Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Chem Senses ; 472022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35997758

RESUMEN

The brain forms robust associations between odors and emotionally salient memories, making odors especially effective at triggering fearful or traumatic memories. Using Pavlovian olfactory fear conditioning (OFC), a variant of the traditional tone-shock paradigm, this study explored the changes involved in its processing. We assessed the expression of neuronal plasticity markers phosphorylated cyclic adenosine monophosphate response element binding protein (pCREB) and phosphorylated mitogen-activated protein kinase (pMAPK) 24 h and 14 days following OFC, in newborn neurons (EdU+) and in brain regions associated with olfactory memory processing; the olfactory bulb, piriform cortex, amygdale, and hippocampus. Here, we show that all proliferating neurons in the dentate gyrus of the hippocampus and glomerular layer of the olfactory bulb were colocalized with pCREB at 24 h and 14 days post-conditioning, and the number of proliferating neurons at both time points were statistically similar. This suggests the occurrence of long-term potentiation within the neurons of this pathway. Finally, OFC significantly increased the density of pCREB- and pMAPK-positive immunoreactive neurons in the medial and cortical subnuclei of the amygdala and the posterior piriform cortex, suggesting their key involvement in its processing. Together, our investigation identifies changes in neuroplasticity within critical neural circuits responsible for olfactory fear memory.


Asunto(s)
Corteza Piriforme , Amígdala del Cerebelo/metabolismo , Proliferación Celular , Miedo/fisiología , Humanos , Recién Nacido , Corteza Piriforme/fisiología , Olfato/fisiología
2.
Pharmacol Ther ; 239: 108195, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35489438

RESUMEN

Post-traumatic stress disorder (PTSD), characterized by abnormally persistent and distressing memories, is a chronic debilitating condition in need of new treatment options. Current treatment guidelines recommend psychotherapy as first line management with only two drugs, sertraline and paroxetine, approved by U.S. Food and Drug Administration (FDA) for treatment of PTSD. These drugs have limited efficacy as they only reduce symptoms related to depression and anxiety without producing permanent remission. PTSD remains a significant public health problem with high morbidity and mortality requiring major advances in therapeutics. Early evidence has emerged for the beneficial effects of psychedelics particularly in combination with psychotherapy for management of PTSD, including psilocybin, MDMA, LSD, cannabinoids, ayahuasca and ketamine. MDMA and psilocybin reduce barrier to therapy by increasing trust between therapist and patient, thus allowing for modification of trauma related memories. Furthermore, research into the memory reconsolidation mechanisms has allowed for identification of various pharmacological targets to disrupt abnormally persistent memories. A number of pre-clinical and clinical studies have investigated novel and re-purposed pharmacological agents to disrupt fear memory in PTSD. Novel therapeutic approaches like neuropeptide Y, oxytocin, cannabinoids and neuroactive steroids have also shown potential for PTSD treatment. Here, we focus on the role of fear memory in the pathophysiology of PTSD and propose that many of these new therapeutic strategies produce benefits through the effect on fear memory. Evaluation of recent research findings suggests that while a number of drugs have shown promising results in preclinical studies and pilot clinical trials, the evidence from large scale clinical trials would be needed for these drugs to be incorporated in clinical practice.


Asunto(s)
Cannabinoides , N-Metil-3,4-metilenodioxianfetamina , Trastornos por Estrés Postraumático , Estados Unidos , Humanos , Trastornos por Estrés Postraumático/tratamiento farmacológico , N-Metil-3,4-metilenodioxianfetamina/farmacología , N-Metil-3,4-metilenodioxianfetamina/uso terapéutico , Psilocibina/uso terapéutico , Miedo/fisiología , Cannabinoides/uso terapéutico
3.
J Psychiatr Res ; 150: 246-256, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35405409

RESUMEN

Post-traumatic stress disorder (PTSD) develops after an exposure to a life-threatening event and is characterized by intrusive memories. According to memory reconsolidation theory retrieval of memory under certain conditions leads to its labilization and subsequent re-storage which could be disrupted by drugs. Propranolol has been the most commonly investigated drug for memory reconsolidation therapy in clinical trials. Intervention with propranolol have shown mixed results in PTSD patients with some studies showing improvement in symptoms while other failing to replicate these findings. We conducted a systematic review and meta-analysis to determine the efficacy of trauma memory disruption by propranolol on PTSD symptoms and physiological responses in PTSD patients. 3224 publications were assessed for eligibility. Seven studies on effects of propranolol on PTSD symptoms and 3 studies on effects of propranolol on physiological responses were incorporated in the meta-analyses. Overall, results indicate that propranolol did not show a beneficial effect on PTSD symptoms (standardized mean difference: 1.29; 95% CI = -2.16 - 0.17). Similarly, propranolol did not influence skin conductance (standardized mean difference: 0.77; 95% CI = -1.85 - 0.31) or EMG response (standardized mean difference: 0.16; 95% CI = -0.65 - 0.33). However, propranolol significantly reduced heart rate after trauma memory recall compared to placebo (standardized mean difference: 0.67; 95% CI = -1.27 to -0.07). This study finds a lack of evidence for the efficacy of propranolol on traumatic memory disruption, in PTSD patients, to recommend its routine clinical use. However, a high level of heterogeneity, variation in propranolol dosage and inadequate sample sizes mean that these findings require cautious interpretation.


Asunto(s)
Propranolol , Trastornos por Estrés Postraumático , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Miedo , Humanos , Memoria/fisiología , Propranolol/farmacología , Propranolol/uso terapéutico , Trastornos por Estrés Postraumático/tratamiento farmacológico
4.
Psychophysiology ; 59(9): e14048, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35324013

RESUMEN

Individual differences in the ability to habitually regulate emotion may impact the efficacy of fear memory extinction. The aim of this study was to assess the relationship between dispositional cognitive reappraisal and expressive suppression with post-retrieval and standard extinction. Fear memory and extinction were measured with the recovery of skin conductance responses. We also examined the relationship between a temporal feature of electrodermal responding (half-recovery time) and each of the emotion regulation strategies. University students (N = 80) underwent a three-day fear conditioning procedure using a within-subject design consisting of acquisition on day one, post-retrieval extinction and standard extinction on day two, and recovery test on day three. Individual difference data on self-reported levels of cognitive reappraisal, expressive suppression, trait anxiety, and depression were collected. We did not detect a relationship between the two emotion regulation strategies measured in this study and acquisition or extinction. We found, however, that increased dispositional use of cognitive reappraisal was associated with lower spontaneous recovery to both the post-retrieval extinction and standard extinction stimulus after controlling for age, trait anxiety, and depression. There were no associations between expressive suppression and conditioned responses. We also observed patterns of faster dissipation of arousal for reappraisal and slower for suppression to the conditioned stimulus during extinction training, which may represent the unique influence of each emotion strategy on the regulation of fear. We conclude greater daily use of cognitive reappraisal, but not expressive suppression, associates with extinction retention after receiving both standard and post-retrieval extinction.


Asunto(s)
Condicionamiento Clásico , Miedo , Ansiedad/psicología , Cognición/fisiología , Condicionamiento Clásico/fisiología , Emociones/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Humanos
5.
Cereb Cortex ; 32(20): 4619-4639, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-35149865

RESUMEN

Adolescence constitutes a period of vulnerability in the emergence of fear-related disorders (FRD), as a massive reorganization occurs in the amygdala-prefrontal cortex network, critical to regulate fear behavior. Genetic and environmental factors during development may predispose to the emergence of FRD at the adult age, but the underlying mechanisms are poorly understood. In the present study, we tested whether a partial knock-down of tuberous sclerosis complex 2 (Tsc2, Tuberin), a risk gene for neurodevelopmental disorders, in the basolateral amygdala (BLA) from adolescence could alter fear-network functionality and create a vulnerability ground to FRD appearance at adulthood. Using bilateral injection of a lentiviral vector expressing a miRNA against Tsc2 in the BLA of early (PN25) or late adolescent (PN50) rats, we show that alteration induced specifically from PN25 resulted in an increased c-Fos activity at adulthood in specific layers of the prelimbic cortex, a resistance to fear extinction and an overgeneralization of fear to a safe, novel stimulus. A developmental dysfunction of the amygdala could thus play a role in the vulnerability to FRD emergence at adulthood. We propose our methodology as an alternative to model the developmental vulnerability to FRD, especially in its comorbidity with TSC2-related autism syndrome.


Asunto(s)
MicroARNs , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa , Amígdala del Cerebelo , Animales , Extinción Psicológica/fisiología , Miedo/fisiología , Corteza Prefrontal/fisiología , Ratas , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética
6.
Front Neural Circuits ; 15: 660199, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34295224

RESUMEN

Post-traumatic stress disorder (PTSD) is a debilitating and chronic fear-based disorder. Pavlovian fear conditioning protocols have long been utilised to manipulate and study these fear-based disorders. Contextual fear conditioning (CFC) is a particular Pavlovian conditioning procedure that pairs fear with a particular context. Studies on the neural mechanisms underlying the development of contextual fear memories have identified the medial prefrontal cortex (mPFC), or more specifically, the pre-limbic cortex (PL) of the mPFC as essential for the expression of contextual fear. Despite this, little research has explored the role of the PL in contextual fear memory maintenance or examined the role of neuronal mitogen-activated protein kinase (pMAPK; ERK 1/2), brain-derived neurotrophic factor (BDNF), and IBA-1 in microglia in the PL as a function of Pavlovian fear conditioning. The current study was designed to evaluate how the maintenance of two different long-term contextual fear memories leads to changes in the number of immune-positive cells for two well-known markers of neural activity (phosphorylation of MAPK and BDNF) and microglia (IBA-1). Therefore, the current experiment is designed to assess the number of immune-positive pMAPK and BDNF cells, microglial number, and morphology in the PL following CFC. Specifically, 2 weeks following conditioning, pMAPK, BDNF, and microglia number and morphology were evaluated using well-validated antibodies and immunohistochemistry (n = 12 rats per group). A standard CFC protocol applied to rats led to increases in pMAPK, BDNF expression and microglia number as compared to control conditions. Rats in the unpaired fear conditioning (UFC) procedure, despite having equivalent levels of fear to context, did not have any change in pMAPK, BDNF expression and microglia number in the PL compared to the control conditions. These data suggest that alterations in the expression of pMAPK, BDNF, and microglia in the PL can occur for up to 2 weeks following CFC. Together the data suggest that MAPK, BDNF, and microglia within the PL of the mPFC may play a role in contextual fear memory maintenance.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Proteínas de Unión al Calcio/biosíntesis , Miedo/fisiología , Memoria/fisiología , Proteínas de Microfilamentos/biosíntesis , Quinasas de Proteína Quinasa Activadas por Mitógenos/biosíntesis , Corteza Prefrontal/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Proteínas de Unión al Calcio/genética , Condicionamiento Clásico/fisiología , Estimulación Eléctrica/efectos adversos , Miedo/psicología , Expresión Génica , Masculino , Proteínas de Microfilamentos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/psicología
7.
ACS Nano ; 14(11): 16140-16155, 2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33186028

RESUMEN

The development of highly efficient and durable earth-abundant hydrogen evolution reaction (HER) catalysts is crucial for the extensive implementation of the hydrogen economy. Members of the 2D MXenes family, particularly Mo2CTx, have recently been identified as promising HER catalysts. However, their inherent oxidative instability in air and aqueous electrolyte solutions is hindering their widespread use. Herein, we present a simple and scalable method to circumvent adventitious oxidation in Mo2CTx MXenes via in situ sulfidation to form a Mo2CTx/2H-MoS2 nanohybrid. The intimate epitaxial coupling at the Mo2CTx/2H-MoS2 nanohybrid interface afforded superior HER activities, requiring only 119 or 182 mV overpotential to yield -10 or -100 mA cm-2geom current densities, respectively. Density functional theory calculations reveal strongest interfacial adhesion was found within the nanohybrid structure as compared to the physisorbed nanohybrid, and the possibility to tune the HER overpotential through manipulating the extent of MXene sulfidation. Critically, the presence of 2H-MoS2 suppresses further oxidation of the MXene layer, enabling the nanohybrid to sustain industrially relevant current densities of over -450 mA cm-2geom with exceptional durability. Less than 30 mV overpotential degradation was observed after 10 continuous days of electrolysis at a fixed -10 mA cm-2geom current density or 100,000 successive cyclic voltammetry cycles. The exceptional HER durability of the Mo2CTx/2H-MoS2 nanohybrid presents a major step forward to realize practical implementation of MXenes as noble metal free catalysts for broad-based applications in water splitting and energy conversion.

8.
Front Cell Neurosci ; 13: 533, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31866826

RESUMEN

An increase in post-synaptic Ca2+ conductance through activation of the ionotropic N-methyl-D-aspartate receptor (NMDAR) and concomitant structural changes are essential for the initiation of long-term potentiation (LTP) and memory formation. Memories can be initiated by coincident events, as occurs in classical conditioning, where the NMDAR can act as a molecular coincidence detector. Binding of glutamate and glycine, together with depolarization of the postsynaptic cell membrane to remove the Mg2+ channel pore block, results in NMDAR opening for Ca2+ conductance. Accumulating evidence has implicated both force-from-lipids and protein tethering mechanisms for mechanosensory transduction in NMDAR, which has been demonstrated by both, membrane stretch and application of amphipathic molecules such as arachidonic acid (AA). The contribution of mechanosensitivity to memory formation and consolidation may be to increase activity of the NMDAR leading to facilitated memory formation. In this review we look back at the progress made toward understanding the physiological and pathological role of NMDA receptor channels in mechanobiology of the nervous system and consider these findings in like of their potential functional implications for memory formation. We examine recent studies identifying mechanisms of both NMDAR and other mechanosensitive channels and discuss functional implications including gain control of NMDA opening probability. Mechanobiology is a rapidly growing area of biology with many important implications for understanding form, function and pathology in the nervous system.

9.
Front Mol Neurosci ; 12: 221, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31607858

RESUMEN

Odors have proven to be the most resilient trigger for memories of high emotional saliency. Fear associated olfactory memories pose a detrimental threat of potentially transforming into severe mental illness such as fear and anxiety-related disorders. Many studies have deliberated on auditory, visual and general contextual fear memory (CFC) processes; however, fewer studies have investigated mechanisms of olfactory fear memory. Evidence strongly suggests that the neuroanatomical representation of olfactory fear memory differs from that of auditory and visual fear memory. The aim of this review article is to revisit the literature regarding the understanding of the neurobiological process of fear conditioning and to illustrate the circuitry of olfactory fear memory.

10.
Front Cell Neurosci ; 13: 214, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31139053

RESUMEN

Contextual fear conditioning is a Pavlovian conditioning paradigm capable of rapidly creating fear memories to contexts, such as rooms or chambers. Contextual fear conditioning protocols have long been utilized to evaluate how fear memories are consolidated, maintained, expressed, recalled, and extinguished within the brain. These studies have identified the lateral portion of the amygdala and the dorsal portion of the hippocampus as essential for contextual fear memory consolidation. The current study was designed to evaluate how two different contextual fear memories alter amygdala and hippocampus microglia, brain derived neurotrophic factor (BDNF), and phosphorylated cyclic-AMP response element binding (pCREB). We find rats provided with standard contextual fear conditioning to have more microglia and more cells expressing BDNF in the dentate gyrus as compared to a context only control group. Additionally, standard contextual fear conditioning altered microglia morphology to become amoeboid in shape - a common response to central nervous system insult, such as traumatic brain injury, infection, ischemia, and more. The unpaired fear conditioning procedure (whereby non-reinforced and non-overlapping auditory tones were provided at random intervals during conditioning), despite producing equivalent levels of fear as the standard procedure, did not alter microglia, BDNF or pCREB number in any dorsal hippocampus or lateral amygdala brain regions. Despite this, the unpaired fear conditioning protocol produced some alterations in microglia morphology, but less compared to rats provided with standard contextual fear conditioning. Results from this study demonstrate that contextual fear conditioning is capable of producing large alterations to dentate gyrus plasticity and microglia, whereas unpaired fear conditioning only produces minor changes to microglia morphology. These data show, for the first time, that Pavlovian fear conditioning protocols can induce similar responses as trauma, infection or other insults within the central nervous system.

11.
Psychopharmacology (Berl) ; 236(1): 383-397, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30610350

RESUMEN

RATIONALE: The precise neural circuitry that encodes fear memory and its extinction within the brain are not yet fully understood. Fearful memories can be persistent, resistant to extinction, and associated with psychiatric disorders, especially post-traumatic stress disorder (PTSD). Here, we investigated the microtopography of neurons activated during the recall of an extinguished fear memory, as well as the influence of time on this microtopography. METHODS: We used the plasticity-related phosphorylated mitogen-activated protein kinase (pMAPK) to identify neurons activated in the recall of consolidated and extinguished auditory Pavlovian fear memories in rats. Quantitatively matched brain regions were used to investigate activity in the amygdala and prefrontal cortex. RESULTS: Recall of a consolidated, nonextinguished auditory fear memory resulted in a significantly greater number of activated neurons located in the dorsolateral subdivision of the lateral amygdala (LADL) when recalled 24 h after consolidation but not when recalled 7 days later. We found that the recall of an extinction memory was associated with pMAPK activation in the ventrolateral subdivision of the lateral amygdala (LAVL). Next, we showed that the pattern of pMAPK expression in the prelimbic cortex differed spatially following temporal variation in the recall of that memory. The deep and superficial layers of the pre-limbic cortex were engaged in recent recall of a fear memory, but only the superficial layers were recruited if the recall occurred 7 days later. CONCLUSIONS: Collectively, our findings demonstrate a functional microtopography of auditory fear memory during consolidation and extinction at the microanatomical level within the lateral amygdala and medial prefrontal cortex.


Asunto(s)
Amígdala del Cerebelo/fisiología , Mapeo Encefálico , Extinción Psicológica/fisiología , Miedo/fisiología , Consolidación de la Memoria/fisiología , Corteza Prefrontal/fisiología , Amígdala del Cerebelo/anatomía & histología , Amígdala del Cerebelo/metabolismo , Animales , Complejo Nuclear Basolateral/fisiología , Condicionamiento Clásico/fisiología , Masculino , Recuerdo Mental/fisiología , Plasticidad Neuronal/fisiología , Corteza Prefrontal/anatomía & histología , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/fisiopatología
12.
Front Neural Circuits ; 12: 84, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30386215

RESUMEN

In order to understand the relationship between neuronal organization and behavior, precise methods that identify and quantify functional cellular ensembles are required. This is especially true in the quest to understand the mechanisms of memory. Brain structures involved in memory formation and storage, as well as the molecular determinates of memory are well-known, however, the microanatomy of functional neuronal networks remain largely unidentified. We developed a novel approach to statistically map molecular markers in neuronal networks through quantitative topographic measurement. Brain nuclei and their subdivisions are well-defined - our approach allows for the identification of new functional micro-regions within established subdivisions. A set of analytic methods relevant for measurement of discrete neuronal data across a diverse range of brain subdivisions are presented. We provide a methodology for the measurement and quantitative comparison of functional micro-neural network activity based on immunohistochemical markers matched across individual brains using micro-binning and heat mapping within brain sub-nuclei. These techniques were applied to the measurement of different memory traces, allowing for greater understanding of the functional encoding within sub-nuclei and its behavior mediated change. These approaches can be used to understand other functional and behavioral questions, including sub-circuit organization, normal memory function and the complexities of pathology. Precise micro-mapping of functional neuronal topography provides essential data to decode network activity underlying behavior.


Asunto(s)
Mapeo Encefálico/métodos , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Neuronas/fisiología , Animales , Encéfalo/citología , Imagenología Tridimensional/métodos , Masculino , Ratas , Ratas Sprague-Dawley
13.
Neurosci Biobehav Rev ; 92: 43-54, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29752958

RESUMEN

Context is an ever-present combination of discrete environmental elements capable of influencing many psychological processes. When context is associated with an aversive stimulus, a permanent contextual fear memory is formed. Context is hypothesized to greatly influence the treatability of various fear-based pathologies, in particular, post-traumatic stress disorder (PTSD). In order to understand how contextual fear memories are encoded and impact underlying fear pathology, delineation of the underlying neural circuitry of contextual fear memory consolidation and maintenance is essential. Past understandings of contextual fear suggest that the hippocampus only creates a unitary, or single, representation of context. This representation is sent to the amygdala, which creates the associative contextual fear memory. In contrast, here we review new evidence from the literature showing contextual fear memories to be consolidated and maintained by both amygdala and hippocampus. Based on this evidence, we revise the current model of contextual fear memory consolidation, highlighting a larger role for hippocampus. This new model may better explain the role of the hippocampus in PTSD.


Asunto(s)
Condicionamiento Psicológico/fisiología , Miedo/psicología , Hipocampo/fisiología , Memoria/fisiología , Lóbulo Temporal/fisiología , Humanos , Trastornos por Estrés Postraumático/patología
15.
Front Behav Neurosci ; 10: 24, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27013997
16.
Neurobiol Learn Mem ; 127: 64-71, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26642919

RESUMEN

Subtle differences in neuronal microanatomy may be coded in individuals with genetic susceptibility for neuropsychiatric disorders. Genetic susceptibility is a significant risk factor in the development of anxiety disorders, including post-traumatic stress disorder (PTSD). Pavlovian fear conditioning has been proposed to model key aspects of PTSD. According to this theory, PTSD begins with the formation of a traumatic memory which connects relevant environmental stimuli to significant threats to life. The lateral amygdala (LA) is considered to be a key network hub for the establishment of Pavlovian fear conditioning. Substantial research has also linked the LA to PTSD. Here we used a genetic mouse model of fear susceptibility (F-S) and resistance (F-R) to investigate the dendritic and spine structure of principal neurons located in the LA. F-S and F-R lines were bi-directionally selected based on divergent levels of contextual and cued conditioned freezing in response to fear-evoking footshocks. We examined LA principal neuron dendritic and spine morphology in the offspring of experimentally naive F-S and F-R mice. We found differences in the spatial distribution of dendritic branch points across the length of the dendrite tree, with a significant increase in branch points at more distal locations in the F-S compared with F-R line. These results suggest a genetic predisposition toward differences in fear memory strength associated with a dendritic branch point organization of principal neurons in the LA. These micro-anatomical differences in neuron structure in a genetic mouse model of fear susceptibility and resistance provide important insights into the cellular mechanisms of pathophysiology underlying genetic predispositions to anxiety and PTSD.


Asunto(s)
Complejo Nuclear Basolateral/patología , Espinas Dendríticas/patología , Miedo/fisiología , Animales , Reacción de Prevención/fisiología , Condicionamiento Clásico/fisiología , Electrochoque , Ratones , Ratones Endogámicos C57BL
17.
Neurobiol Learn Mem ; 116: 1-13, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25076183

RESUMEN

Pavlovian fear conditioning is an evolutionary conserved and extensively studied form of associative learning and memory. In mammals, the lateral amygdala (LA) is an essential locus for Pavlovian fear learning and memory. Despite significant progress unraveling the cellular mechanisms responsible for fear conditioning, very little is known about the anatomical organization of neurons encoding fear conditioning in the LA. One key question is how fear conditioning to different sensory stimuli is organized in LA neuronal ensembles. Here we show that Pavlovian fear conditioning, formed through either the auditory or visual sensory modality, activates a similar density of LA neurons expressing a learning-induced phosphorylated extracellular signal-regulated kinase (p-ERK1/2). While the size of the neuron population specific to either memory was similar, the anatomical distribution differed. Several discrete sites in the LA contained a small but significant number of p-ERK1/2-expressing neurons specific to either sensory modality. The sites were anatomically localized to different levels of the longitudinal plane and were independent of both memory strength and the relative size of the activated neuronal population, suggesting some portion of the memory trace for auditory and visually cued fear conditioning is allocated differently in the LA. Presenting the visual stimulus by itself did not activate the same p-ERK1/2 neuron density or pattern, confirming the novelty of light alone cannot account for the specific pattern of activated neurons after visual fear conditioning. Together, these findings reveal an anatomical distribution of visual and auditory fear conditioning at the level of neuronal ensembles in the LA.


Asunto(s)
Amígdala del Cerebelo/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Neuronas/fisiología , Estimulación Acústica , Amígdala del Cerebelo/metabolismo , Animales , Recuento de Células , Señales (Psicología) , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Memoria/fisiología , Neuronas/metabolismo , Fosforilación , Estimulación Luminosa , Ratas , Ratas Sprague-Dawley
18.
Neurobiol Learn Mem ; 112: 195-203, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23811025

RESUMEN

Individual variability in the acquisition, consolidation and extinction of conditioned fear potentially contributes to the development of fear pathology including posttraumatic stress disorder (PTSD). Pavlovian fear conditioning is a key tool for the study of fundamental aspects of fear learning. Here, we used a selected mouse line of High and Low Pavlovian conditioned fear created from an advanced intercrossed line (AIL) in order to begin to identify the cellular basis of phenotypic divergence in Pavlovian fear conditioning. We investigated whether phosphorylated MAPK (p44/42 ERK/MAPK), a protein kinase required in the amygdala for the acquisition and consolidation of Pavlovian fear memory, is differentially expressed following Pavlovian fear learning in the High and Low fear lines. We found that following Pavlovian auditory fear conditioning, High and Low line mice differ in the number of pMAPK-expressing neurons in the dorsal sub nucleus of the lateral amygdala (LAd). In contrast, this difference was not detected in the ventral medial (LAvm) or ventral lateral (LAvl) amygdala sub nuclei or in control animals. We propose that this apparent increase in plasticity at a known locus of fear memory acquisition and consolidation relates to intrinsic differences between the two fear phenotypes. These data provide important insights into the micronetwork mechanisms encoding phenotypic differences in fear. Understanding the circuit level cellular and molecular mechanisms that underlie individual variability in fear learning is critical for the development of effective treatment of fear-related illnesses such as PTSD.


Asunto(s)
Complejo Nuclear Basolateral/enzimología , Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/enzimología , Animales , Complejo Nuclear Basolateral/citología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Fenotipo , Distribución Aleatoria
19.
Behav Brain Res ; 257: 39-48, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24070856

RESUMEN

Anxiety traits can be stable and permanent characteristics of an individual across time that is less susceptible of influences by a particular situation. One way to study trait anxiety in an experimental context is through the use of rat lines, selected according to contrasting phenotypes of fear and anxiety. It is not clear whether the behavioral differences between two contrasting rat lines in one given anxiety test are also present in others paradigms of state anxiety. Here, we examine the extent to which multiple anxiety traits generalize across selected animal lines originally selected for a single anxiety trait. We review the behavioral results available in the literature of eight rat genetic models of trait anxiety - namely Maudsley Reactive and Non-reactive rats, Floripa H and L rats, Tsukuba High and Low Emotional rats, High and Low Anxiety-related rats, High and Low Ultrasonic Vocalization rats, Roman High and Low Avoidance rats, Syracuse High and Low Avoidance rats, and Carioca High and Low Conditioned Freezing rats - across 11 behavioral paradigms of innate anxiety or aversive learning frequently used in the experimental setting. We observed both convergence and divergence of behavioral responses in these selected lines across the 11 paradigms. We find that predisposition for specific anxiety traits will usually be generalized to other anxiety provoking stimuli. However this generalization is not observed across all genetic models indicating some unique trait and state interactions. Genetic models of enhanced-anxiety related responses are beginning to help define how anxiety can manifest differently depending on the underlying traits and the current environmentally induced state.


Asunto(s)
Ansiedad , Conducta Animal/fisiología , Animales , Ansiedad/genética , Ansiedad/fisiopatología , Ansiedad/psicología , Modelos Animales de Enfermedad , Genotipo , Ratas
20.
Eur J Neurosci ; 38(9): 3314-24, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23968228

RESUMEN

Genetic variability in the strength and precision of fear memory is hypothesised to contribute to the etiology of anxiety disorders, including post-traumatic stress disorder. We generated fear-susceptible (F-S) or fear-resistant (F-R) phenotypes from an F8 advanced intercross line (AIL) of C57BL/6J and DBA/2J inbred mice by selective breeding. We identified specific traits underlying individual variability in Pavlovian conditioned fear learning and memory. Offspring of selected lines differed in the acquisition of conditioned fear. Furthermore, F-S mice showed greater cued fear memory and generalised fear in response to a novel context than F-R mice. F-S mice showed greater basal corticosterone levels and hypothalamic corticotrophin-releasing hormone (CRH) mRNA levels than F-R mice, consistent with higher hypothalamic-pituitary-adrenal (HPA) axis drive. Hypothalamic mineralocorticoid receptor and CRH receptor 1 mRNA levels were decreased in F-S mice as compared with F-R mice. Manganese-enhanced magnetic resonance imaging (MEMRI) was used to investigate basal levels of brain activity. MEMRI identified a pattern of increased brain activity in F-S mice that was driven primarily by the hippocampus and amygdala, indicating excessive limbic circuit activity in F-S mice as compared with F-R mice. Thus, selection pressure applied to the AIL population leads to the accumulation of heritable trait-relevant characteristics within each line, whereas non-behaviorally relevant traits remain distributed. Selected lines therefore minimise false-positive associations between behavioral phenotypes and physiology. We demonstrate that intrinsic differences in HPA axis function and limbic excitability contribute to phenotypic differences in the acquisition and consolidation of associative fear memory. Identification of system-wide traits predisposing to variability in fear memory may help in the direction of more targeted and efficacious treatments for fear-related pathology.


Asunto(s)
Miedo , Carácter Cuantitativo Heredable , Retención en Psicología , Animales , Condicionamiento Clásico , Corticosterona/sangre , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Generalización Psicológica , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiología , Endogamia , Ratones , Ratones Endogámicos C57BL , Fenotipo , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...