Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38798391

RESUMEN

Combining mouse genetics, electrophysiology, and behavioral training and testing, we explored how sleep disruption may affect the function of anxiety-controlling circuits, focusing on projections from the basolateral nucleus of the amygdala (BLA) to CRF-positive cells in the lateral division of the central amygdala (CeL). We found in Crh-IRES-Cre::Ai14(tdTomato) reporter female mice that 6 hours of sleep disruption during their non-active (light) cycle may be anxiogenic. Notably, the AMPAR/NMDAR EPSC amplitude ratio at the BLA inputs to CRF-CeL cells (CRF CeL ), assessed with whole-cell recordings in ex vivo experiments, was enhanced in slices from sleep-disrupted mice, whereas paired-pulse ratio (PPR) of the EPSCs induced by two closely spaced presynaptic stimuli remained unchanged. These findings indicate that sleep disruption-associated synaptic enhancements in glutamatergic projections from the BLA to CRF-CeL neurons may be postsynaptically expressed. We found also that the excitation/inhibition (E/I) ratio in the BLA to CRF CeL inputs was increased in sleep-disrupted mice, suggesting that the functional efficiency of excitation in BLA inputs to CRF CeL cells has increased following sleep disruption, thus resulting in their enhanced activation. The latter could be translated into enhanced anxiogenesis as activation of CRF cells in the CeL was shown to promote anxiety-like behaviors.

2.
Sci Rep ; 14(1): 8919, 2024 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637645

RESUMEN

The natural alignment of animals into social dominance hierarchies produces adaptive, and potentially maladaptive, changes in the brain that influence health and behavior. Aggressive and submissive behaviors assumed by animals through dominance interactions engage stress-dependent neural and hormonal systems that have been shown to correspond with social rank. Here, we examined the association between social dominance hierarchy status established within cages of group-housed mice and the expression of the stress peptide PACAP in the bed nucleus of the stria terminalis (BNST) and central nucleus of the amygdala (CeA). We also examined the relationship between social dominance rank and blood corticosterone (CORT) levels, body weight, motor coordination (rotorod) and acoustic startle. Male C57BL/6 mice were ranked as either Dominant, Submissive, or Intermediate based on counts of aggressive/submissive encounters assessed at 12 weeks-old following a change in homecage conditions. PACAP expression was significantly higher in the BNST, but not the CeA, of Submissive mice compared to the other groups. CORT levels were lowest in Submissive mice and appeared to reflect a blunted response following events where dominance status is recapitulated. Together, these data reveal changes in specific neural/neuroendocrine systems that are predominant in animals of lowest social dominance rank, and implicate PACAP in brain adaptations that occur through the development of social dominance hierarchies.


Asunto(s)
Corticosterona , Núcleos Septales , Animales , Masculino , Ratones , Amígdala del Cerebelo/metabolismo , Ratones Endogámicos C57BL , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Núcleos Septales/metabolismo , Predominio Social , Estrés Psicológico/metabolismo
3.
Drug Alcohol Depend ; 255: 110967, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38150894

RESUMEN

BACKGROUND: Opioid withdrawal symptoms (OWS) are highly aversive and prompt unprescribed opioid use, which increases morbidity, mortality, and, among individuals being treated for opioid use disorder (OUD), recurrence. OWS are driven by sympathetic nervous system (SNS) hyperactivity that occurs when blood opioid levels wane. We tested whether brief inhalation of xenon gas, which inhibits SNS activity and is used clinically for anesthesia and diagnostic imaging, attenuates naltrexone-precipitated withdrawal-like signs in morphine-dependent mice. METHODS: Adult CD-1 mice were implanted with morphine sulfate-loaded (60 mg/ml) minipumps and maintained for 6 days to establish morphine dependence. On day 7, mice were given subcutaneous naltrexone (0.3 mg/kg) and placed in a sealed exposure chamber containing either 21% oxygen/balance nitrogen (controls) or 21% oxygen/added xenon peaking at 30%/balance nitrogen. After 10 minutes, mice were transferred to observation chambers and videorecorded for 45 minutes. Videos were scored in a blind manner for morphine withdrawal behaviors. Data were analyzed using 2-way ANOVAs testing for treatment and sex effects. RESULTS AND CONCLUSIONS: Xenon-exposed mice exhibited fewer jumps (P = 0.010) and jumping suppression was detectible within the first 10-minute video segment, but no sex differences were detected. Brief inhalation of low concentration xenon rapidly and substantially attenuated naltrexone-precipitated jumping in morphine-dependent mice, suggesting that it can inhibit OWS. If xenon effects translate to humans with OUD, xenon inhalation may be effective for reducing OWS, unprescribed opioid use, and for easing OUD treatment initiation, which could help lower excess morbidity and mortality associated with OUD.


Asunto(s)
Dependencia de Morfina , Trastornos Relacionados con Opioides , Síndrome de Abstinencia a Sustancias , Humanos , Adulto , Ratones , Animales , Naltrexona/farmacología , Naltrexona/uso terapéutico , Analgésicos Opioides/uso terapéutico , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/uso terapéutico , Morfina/farmacología , Morfina/uso terapéutico , Narcóticos/uso terapéutico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Trastornos Relacionados con Opioides/tratamiento farmacológico , Nitrógeno/uso terapéutico , Oxígeno/uso terapéutico
4.
bioRxiv ; 2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37205328

RESUMEN

The natural alignment of animals into social dominance hierarchies produces adaptive, and potentially maladaptive, changes in the brain that influence health and behavior. Aggressive and submissive behaviors assumed by animals through dominance interactions engage stress-dependent neural and hormonal systems that have been shown to correspond with social rank. Here, we examined the impact of social dominance hierarchies established within cages of group-housed laboratory mice on expression of the stress peptide pituitary adenylate cyclase-activating polypeptide (PACAP) in areas of the extended amygdala comprising the bed nucleus of the stria terminalis (BNST) and central nucleus of the amygdala (CeA). We also quantified the impact of dominance rank on corticosterone (CORT), body weight, and behavior including rotorod and acoustic startle response. Weight-matched male C57BL/6 mice, group-housed (4/cage) starting at 3 weeks of age, were ranked as either most-dominant (Dominant), least-dominant (Submissive) or in-between rank (Intermediate) based on counts of aggressive and submissive encounters assessed at 12 weeks-old following a change in homecage conditions. We found that PACAP expression was significantly higher in the BNST, but not the CeA, of Submissive mice compared to the other two groups. CORT levels were lowest in Submissive mice and appeared to reflect a blunted response following social dominance interactions. Body weight, motor coordination, and acoustic startle were not significantly different between the groups. Together, these data reveal changes in specific neural/neuroendocrine systems that are predominant in animals of lowest social dominance rank, and implicate PACAP in brain adaptations that occur through the development of social dominance hierarchies.

5.
Nat Rev Neurol ; 18(5): 273-288, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35352034

RESUMEN

Post-traumatic stress disorder (PTSD) is a maladaptive and debilitating psychiatric disorder, characterized by re-experiencing, avoidance, negative emotions and thoughts, and hyperarousal in the months and years following exposure to severe trauma. PTSD has a prevalence of approximately 6-8% in the general population, although this can increase to 25% among groups who have experienced severe psychological trauma, such as combat veterans, refugees and victims of assault. The risk of developing PTSD in the aftermath of severe trauma is determined by multiple factors, including genetics - at least 30-40% of the risk of PTSD is heritable - and past history, for example, prior adult and childhood trauma. Many of the primary symptoms of PTSD, including hyperarousal and sleep dysregulation, are increasingly understood through translational neuroscience. In addition, a large amount of evidence suggests that PTSD can be viewed, at least in part, as a disorder that involves dysregulation of normal fear processes. The neural circuitry underlying fear and threat-related behaviour and learning in mammals, including the amygdala-hippocampus-medial prefrontal cortex circuit, is among the most well-understood in behavioural neuroscience. Furthermore, the study of threat-responding and its underlying circuitry has led to rapid progress in understanding learning and memory processes. By combining molecular-genetic approaches with a translational, mechanistic knowledge of fear circuitry, transformational advances in the conceptual framework, diagnosis and treatment of PTSD are possible. In this Review, we describe the clinical features and current treatments for PTSD, examine the neurobiology of symptom domains, highlight genomic advances and discuss translational approaches to understanding mechanisms and identifying new treatments and interventions for this devastating syndrome.


Asunto(s)
Trastornos por Estrés Postraumático , Veteranos , Adulto , Amígdala del Cerebelo , Animales , Miedo/fisiología , Hipocampo , Humanos , Mamíferos , Trastornos por Estrés Postraumático/diagnóstico , Trastornos por Estrés Postraumático/terapia
6.
Int J Neuropsychopharmacol ; 23(7): 469-479, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32242615

RESUMEN

BACKGROUND: Tobacco use is prevalent in individuals who are routinely exposed to stress. However, little is known about how nicotine affects responses to trauma. We examined in rats how nicotine exposure affects fear conditioning, a procedure often used to study stress-related psychiatric illness. METHODS: We examined 2 methods of nicotine exposure: self-administration, modeling voluntary use, and experimenter-programmed subcutaneous administration, modeling medicinal administration (nicotine patch). For self-administered nicotine, rats trained to self-administer nicotine i.v. were fear conditioned (via light cue preceding foot-shock) either immediately after a 12-hour self-administration session or 12 hours later during a period with somatic signs of nicotine withdrawal. For experimenter-delivered nicotine, rats were conditioned after 1-21 days of nicotine delivered by programmable (12 hours on) subcutaneous mini-pumps. Tests to evaluate acoustic startle responses to the conditioning environment (context-potentiated startle) and in the presence or absence of the light cue (fear-potentiated startle) occurred after a 10-day period. RESULTS: Rats fear conditioned immediately after nicotine self-administration showed reduced responses to the shock-associated context, whereas those trained during nicotine withdrawal showed exaggerated responses. Experimenter-programmed nicotine produced effects qualitatively similar to those seen with self-administered nicotine. CONCLUSIONS: Self-administration or experimenter-programmed delivery of nicotine immediately before exposure to aversive events can reduce conditioned fear responses. In contrast, exposure to aversive events during nicotine withdrawal exacerbates fear responses. These studies raise the possibility of developing safe and effective methods to deliver nicotine or related drugs to mitigate the effects of stress while also highlighting the importance of preventing withdrawal in nicotine-dependent individuals.


Asunto(s)
Miedo/psicología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Síndrome de Abstinencia a Sustancias/psicología , Tabaquismo/psicología , Estimulación Acústica , Animales , Condicionamiento Clásico , Señales (Psicología) , Bombas de Infusión Implantables , Inyecciones Intravenosas , Inyecciones Subcutáneas , Luz , Masculino , Ratas , Ratas Long-Evans , Reflejo de Sobresalto/efectos de los fármacos , Autoadministración , Tabaquismo/fisiopatología
7.
Artículo en Inglés | MEDLINE | ID: mdl-31538751

RESUMEN

Posttraumatic stress disorder (PTSD) is a debilitating disease with limited available treatment options and for which novel effective interventions constitute a significant unmet need. This case report describes successful treatment of a patient with panic disorder and PTSD stemming from the 2010 Moscow subway terrorist attacks through the combination of script-driven trauma memory reactivation and inhalation of a xenon-based gas mixture. Xenon is a competitive inhibitor of N-methyl-d-aspartate receptors known to play a role in memory reconsolidation, a learning and memory process wherein memories temporarily enter a labile state after reactivation and may be modified. Literature describing current pharmacologic and exposure-based treatments is reviewed and provides the basis for use of this novel treatment strategy to target and modify emotional memories.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Terapia Implosiva/métodos , Trastorno de Pánico/tratamiento farmacológico , Trastornos por Estrés Postraumático/tratamiento farmacológico , Xenón/farmacología , Adulto , Terapia Combinada , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Femenino , Humanos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Xenón/administración & dosificación
8.
Neurobiol Learn Mem ; 157: 24-34, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30458282

RESUMEN

The stress-related neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is implicated in neuromodulation of learning and memory. PACAP can alter synaptic plasticity and has direct actions on neurons in the amygdala and hippocampus that could contribute to its acute and persistent effects on the consolidation and expression of conditioned fear. We recently demonstrated that intracerebroventricular (ICV) infusion of PACAP prior to fear conditioning (FC) results in initial amnestic-like effects followed by hyper-expression of conditioned freezing with repeated testing, and analyses of immediate-early gene c-Fos expression suggested that the central nucleus of the amygdala (CeA), but not the lateral/basolateral amygdala (LA/BLA) or hippocampus, are involved in these PACAP effects. Here, we extend that work by examining the expression of the synaptic plasticity marker activity-regulated cytoskeleton-associated protein (Arc/Arg 3.1) after PACAP administration and FC. Male Sprague-Dawley rats were implanted with cannula for ICV infusion of PACAP-38 (1.5 µg) or vehicle followed by FC and tests for conditioned freezing. One hour after FC, Arc protein expression was significantly elevated in the CeA and bed nucleus of the stria terminalis (BNST), interconnected structures that are key elements of the extended amygdala, in rats that received the combination of PACAP + FC. In contrast, Arc expression within the subdivisions of the hippocampus, or the LA/BLA, were unchanged. A subpopulation of Arc-positive cells in both the CeA and BNST also express PKCdelta, an intracellular marker that has been used to identify microcircuits that gate conditioned fear in the CeA. Consistent with our previous findings, on the following day conditioned freezing behavior was reduced in rats that had been given the combination of PACAP + FC-an amnestic-like effect-and Arc expression levels had returned to baseline. Given the established role of Arc in modifying synaptic plasticity and memory formation, our findings suggest that PACAP-induced overexpression of Arc following fear conditioning may disrupt neuroplastic changes within populations of CeA and BNST neurons normally responsible for encoding fear-related cues that, in this case, results in altered fear memory consolidation. Hence, PACAP systems may represent an axis on which stress and experience-driven neurotransmission converge to alter emotional memory, and mediate pathologies that are characteristic of psychiatric illnesses such as post-traumatic stress disorder.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Proteínas del Citoesqueleto/metabolismo , Miedo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Núcleos Septales/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Animales , Condicionamiento Clásico , Masculino , Plasticidad Neuronal , Neuronas/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Ratas Sprague-Dawley , Núcleos Septales/efectos de los fármacos
9.
Psychoneuroendocrinology ; 64: 12-21, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26590791

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is implicated in stress regulation and learning and memory. PACAP has neuromodulatory actions on brain structures within the limbic system that could contribute to its acute and persistent effects in animal models of stress and anxiety-like behavior. Here, male Sprague-Dawley rats were implanted with intracerebroventricular (ICV) cannula for infusion of PACAP-38 (0.5, 1, or 1.5 µg) or vehicle followed 30 min later by fear conditioning. Freezing was measured early (1, 4, and 7 days) or following a delay (7, 10, and 13 days) after conditioning. PACAP (1.5 µg) produced a bi-phasic response in freezing behavior across test days: relative to controls, PACAP-treated rats showed a reduction in freezing when tested 1 or 7 days after fear conditioning that evolved into a significant elevation in freezing by the third test session in the early, but not delayed, group. Corticosterone (CORT) levels were significantly elevated in PACAP-treated rats following fear conditioning, but not at the time of testing (Day 1). Brain c-Fos expression revealed PACAP-dependent alterations within, as well as outside of, areas typically implicated in fear conditioning. Our findings raise the possibility that PACAP disrupts fear memory consolidation by altering synaptic plasticity within neurocircuits normally responsible for encoding fear-related cues, producing a type of dissociation or peritraumatic amnesia often seen in people early after exposure to a traumatic event. However, fear memories are retained such that repeated testing and memory reactivation (e.g., re-experiencing) causes the freezing response to emerge and persist at elevated levels. PACAP systems may represent an axis on which stress and exposure to trauma converge to promote maladaptive behavioral responses characteristic of psychiatric illnesses such as post-traumatic stress disorder (PTSD).


Asunto(s)
Conducta Animal/efectos de los fármacos , Condicionamiento Psicológico , Miedo/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Corticosterona/sangre , Relación Dosis-Respuesta a Droga , Reacción Cataléptica de Congelación/efectos de los fármacos , Infusiones Intraventriculares , Masculino , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Ratas , Factores de Tiempo
10.
Biol Psychiatry ; 80(12): 955-964, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-26229039

RESUMEN

BACKGROUND: Severe or prolonged stress can trigger psychiatric illnesses including mood and anxiety disorders. Recent work indicates that pituitary adenylate cyclase-activating polypeptide (PACAP) plays an important role in regulating stress effects. In rodents, exogenous PACAP administration can produce persistent elevations in the acoustic startle response, which may reflect anxiety-like signs including hypervigilance. We investigated whether PACAP causes acute or persistent alterations in behaviors that reflect other core features of mood and anxiety disorders (motivation, social interaction, and attention). METHODS: Using male Sprague Dawley rats, we examined if PACAP (.25-1.0 µg, intracerebroventricular infusion) affects motivation as measured in the intracranial self-stimulation test. We also examined if PACAP alters interactions with a conspecific in the social interaction test. Finally, we examined if PACAP affects performance in the 5-choice serial reaction time task, which quantifies attention and error processing. RESULTS: Dose-dependent disruptions in motivation, social interaction, and attention were produced by PACAP, as reflected by increases in reward thresholds, decreases in social behaviors, and decreases in correct responses and alterations in posterror accuracy. Behavior normalized quickly in the intracranial self-stimulation and 5-choice serial reaction time task tests but remained dysregulated in the social interaction test. Effects on attention were attenuated by the corticotropin-releasing factor receptor-1 antagonist antalarmin but not the κ opioid receptor antagonist JDTic. CONCLUSIONS: Our findings suggest that PACAP affects numerous domains often dysregulated in mood and anxiety disorders, but that individual signs depend on brain substrates that are at least partially independent. This work may help to devise therapeutics that mitigate specific signs of these disorders.


Asunto(s)
Anhedonia/efectos de los fármacos , Atención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Motivación/efectos de los fármacos , Neurotransmisores/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Conducta Social , Animales , Modelos Animales de Enfermedad , Masculino , Neurotransmisores/administración & dosificación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/administración & dosificación , Ratas , Ratas Sprague-Dawley
11.
PLoS One ; 9(8): e106189, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25162644

RESUMEN

Xenon (Xe) is a noble gas that has been developed for use in people as an inhalational anesthestic and a diagnostic imaging agent. Xe inhibits glutamatergic N-methyl-D-aspartate (NMDA) receptors involved in learning and memory and can affect synaptic plasticity in the amygdala and hippocampus, two brain areas known to play a role in fear conditioning models of post-traumatic stress disorder (PTSD). Because glutamate receptors also have been shown to play a role in fear memory reconsolidation--a state in which recalled memories become susceptible to modification--we examined whether Xe administered after fear memory reactivation could affect subsequent expression of fear-like behavior (freezing) in rats. Male Sprague-Dawley rats were trained for contextual and cued fear conditioning and the effects of inhaled Xe (25%, 1 hr) on fear memory reconsolidation were tested using conditioned freezing measured days or weeks after reactivation/Xe administration. Xe administration immediately after fear memory reactivation significantly reduced conditioned freezing when tested 48 h, 96 h or 18 d after reactivation/Xe administration. Xe did not affect freezing when treatment was delayed until 2 h after reactivation or when administered in the absence of fear memory reactivation. These data suggest that Xe substantially and persistently inhibits memory reconsolidation in a reactivation and time-dependent manner, that it could be used as a new research tool to characterize reconsolidation and other memory processes, and that it could be developed to treat people with PTSD and other disorders related to emotional memory.


Asunto(s)
Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Memoria/efectos de los fármacos , Trastornos por Estrés Postraumático/tratamiento farmacológico , Tranquilizantes/farmacología , Xenón/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Señales (Psicología) , Modelos Animales de Enfermedad , Emociones/fisiología , Miedo/psicología , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Hipocampo/efectos de los fármacos , Masculino , Memoria/fisiología , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/psicología
12.
Proc Natl Acad Sci U S A ; 110(12): 4798-803, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487762

RESUMEN

Synaptic mechanisms underlying memory reconsolidation after retrieval are largely unknown. Here we report that synapses in projections to the lateral nucleus of the amygdala implicated in auditory fear conditioning, which are potentiated by learning, enter a labile state after memory reactivation, and must be restabilized through a postsynaptic mechanism implicating the mammalian target of rapamycin kinase-dependent signaling. Fear-conditioning-induced synaptic enhancements were primarily presynaptic in origin. Reconsolidation blockade with rapamycin, inhibiting mammalian target of rapamycin kinase activity, suppressed synaptic potentiation in slices from fear-conditioned rats. Surprisingly, this reduction of synaptic efficacy was mediated by post- but not presynaptic mechanisms. These findings suggest that different plasticity rules may apply to the processes underlying the acquisition of original fear memory and postreactivational stabilization of fear-conditioning-induced synaptic enhancements mediating fear memory reconsolidation.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Miedo/fisiología , Aprendizaje/fisiología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Amígdala del Cerebelo/citología , Animales , Antibacterianos/farmacología , Masculino , Microdisección , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Sirolimus/farmacología , Transmisión Sináptica/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Técnicas de Cultivo de Tejidos
13.
J Neurosci ; 32(41): 14165-77, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23055486

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide expressed in the brain, where it may act as a neuromodulator or neurotransmitter contributing to different behavioral processes and stress responses. PACAP is highly expressed in the amygdala, a subcortical brain area involved in both innate and learned fear, suggesting a role for PACAP-mediated signaling in fear-related behaviors. It remains unknown, however, whether and how PACAP affects neuronal and synaptic functions in the amygdala. In this study, we focused on neurons in the lateral division of the central nucleus (CeL), where PACAP-positive presynaptic terminals were predominantly found within the amygdala. In our experiments on rat brain slices, exogenous application of PACAP did not affect either resting membrane potential or membrane excitability of CeL neurons. PACAP enhanced, however, excitatory synaptic transmission in projections from the basolateral nucleus (BLA) to the CeL, while inhibitory transmission in the same pathway was unaffected. PACAP-induced potentiation of glutamatergic synaptic responses persisted after the washout of PACAP and was blocked by the VPAC1 receptor antagonist, suggesting that VPAC1 receptors might mediate synaptic effects of PACAP in the CeL. Moreover, potentiation of synaptic transmission by PACAP was dependent on postsynaptic activation of protein kinase A and calcium/calmodulin-dependent protein kinase II, as well as synaptic targeting of GluR1 subunit-containing AMPA receptors. Thus, PACAP may upregulate excitatory neurotransmission in the BLA-CeL pathway postsynaptically, consistent with the known roles of PACAP in control of fear-related behaviors.


Asunto(s)
Amígdala del Cerebelo/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Regulación de la Expresión Génica , Red Nerviosa/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Animales , Femenino , Masculino , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/biosíntesis , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Ratas , Ratas Sprague-Dawley
14.
Neuropharmacology ; 63(2): 250-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22465203

RESUMEN

GABA(A) receptor modulating drugs such as benzodiazepines (BZs) have been used to treat anxiety disorders for over five decades. In order to determine whether the same or different GABA(A) receptor subtypes are necessary for the anxiolytic-like action of BZs in unconditioned anxiety and conditioned fear models, we investigated the role of different GABA(A) receptor subtypes by challenging wild type, α1(H101R), α2(H101R) and α3(H126R) mice bred on the C57BL/6J background with diazepam or chlordiazepoxide in the elevated plus maze and the fear-potentiated startle paradigms. Both drugs significantly increased open arm exploration in the elevated plus maze in wild type, α1(H101R) and α3(H126R), but this effect was abolished in α2(H101R) mice; these were expected results based on previous published results. In contrast, while administration of diazepam and chlordiazepoxide significantly attenuated fear-potentiated startle (FPS) in wild type mice and α3(H126R) mice, the fear-reducing effects of these drugs were absent in both α1(H101R) and α2(H101R) point mutants, indicating that both α1- and α2-containing GABA(A) receptors are necessary for BZs to exert their effects on conditioned fear responses. Our findings illustrate both an overlap and a divergence between the GABA(A) receptor subtype requirements for the impact of BZs, specifically that both α1- and α2-containing GABA(A) receptors are necessary for BZs to reduce conditioned fear whereas only α2-containing GABA(A) receptors are needed for BZ-induced anxiolysis in unconditioned tests of anxiety. This raises the possibility that GABAergic pharmacological interventions for specific anxiety disorders can be differentially tailored.


Asunto(s)
Ansiolíticos/uso terapéutico , Ansiedad/tratamiento farmacológico , Clordiazepóxido/uso terapéutico , Condicionamiento Psicológico/efectos de los fármacos , Diazepam/uso terapéutico , Miedo/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Animales , Ansiolíticos/farmacología , Ansiedad/genética , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Clordiazepóxido/farmacología , Condicionamiento Psicológico/fisiología , Diazepam/farmacología , Miedo/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Subunidades de Proteína/genética , Receptores de GABA-A/genética , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología
15.
Cell Mol Neurobiol ; 32(5): 737-48, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22090159

RESUMEN

Epinephrine (Epi), which initiates short-term responses to cope with stress, is, in part, stress-regulated via genetic control of its biosynthetic enzyme, phenylethanolamine N-methyltransferase (PNMT). In rats, immobilization (IMMO) stress activates the PNMT gene in the adrenal medulla via Egr-1 and Sp1 induction. Yet, elevated Epi induced by acute and chronic stress is associated with stress induced, chronic illnesses of cardiovascular, immune, cancerous, and behavioral etiologies. Major sources of Epi include the adrenal medulla and brainstem. Although catecholamines do not cross the blood-brain barrier, circulating Epi from the adrenal medulla may communicate with the central nervous system and stress circuitry by activating vagal nerve ß-adrenergic receptors to release norepinephrine, which could then stimulate release of the same from the nucleus tractus solitarius and locus coeruleus. In turn, the basal lateral amygdala (BLA) may activate to stimulate afferents to the hypothalamus, neocortex, hippocampus, caudate nucleus, and other brain regions sequentially. Recently, we have shown that repeated IMMO or force swim stress may evoke stress resiliency, as suggested by changes in expression and extinction of fear memory in the fear-potentiated startle paradigm. However, concomitant adrenergic changes seem stressor dependent. Present studies aim to identify stressful conditions that elicit stress resiliency versus stress sensitivity, with the goal of developing a model to investigate the potential role of Epi in stress-associated illness. If chronic Epi over expression does elicit illness, possibilities for alternative therapeutics exist through regulating stress-induced Epi expression, adrenergic receptor function and/or corticosteroid effects on Epi, adrenergic receptors and the stress axis.


Asunto(s)
Enfermedad , Epinefrina/metabolismo , Estrés Psicológico/metabolismo , Adrenérgicos , Animales , Epinefrina/química , Humanos , Transducción de Señal , Factores de Tiempo
16.
Nat Neurosci ; 15(1): 113-22, 2011 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-22158512

RESUMEN

Long-term synaptic enhancements in cortical and thalamic auditory inputs to the lateral nucleus of the amygdala (LAn) mediate encoding of conditioned fear memory. It is not known, however, whether the convergent auditory conditioned stimulus (CSa) pathways interact with each other to produce changes in their synaptic function. We found that continuous paired stimulation of thalamic and cortical auditory inputs to the LAn with the interstimulus delay approximately mimicking a temporal pattern of their activation in behaving animals during auditory fear conditioning resulted in persistent potentiation of synaptic transmission in the cortico-amygdala pathway in rat brain slices. This form of input timing-dependent plasticity (ITDP) in cortical input depends on inositol 1,4,5-trisphosphate (InsP(3))-sensitive Ca(2+) release from internal stores and postsynaptic Ca(2+) influx through calcium-permeable kainate receptors during its induction. ITDP in the auditory projections to the LAn, determined by characteristics of presynaptic activity patterns, may contribute to the encoding of the complex CSa.


Asunto(s)
Amígdala del Cerebelo/fisiología , Corteza Cerebral/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Tálamo/fisiología , Animales , Señalización del Calcio/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Inositol 1,4,5-Trifosfato/metabolismo , Vías Nerviosas/fisiología , Ratas , Receptores de Ácido Kaínico/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología
17.
Biol Psychiatry ; 70(5): 425-33, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531393

RESUMEN

BACKGROUND: The kappa opioid receptor (KOR) system contributes to the prodepressive and aversive consequences of stress and is implicated in the facilitation of conditioned fear and anxiety in rodents. Here, we sought to identify neural circuits that mediate KOR system effects on fear and anxiety in rats. METHODS: We assessed whether fear conditioning induces plasticity in KOR or dynorphin (the endogenous KOR ligand) messenger RNA (mRNA) expression in the basolateral (BLA) and central (CeA) nuclei of the amygdala, hippocampus, or striatum. We then assessed whether microinfusions of the KOR antagonist JDTic (0-10 µg/side) into the BLA or CeA affect the expression of conditioned fear or anxiety. Finally, we examined whether fear extinction induces plasticity in KOR mRNA expression that relates to the quality of fear extinction. RESULTS: Fear conditioning upregulated KOR mRNA in the BLA by 65% and downregulated it in the striatum by 22%, without affecting KOR levels in the CeA or hippocampus, or dynorphin levels in any region. KOR antagonism in either the BLA or CeA decreased conditioned fear in the fear-potentiated startle paradigm, whereas KOR antagonism in the BLA, but not the CeA, produced anxiolytic-like effects in the elevated plus maze. Effective fear extinction was associated with a 67% reduction in KOR mRNA in the BLA. CONCLUSIONS: These findings suggest that fear conditioning and extinction dynamically regulate KOR expression in the BLA and provide evidence that the BLA and CeA are important neural substrates mediating the anxiolytic-like effects of KOR antagonists in models of fear and anxiety.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Condicionamiento Psicológico/fisiología , Dinorfinas/fisiología , Miedo/fisiología , Receptores Opioides kappa/fisiología , Transducción de Señal/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Condicionamiento Psicológico/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Extinción Psicológica/fisiología , Miedo/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Microinyecciones , Piperidinas/administración & dosificación , Piperidinas/farmacología , Ratas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/biosíntesis , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Transducción de Señal/efectos de los fármacos , Tetrahidroisoquinolinas/administración & dosificación , Tetrahidroisoquinolinas/farmacología
18.
J Neurosci ; 31(8): 3095-103, 2011 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-21414930

RESUMEN

Stress triggers psychiatric conditions including depressive and anxiety disorders. The mechanisms by which stress produces persistent changes in behavior are not fully understood. Here we show in rats that stress (footshock) activates the transcription factor cAMP response element binding protein (CREB) within the nucleus accumbens shell (NAS), a brain area involved in encoding reward and aversion. To examine the behavioral significance of altered CREB function in the NAS, we used viral vectors to elevate or disrupt CREB in this region. Elevated CREB produced increases in intracranial self-stimulation thresholds, a depressive-like sign reflecting anhedonia (decreased sensitivity to reward), whereas disruption of CREB function by expression of a dominant-negative CREB had the opposite effect. To determine whether neuroadaptations that produce anhedonia subsequently affect vulnerability to stress-induced behavioral adaptations, we subjected rats with altered CREB function in the NAS to fear conditioning. Although neither elevation nor disruption of CREB function altered the development of conditioned fear, elevation of CREB impaired extinction of conditioned fear. To mimic downstream effects of CREB activation on expression of the opioid peptide dynorphin, we microinjected the κ-opioid receptor (KOR) agonist U50,488 directly into the NAS. KOR stimulation produced anhedonia but had no effect on expression or extinction of conditioned fear. These findings demonstrate that activation of CREB in the NAS produces multiple behavioral signs (anhedonia, impaired extinction) characteristic of experience-dependent psychiatric conditions such as posttraumatic stress disorder. Although CREB activation is a common trigger, expression of these individual signs appears to involve divergent downstream mechanisms.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Extinción Psicológica/fisiología , Miedo/fisiología , Núcleo Accumbens/metabolismo , Recompensa , Estrés Psicológico/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiopatología , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/fisiopatología , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología
19.
Psychopharmacology (Berl) ; 213(4): 697-706, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20922362

RESUMEN

RATIONALE: Anxiety disorders affect 18% of the United States adult population annually. Recent surges in the diagnosis of posttraumatic stress disorder (PTSD) from combat-exposed veterans have prompted an urgent need to understand the pathophysiology underlying this debilitating condition. OBJECTIVES: Anxiety and fear responses are partly modulated by gamma aminobutyric acid type A (GABA(A)) receptor-mediated synaptic inhibition; benzodiazepines potentiate GABAergic inhibition and are effective anxiolytics. Many genetically modified mouse lines are generated and/or maintained on the C57BL/6J background, a strain where manipulation of anxiety-like behavior using benzodiazepines is difficult. Fear-potentiated startle (FPS), a test of conditioned fear, is a useful preclinical tool to study PTSD-like responses but has been difficult to establish in C57BL/6J mice. METHODS: We modified several FPS experimental parameters and developed a paradigm to assess conditioned fear in C57BL/6J mice. The 6-day protocol consisted of three startle Acclimation days, a Pre-Test day followed by Training and Testing for FPS. Subject responses to the effects of three benzodiazepines were also examined. RESULTS: C57BL/6J mice had low levels of unconditioned fear assessed during Pre-Test (15-18%) but showed robust FPS (80-120%) during the Test session. Conditioned fear responses extinguished over repeated test sessions. Administration of the benzodiazepines alprazolam (0.5 and 1 mg/kg, i.p.), chlordiazepoxide (5 and 10 mg/kg, i.p.), and diazepam (1, 2, and 4 mg/kg, i.p.) significantly reduced FPS to Pre-Test levels. CONCLUSIONS: We used a modified and pharmacologically-validated paradigm to assess FPS in mice thereby providing a powerful tool to examine the neurobiology of PTSD in genetic models of anxiety generated on the C57BL/6J background.


Asunto(s)
Ansiolíticos/farmacología , Benzodiazepinas/farmacología , Miedo/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Alprazolam/administración & dosificación , Alprazolam/farmacología , Animales , Ansiolíticos/administración & dosificación , Benzodiazepinas/administración & dosificación , Clordiazepóxido/administración & dosificación , Clordiazepóxido/farmacología , Diazepam/administración & dosificación , Diazepam/farmacología , Relación Dosis-Respuesta a Droga , Moduladores del GABA/administración & dosificación , Moduladores del GABA/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo
20.
Cell ; 137(4): 761-72, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19450521

RESUMEN

The transient receptor potential channel 5 (TRPC5) is predominantly expressed in the brain where it can form heterotetrameric complexes with TRPC1 and TRPC4 channel subunits. These excitatory, nonselective cationic channels are regulated by G protein, phospholipase C-coupled receptors. Here, we show that TRPC5(-/-) mice exhibit diminished innate fear levels in response to innately aversive stimuli. Moreover, mutant mice exhibited significant reductions in responses mediated by synaptic activation of Group I metabotropic glutamate and cholecystokinin 2 receptors in neurons of the amygdala. Synaptic strength at afferent inputs to the amygdala was diminished in P10-P13 null mice. In contrast, baseline synaptic transmission, membrane excitability, and spike timing-dependent long-term potentiation at cortical and thalamic inputs to the amygdala were largely normal in older null mice. These experiments provide genetic evidence that TRPC5, activated via G protein-coupled neuronal receptors, has an essential function in innate fear.


Asunto(s)
Amígdala del Cerebelo/fisiología , Miedo , Canales Catiónicos TRPC/fisiología , Animales , Encéfalo , Condicionamiento Psicológico , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Noqueados , Receptores de Glutamato Metabotrópico/fisiología , Transmisión Sináptica , Canales Catiónicos TRPC/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...