperactivity disorder (ADHD), bipolar disorder, and schizophrenia susceptibility. In addition, levels of calcyon protein and transcripts are also significantly increased in postmortem tissue from schizophrenic brains. But whether altered calcyon expression might be part of the disease etiology or merely a patho-physiological side effect is not known. To begin to address this issue, we generated a transgenic mouse line (Cal(OE)) using the human calcyon cDNA in which calcyon expression is up-regulated in a number of forebrain structures including the hippocampus, prefrontal cortex (PFC), striatum, and amygdala. Compared to control littermates, the Cal(OE) mice display a range of abnormal behaviors including spontaneous hyperactivity, reduced anxiety, and/or impaired restraint (harm avoidance) that would indicate that calcyon up-regulation leads to deficits in control over behavioral output.


Asunto(s)
Conducta Exploratoria/fisiología , Hipercinesia/metabolismo , Conducta Impulsiva/metabolismo , Proteínas de la Membrana/metabolismo , Prosencéfalo/metabolismo , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/metabolismo , Reacción de Prevención/fisiología , Hipocampo/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Actividad Motora/fisiología , Neostriado/metabolismo , Corteza Prefrontal/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA