Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Learn Mem ; 29(3): 83-92, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35169047

RESUMO

NMDA receptors (NMDARs) and AMPA receptors (AMPARs) in amygdala nuclei and the dorsal hippocampus (dHipp) are critical for fear conditioning. Enhancements in synaptic AMPAR expression in amygdala nuclei and the dHipp are critical for fear conditioning, with some studies observing changes in AMPAR expression across many neurons in these brain regions. Whether similar changes occur in other nodes of the fear circuit (e.g., ventral hippocampus [vHipp]) or changes in NMDAR expression in the fear circuit occur with fear conditioning have not been sufficiently examined. To address this we used near-infrared immunohistochemistry (IHC) to measure AMPAR and NMDAR subunit expression in several nodes of the fear circuit. Long-term changes in GluR1 and GluR2 expression in the ventral hippocampus (vHipp) and anterior cingulate cortex (ACC), enhanced NR2A expression in amygdala nuclei, and changes in the ratio of GluR1/NR2A and GluR2/NR2A in the dHipp was observed with fear conditioning. Most of these changes were dependent on protein synthesis during fear conditioning and were not observed immediately after fear conditioning. The results of the study suggest that global changes in AMPARs and NMDARs occur in multiple nodes within the fear circuit and raise the possibility that these changes contribute to fear memory. Further research examining how global changes in AMPAR, NMDAR, and AMPAR/NMDAR ratios within nodes of the fear circuit contribute to fear memory is needed.


Assuntos
Receptores de AMPA , Receptores de N-Metil-D-Aspartato , Medo/fisiologia , Hipocampo/metabolismo , Aprendizagem , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
2.
Int J Neuropsychopharmacol ; 24(3): 229-238, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33151288

RESUMO

BACKGROUND: A core symptom of posttraumatic stress disorder is persistent fear memory, which can be defined as fear memory that is resistant to updating, inhibition, or extinction. posttraumatic stress disorder emerges after traumatic stress exposure, but neurobiological mechanisms via which traumatic stress leads to persistent fear memory are not well defined. Akt signaling within the amygdala (Amy) is enhanced with traumatic stress, and phosphatidylinositol kinase 3 (PI3K) activation of Akt within the basolateral Amy (BLA) has been implicated as critical to fear memory formation. These findings raise the possibility that traumatic stress enhances PI3K→Akt signaling in the BLA, which leads to persistent fear memory. METHODS: To test this hypothesis, rats were exposed to traumatic stress using the single prolonged stress model, and changes in Akt phosphorylation were assayed in the Amy at 0 and 30 minutes after fear conditioning (FC). In a separate experiment, we inhibited PI3K→Akt signaling in the BLA prior to FC and observed the effect this had on acquisition, expression, and extinction of FC in stressed and control rats. RESULTS: Enhanced Akt phosphorylation in the Amy at both time points was observed in stressed rats, but not in control rats. PI3K→Akt inhibition in the BLA had no effect on freezing in control rats but decreased freezing during extinction training and testing in stressed rats. CONCLUSION: These findings suggest that PI3K→Akt signaling in the BLA could be a mechanism via which traumatic stress leads to fear memory that is resistant to extinction.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Extinção Psicológica , Medo/fisiologia , Rememoração Mental , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transtornos de Estresse Pós-Traumáticos , Estresse Psicológico , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Complexo Nuclear Basolateral da Amígdala/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Masculino , Rememoração Mental/efeitos dos fármacos , Rememoração Mental/fisiologia , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transtornos de Estresse Pós-Traumáticos/metabolismo , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
3.
Learn Mem ; 26(10): 1-9, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31527186

RESUMO

Clinical and preclinical studies that have examined the neurobiology of persistent fear memory in posttraumatic stress disorder (PTSD) have focused on the medial prefrontal cortex, hippocampus, and amygdala. Sensory systems, the periaqueductal gray (PAG), and midline thalamic nuclei have been implicated in fear and extinction memory, but whether neural activity in these substrates is sensitive to traumatic stress (at baseline or during emotional learning and memory) remains unexplored. To address this, we used the single prolonged stress (SPS) model of traumatic stress. SPS and control rats were either subjected to fear conditioning (CS-fear) or presented with CSs alone (CS-only) during fear conditioning. All rats were then subjected to extinction training and testing. A subset of rats were euthanized after each behavioral stage and c-Fos and c-Jun used to measure neural activation in all substrates. SPS lowered c-Jun levels in the dorsomedial and lateral PAG at baseline, but the elevated c-Jun expression in the PAG during emotional learning and memory. SPS also altered c-Fos expression during fear and extinction learning/memory in midline thalamic nuclei. These findings suggest changes in neural function in the PAG and midline thalamic nuclei could contribute to persistent fear memory induced by traumatic stress. Interestingly, SPS effects were also observed in animals that never learned fear or extinction (i.e., CS-only). This raises the possibility that traumatic stress could have broader effects on the psychological function that are dependent on the PAG and midline thalamic nuclei.


Assuntos
Condicionamento Clássico/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Memória/fisiologia , Núcleos da Linha Média do Tálamo/fisiopatologia , Substância Cinzenta Periaquedutal/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transtornos de Estresse Traumático/fisiopatologia , Estresse Psicológico/fisiopatologia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Núcleos da Linha Média do Tálamo/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Ratos , Transtornos de Estresse Traumático/metabolismo , Estresse Psicológico/metabolismo
4.
Neurobiol Learn Mem ; 152: 71-79, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29783059

RESUMO

In classical fear conditioning, a neutral conditioned stimulus (CS) is paired with an aversive unconditioned stimulus (US), which leads to a fear memory. If the CS is repeatedly presented without the US after fear conditioning, the formation of an extinction memory occurs, which inhibits fear memory expression. A previous study has demonstrated that selective cholinergic lesions in the medial septum and vertical limb of the diagonal bands of Broca (MS/vDBB) prior to fear and extinction learning disrupt contextual fear memory discrimination and acquisition of extinction memory. MS/vDBB cholinergic neurons project to a number of substrates that are critical for fear and extinction memory. However, it is currently unknown which of these efferent projections are critical for contextual fear memory discrimination and extinction memory. To address this, we induced cholinergic lesions in efferent targets of MS/vDBB cholinergic neurons. These included the dorsal hippocampus (dHipp), ventral hippocampus (vHipp), medial prefrontal cortex (mPFC), and in the mPFC and dHipp combined. None of these lesion groups exhibited deficits in contextual fear memory discrimination or extinction memory. However, vHipp cholinergic lesions disrupted auditory fear memory. Because MS/vDBB cholinergic neurons are the sole source of acetylcholine in the vHipp, these results suggest that MS/vDBB cholinergic input to the vHipp is critical for auditory fear memory. Taken together with previous findings, the results of this study suggest that MS/vDBB cholinergic neurons are critical for fear and extinction memory, though further research is needed to elucidate the role of MS/vDBB cholinergic neurons in these types of emotional memory.


Assuntos
Neurônios Colinérgicos/fisiologia , Feixe Diagonal de Broca/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Hipocampo/fisiologia , Memória/fisiologia , Núcleos Septais/fisiologia , Estimulação Acústica , Animais , Condicionamento Clássico , Masculino , Vias Neurais/fisiologia , Ratos Sprague-Dawley
5.
Learn Mem ; 23(12): 689-698, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27918273

RESUMO

Single prolonged stress (SPS) has been used to examine mechanisms via which stress exposure leads to post-traumatic stress disorder symptoms. SPS induces fear extinction retention deficits, but neural circuits critical for mediating these deficits are unknown. To address this gap, we examined the effect of SPS on neural activity in brain regions critical for extinction retention (i.e., fear extinction circuit). These were the ventral hippocampus (vHipp), dorsal hippocampus (dHipp), basolateral amygdala (BLA), prelimbic cortex (PL), and infralimbic cortex (IL). SPS or control rats were fear conditioned then subjected to extinction training and testing. Subsets of rats were euthanized after extinction training, extinction testing, or immediate removal from the housing colony (baseline condition) to assay c-Fos levels (measure of neural activity) in respective brain region. SPS induced extinction retention deficits. During extinction training SPS disrupted enhanced IL neural activity and inhibited BLA neural activity. SPS also disrupted inhibited BLA and vHipp neural activity during extinction testing. Statistical analyses suggested that SPS disrupted functional connectivity within the dHipp during extinction training and increased functional connectivity between the BLA and vHipp during extinction testing. Our findings suggest that SPS induces extinction retention deficits by disrupting both excitatory and inhibitory changes in neural activity within the fear extinction circuit and inducing changes in functional connectivity within the Hipp and BLA.


Assuntos
Tonsila do Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Extinção Psicológica/fisiologia , Medo/fisiologia , Hipocampo/metabolismo , Estresse Psicológico/metabolismo , Tonsila do Cerebelo/patologia , Animais , Córtex Cerebral/patologia , Eletrochoque , Análise Fatorial , Reação de Congelamento Cataléptica , Hipocampo/patologia , Imuno-Histoquímica , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Vias Neurais/metabolismo , Vias Neurais/patologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Sprague-Dawley , Estresse Psicológico/patologia
6.
Hippocampus ; 26(6): 718-26, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26606423

RESUMO

Previous research has shown that the ventral medial prefrontal cortex (vmPFC) and hippocampus (Hipp) are critical for extinction memory. Basal forebrain (BF) cholinergic input to the vmPFC and Hipp is critical for neural function in these substrates, which suggests BF cholinergic neurons may be critical for extinction memory. In order to test this hypothesis, we applied cholinergic lesions to different regions of the BF and observed the effects these lesions had on extinction memory. Complete BF cholinergic lesions induced contextual fear memory generalization, and this generalized fear was resistant to extinction. Animals with complete BF cholinergic lesions could not acquire cued fear extinction. Restricted cholinergic lesions in the medial septum and vertical diagonal bands of Broca (MS/vDBB) mimicked the effects that BF cholinergic lesions had on contextual fear memory generalization and acquisition of fear extinction. Cholinergic lesions in the horizontal diagonal band of Broca and nucleus basalis (hDBB/NBM) induced a small deficit in extinction of generalized contextual fear memory with no accompanying deficits in cued fear extinction. The results of this study reveal that MS/vDBB cholinergic neurons are critical for inhibition and extinction of generalized contextual fear memory, and via this process, may be critical for acquisition of cued fear extinction. Further studies delineating neural circuits and mechanisms through which MS/vDBB cholinergic neurons facilitate these emotional memory processes are needed. © 2015 Wiley Periodicals, Inc.


Assuntos
Neurônios Colinérgicos/fisiologia , Feixe Diagonal de Broca/fisiopatologia , Medo/fisiologia , Generalização Psicológica/fisiologia , Memória/fisiologia , Septo do Cérebro/fisiopatologia , Animais , Neurônios Colinérgicos/patologia , Condicionamento Clássico/fisiologia , Sinais (Psicologia) , Feixe Diagonal de Broca/patologia , Extinção Psicológica/fisiologia , Reação de Congelamento Cataléptica , Imuno-Histoquímica , Masculino , Testes Neuropsicológicos , Ratos Sprague-Dawley , Septo do Cérebro/patologia
7.
Neurobiol Learn Mem ; 133: 39-52, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27264248

RESUMO

Cholinergic input to the neocortex, dorsal hippocampus (dHipp), and basolateral amygdala (BLA) is critical for neural function and synaptic plasticity in these brain regions. Synaptic plasticity in the neocortex, dHipp, ventral Hipp (vHipp), and BLA has also been implicated in fear and extinction memory. This finding raises the possibility that basal forebrain (BF) cholinergic neurons, the predominant source of acetylcholine in these brain regions, have an important role in mediating fear and extinction memory. While empirical studies support this hypothesis, there are interesting inconsistencies among these studies that raise questions about how best to define the role of BF cholinergic neurons in fear and extinction memory. Nucleus basalis magnocellularis (NBM) cholinergic neurons that project to the BLA are critical for fear memory and contextual fear extinction memory. NBM cholinergic neurons that project to the neocortex are critical for cued and contextual fear conditioned suppression, but are not critical for fear memory in other behavioral paradigms and in the inhibitory avoidance paradigm may even inhibit contextual fear memory formation. Medial septum and diagonal band of Broca cholinergic neurons are critical for contextual fear memory and acquisition of cued fear extinction. Thus, even though the results of previous studies suggest BF cholinergic neurons modulate fear and extinction memory, inconsistent findings among these studies necessitates more research to better define the neural circuits and molecular processes through which BF cholinergic neurons modulate fear and extinction memory. Furthermore, studies determining if BF cholinergic neurons can be manipulated in such a manner so as to treat excessive fear in anxiety disorders are needed.


Assuntos
Prosencéfalo Basal/fisiologia , Núcleo Basal de Meynert/fisiologia , Complexo Nuclear Basolateral da Amígdala/fisiologia , Neurônios Colinérgicos/fisiologia , Condicionamento Clássico/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Rememoração Mental/fisiologia , Animais , Humanos
8.
Neurosci Biobehav Rev ; 159: 105569, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38309497

RESUMO

Contextual and spatial systems facilitate changes in emotional memory regulation brought on by traumatic stress. Cholinergic basal forebrain (chBF) neurons provide input to contextual/spatial systems and although chBF neurons are important for emotional memory, it is unknown how they contribute to the traumatic stress effects on emotional memory. Clusters of chBF neurons that project to the prefrontal cortex (PFC) modulate fear conditioned suppression and passive avoidance, while clusters of chBF neurons that project to the hippocampus (Hipp) and PFC (i.e. cholinergic medial septum and diagonal bands of Broca (chMS/DBB neurons) are critical for fear extinction. Interestingly, neither Hipp nor PFC projecting chMS/DBB neurons are critical for fear extinction. The retrosplenial cortex (RSC) is a contextual/spatial memory system that receives input from chMS/DBB neurons, but whether this chMS/DBB-RSC circuit facilitates traumatic stress effects on emotional memory remain unexplored. Traumatic stress leads to neuroinflammation and the buildup of reactive oxygen species. These two molecular processes may converge to disrupt chBF circuits enhancing the impact of traumatic stress on emotional memory.


Assuntos
Prosencéfalo Basal , Extinção Psicológica , Humanos , Extinção Psicológica/fisiologia , Medo/fisiologia , Hipocampo/fisiologia , Neurônios Colinérgicos
9.
Eur J Neurosci ; 37(6): 901-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23279008

RESUMO

Data from preclinical and clinical studies have implicated the norepinephrine system in the development and maintenance of post-traumatic stress disorder. The primary source of norepinephrine in the forebrain is the locus coeruleus (LC); however, LC activity cannot be directly measured in humans, and previous research has often relied upon peripheral measures of norepinephrine to infer changes in central LC-norepinephrine function. To directly assess LC-norepinephrine function, we measured single-unit activity of LC neurons in a validated rat model of post-traumatic stress disorder - single prolonged stress (SPS). We also examined tyrosine hydroxylase mRNA levels in the LC of SPS and control rats as an index of norepinephrine utilisation. For electrophysiological recordings, 92 LC neurons were identified from 19 rats (SPS, 12; control, 7), and spontaneous and evoked responses to a noxious event (paw compression) were recorded. Baseline and restraint stress-evoked tyrosine hydroxylase mRNA expression levels were measured in SPS and control rats (n = 16 per group) in a separate experiment. SPS rats showed lower spontaneous activity but higher evoked responses, leading to an enhanced signal-to-noise ratio of LC neurons, accompanied by impaired recovery from post-stimulus inhibition. In concert, tyrosine hydroxylase mRNA expression in the LC of SPS rats tended to be lower at baseline, but was exaggerated following restraint stress. These data demonstrate persistent changes in LC function following stress/trauma in a rat model of post-traumatic stress, as measured by differences in both the electrophysiological properties of LC neurons and tyrosine hydroxylase mRNA transcription.


Assuntos
Locus Cerúleo/metabolismo , Norepinefrina/metabolismo , Estresse Psicológico/fisiopatologia , Potenciais de Ação , Animais , Locus Cerúleo/citologia , Locus Cerúleo/fisiopatologia , Masculino , Inibição Neural , Neurônios/metabolismo , Neurônios/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Restrição Física , Transtornos de Estresse Pós-Traumáticos/metabolismo , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Estresse Psicológico/metabolismo , Transcrição Gênica , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
10.
Learn Mem ; 19(2): 43-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22240323

RESUMO

Clinical research has linked post-traumatic stress disorder (PTSD) with deficits in fear extinction. However, it is not clear whether these deficits result from stress-related changes in the acquisition or retention of extinction or in the regulation of extinction memories by context, for example. In this study, we used the single prolonged stress (SPS) animal model of PTSD and fear conditioning procedures to examine the effects of prior traumatic stress on the acquisition, retention, and context-specificity of extinction. SPS administered one week prior to fear conditioning had no effect on the acquisition of fear conditioning or extinction but disrupted the retention of extinction memories for both contextual and cued fear. This SPS effect required a post-stress incubation period to manifest. The results demonstrate that SPS disrupts extinction retention, leading to enhanced fear renewal; further research is needed to identify the neurobiological processes through which SPS induces these effects.


Assuntos
Extinção Psicológica/fisiologia , Medo/psicologia , Transtornos da Memória/etiologia , Retenção Psicológica/fisiologia , Estresse Psicológico/complicações , Estimulação Acústica , Análise de Variância , Animais , Condicionamento Clássico/fisiologia , Sinais (Psicologia) , Modelos Animais de Doenças , Eletrochoque/efeitos adversos , Reação de Congelamento Cataléptica/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley
11.
Psychopharmacology (Berl) ; 240(5): 1049-1061, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36879072

RESUMO

RATIONALE: Traumatic stress leads to persistent fear, which is a core feature of post-traumatic stress disorder (PTSD). Women are more likely than men to develop PTSD after trauma exposure, which suggests women are differentially sensitive to traumatic stress. However, it is unclear how this differential sensitivity manifests. Cyclical changes in vascular estrogen release could be a contributing factor where levels of vascular estrogens (and activation of estrogen receptors) at the time of traumatic stress alter the impact of traumatic stress. METHODS: To examine this, we manipulated estrogen receptors at the time of stress and observed the effect this had on fear and extinction memory (within the single prolonged stress (SPS) paradigm) in female rats. In all experiments, freezing and darting were used to measure fear and extinction memory. RESULTS: In Experiment 1, SPS enhanced freezing during extinction testing, and this effect was blocked by nuclear estrogen receptor antagonism prior to SPS. In Experiment 2, SPS decreased conditioned freezing during the acquisition and testing of extinction. Administration of 17ß-estradiol altered freezing in control and SPS animals during the acquisition of extinction, but this treatment had no effect on freezing during the testing of extinction memory. In all experiments, darting was only observed to footshock onset during fear conditioning. CONCLUSION: The results suggest multiple behaviors (or different behavioral paradigms) are needed to characterize the nature of traumatic stress effects on emotional memory in female rats and that nuclear estrogen receptor antagonism prior to SPS blocks SPS effects on emotional memory in female rats.


Assuntos
Extinção Psicológica , Transtornos de Estresse Pós-Traumáticos , Ratos , Feminino , Animais , Extinção Psicológica/fisiologia , Receptores de Estrogênio , Ratos Sprague-Dawley , Medo/fisiologia , Transtornos de Estresse Pós-Traumáticos/psicologia , Estresse Psicológico , Modelos Animais de Doenças
12.
Behav Brain Res ; 452: 114579, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37451551

RESUMO

The incidence of post traumatic stress disorder (PTSD) is greater in women than men, but mechanisms via which this difference manifests remain under explored. The single prolonged stress (SPS) rodent model of traumatic stress has been used to identify mechanisms through which traumatic stress leads to deficits in retaining extinction (a core PTSD symptom), but has been mostly utilized in male model systems. Recent studies have observed that SPS leads to changes in persistent fear memory in female rats, though these results are variable. This variability could be driven by changes in behavioral strategy in females during extinction, but this possibility has not been sufficiently explored. To address this, we examined the impact of SPS on freezing and avoidance (a core PTSD symptom) during extinction in male and female rats. In male rats, SPS enhanced acquisition of conditioned freezing, but did not enhance freezing during extinction training or testing. SPS also decreased avoidance during extinction training, but not extinction testing. In female rats, SPS had no impact on conditioned freezing. Avoidance was not observed in control rats, but emerged in SPS/female rats during extinction testing. Furthermore, avoidance was negatively correlated with freezing in female rats (high avoidance associated with lower freezing), but this relationship was disrupted with SPS. The results suggest that introducing avoidance during extinction negates SPS effects on extinction retention in male and female rats, control/female rats engage in avoidance to regulate fear expression, and this relationship is disrupted with SPS.


Assuntos
Extinção Psicológica , Transtornos de Estresse Pós-Traumáticos , Ratos , Feminino , Masculino , Animais , Extinção Psicológica/fisiologia , Ratos Sprague-Dawley , Medo/fisiologia , Transtornos de Estresse Pós-Traumáticos/metabolismo , Modelos Animais de Doenças , Estresse Psicológico/metabolismo
13.
Neurobiol Learn Mem ; 97(4): 386-92, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22446013

RESUMO

It has been well established that expression of conditioned fear is context independent, but the context dependency of unconditioned fear expression has rarely been explored. A recent study reported that unconditioned freezing in rats is enhanced in a familiar context, which suggests that unconditioned fear expression can be modulated by contextual processing. In order to further explore this possibility we examined unconditioned freezing in novel, familiar, and appetitive contexts; and attempted to identify brain regions critical for context-related changes in unconditioned freezing by measuring c-Fos mRNA levels in emotional circuits. Unconditioned freezing was enhanced in the appetitive context, and this enhancement was accompanied by increased c-Fos mRNA expression in the medial amygdala and hippocampus, but attenuated expression in the medial prefrontal cortex. In the appetitive context, expectation of a reward coupled with detection of threat may have enhanced unconditioned fear expression, which suggests that unconditioned fear expression can be modulated by contextual factors. Context-related expectancy mismatch may explain the enhancement of unconditioned fear expression seen in this study and warrants further examination.


Assuntos
Encéfalo/metabolismo , Condicionamento Psicológico/fisiologia , Medo/fisiologia , Reação de Congelamento Cataléptica/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo , Tonsila do Cerebelo/metabolismo , Animais , Corticosterona/metabolismo , Hipocampo/metabolismo , Masculino , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Tiazóis
14.
Front Behav Neurosci ; 15: 766505, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34955778

RESUMO

Post-traumatic stress disorder (PTSD) is a debilitating psychiatric disorder with a high economic burden. Two risk factors for increasing the chances of developing PTSD are sex (being female) and early life stress. These risk factors suggest that early life stress-induced changes and sex differences in emotional circuits and neuroendocrinological systems lead to susceptibility to traumatic stress. Exploring mechanisms via which stress leads to specific effects can be accomplished in animal models, but reliable animal models that allow for an examination of how early life stress interacts with sex to increase susceptibility to traumatic stress is lacking. To address this, we examined the effects of early life stress [using the maternal separation (MS) model] and late adolescence/early adult traumatic stress [using the single prolonged stress (SPS) model] on startle reactivity, anxiety-like behavior in the open field (OF), and basal corticosterone levels in male and female rats. Female rats exposed to MS and SPS (MS/SPS) showed enhanced startle reactivity relative to MS/control female rats. Enhanced startle reactivity was not observed in MS/SPS male rats. Instead, non-maternally separated male rats that were exposed to SPS showed enhanced startle reactivity relative to controls. Female rats had enhanced locomotor activity in the OF and higher basal corticosterone levels in comparison to males, but measures in the OF and basal corticosterone were not affected by MS or SPS. Overall the results suggest that the combined MS and SPS models can be used to explore how changes in maternal care during infancy lead to sex differences in sensitivity to the effects of traumatic stress as adolescents and adults.

15.
Front Behav Neurosci ; 15: 652636, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054443

RESUMO

Posttraumatic stress disorder (PTSD) is a mental health condition triggered by experiencing or witnessing a terrifying event that can lead to lifelong burden that increases mortality and adverse health outcomes. Yet, no new treatments have reached the market in two decades. Thus, screening potential interventions for PTSD is of high priority. Animal models often serve as a critical translational tool to bring new therapeutics from bench to bedside. However, the lack of concordance of some human clinical trial outcomes with preclinical animal efficacy findings has led to a questioning of the methods of how animal studies are conducted and translational validity established. Thus, we conducted a systematic review to determine methodological variability in studies that applied a prominent animal model of trauma-like stress, single prolonged stress (SPS). The SPS model has been utilized to evaluate a myriad of PTSD-relevant outcomes including extinction retention. Rodents exposed to SPS express an extinction retention deficit, a phenotype identified in humans with PTSD, in which fear memory is aberrantly retained after fear memory extinction. The current systematic review examines methodological variation across all phases of the SPS paradigm, as well as strategies for behavioral coding, data processing, statistical approach, and the depiction of data. Solutions for key challenges and sources of variation within these domains are discussed. In response to methodological variation in SPS studies, an expert panel was convened to generate methodological considerations to guide researchers in the application of SPS and the evaluation of extinction retention as a test for a PTSD-like phenotype. Many of these guidelines are applicable to all rodent paradigms developed to model trauma effects or learned fear processes relevant to PTSD, and not limited to SPS. Efforts toward optimizing preclinical model application are essential for enhancing the reproducibility and translational validity of preclinical findings, and should be conducted for all preclinical psychiatric research models.

16.
J Vis Exp ; (147)2019 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-31180361

RESUMO

Neuroscience is the study of how cells in the brain mediate various functions. Measuring protein expression in neurons and glia is critical for the study of neuroscience as cellular function is determined by the composition and activity of cellular proteins. In this article, we describe how immunocytochemistry can be combined with near-infrared high-resolution scanning to provide a semi-quantitative measure of protein expression in distinct brain regions. This technique can be used for single or double protein expression in the same brain region. Measuring proteins in this fashion can be used to obtain a relative change in protein expression with an experimental manipulation, molecular signature of learning and memory, activity in molecular pathways, and neural activity in multiple brain regions. Using the correct proteins and statistical analysis, functional connectivity among brain regions can be determined as well. Given the ease of implementing immunocytochemistry in a laboratory, using immunocytochemistry with near-infrared high-resolution scanning can expand the ability of the neuroscientist to examine neurobiological processes at a systems level.


Assuntos
Encéfalo/diagnóstico por imagem , Fluorescência , Raios Infravermelhos/uso terapêutico , Proteínas/metabolismo , Cintilografia/métodos , Animais , Encéfalo/patologia , Humanos , Roedores
17.
Behav Neurosci ; 122(3): 601-10, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18513130

RESUMO

Previous studies have demonstrated that corticopetal cholinergic lesions applied to the nucleus basalis magnocellularis and substantia innominata (NBM/SI) attenuate operant suppression induced by aversive events. However, these lesions have no effect on open-arm behavior in the elevated plus-maze or changes in startle reactivity induced by bright light. This raises the possibility that NBM/SI corticopetal cholinergic lesions alter operant behavior and/or appetitive state, as opposed to the aversive state operant suppression is supposed to index. To address this concern, the authors documented the effect of NBM/SI corticopetal cholinergic lesions on freezing induced by a component of fox feces (2,4,5-trimethylthiazoline [TMT]), a paradigm that does not involve food deprivation or operant performance. TMT presentation induced freezing behavior, and this effect was attenuated by NBM/SI corticopetal cholinergic lesions. Because predator odor presentation, but not presentation of a predator, induces defense behaviors that are sensitive to anxiolytic drugs, the results of the study suggest that NBM/SI corticopetal cholinergic lesions attenuate anxiety-like states.


Assuntos
Acetilcolina/metabolismo , Núcleo Basal de Meynert/lesões , Reação de Congelamento Cataléptica/efeitos dos fármacos , Odorantes , Substância Inominada/lesões , Acetilcolinesterase/metabolismo , Animais , Núcleo Basal de Meynert/metabolismo , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Contagem de Células , Colina O-Acetiltransferase/metabolismo , Reação de Congelamento Cataléptica/fisiologia , Masculino , Modelos Biológicos , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Ratos , Ratos Sprague-Dawley , Substância Inominada/metabolismo
18.
Brain Res ; 1227: 142-52, 2008 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-18619423

RESUMO

Previous research has demonstrated that nucleus basalis magnocellularis (nbm) corticopetal cholinergic neurons modulate anxiety-like states, but cortical modulation by these neurons during anxiety-like states has not been characterized. In order to address this, we documented the effect of nbm corticopetal cholinergic lesions on cortical activity in direct (prefrontal cortex) and indirect (retrosplenial cortex) targets of nbm corticopetal cholinergic neurons during footshock induced operant suppression. The gamma/delta ratio and theta were used as indices of cortical activity, because these components of the electroencephalogram (EEG) are sensitive to basal forebrain corticopetal cholinergic modulation. During operant suppression, increases in the gamma/delta ratio and augmented theta were observed in both cortical EEGs. Lesions attenuated operant suppression and the gamma/delta ratio, but had no effect on increased theta. The effect of nbm corticopetal cholinergic lesions on the gamma/delta ratio was driven by the effect of the lesions on the delta band. The results of the study demonstrate that during anxiety-like states 1) decreases in delta reflect the action of nbm corticopetal cholinergic neurons, 2) nbm corticopetal cholinergic neurons alter neural processes in direct and indirect cortical targets, and 3) cortical theta is not dependent on the integrity of nbm corticopetal cholinergic neurons.


Assuntos
Ansiedade/fisiopatologia , Núcleo Basal de Meynert/fisiologia , Comportamento Animal/fisiologia , Córtex Cerebral/fisiologia , Fibras Colinérgicas/fisiologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/toxicidade , Núcleo Basal de Meynert/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Fibras Colinérgicas/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Eletroencefalografia/efeitos dos fármacos , Eletrochoque/métodos , Imunotoxinas/administração & dosagem , Imunotoxinas/toxicidade , Masculino , Microinjeções , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiologia , Ratos , Ratos Sprague-Dawley , Proteínas Inativadoras de Ribossomos Tipo 1/administração & dosagem , Proteínas Inativadoras de Ribossomos Tipo 1/toxicidade , Saporinas
19.
PLoS One ; 13(12): e0205144, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30532228

RESUMO

Glucocorticoid receptors (GRs) shuttle from the cytoplasm (cy) to the nucleus (nu) when bound with glucocorticoids (i.e. GR internalization) and alter transcriptional activity. GR activation within the fear circuit has been implicated in fear memory and post traumatic stress disorder (PTSD). However, no study to date has characterized GR internalization within the fear circuit during fear memory formation or examined how traumatic stress impacts this process. To address this, we assayed cy and nu GR levels at baseline and after auditory fear conditioning (FC) in the single prolonged stress (SPS) model of PTSD. Cy and nu GRs within the medial prefrontal cortex (mPFC), dorsal hippocampus (dHipp), ventral hippocampus (vHipp), and amygdala (AMY) were assayed using western blot. The distribution of GR in the cy and nu (at baseline and after FC) was varied across individual nodes of the fear circuit. At baseline, SPS enhanced cyGRs in the dHipp, but decreased cyGRs in the AMY. FC only enhanced GR internalization in the AMY and this effect was attenuated by SPS exposure. SPS also decreased cyGRs in the dHipp after FC. The results of this study suggests that GR internalization is varied across the fear circuit, which in turn suggests GR activation is selectively regulated within individual nodes of the fear circuit. The findings also suggest that changes in GR dynamics in the dHipp and AMY modulate the enhancing effect SPS has on fear memory persistence.


Assuntos
Encéfalo/metabolismo , Medo , Memória , Receptores de Glucocorticoides/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Transtornos de Estresse Pós-Traumáticos/patologia , Transtornos de Estresse Pós-Traumáticos/fisiopatologia
20.
Behav Brain Res ; 341: 189-197, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29292158

RESUMO

Neural circuits via which stress leads to disruptions in fear extinction is often explored in animal stress models. Using the single prolonged stress (SPS) model of post traumatic stress disorder and the immediate early gene (IEG) c-Fos as a measure of neural activity, we previously identified patterns of neural activity through which SPS disrupts extinction retention. However, none of these stress effects were specific to fear or extinction learning and memory. C-Jun is another IEG that is sometimes regulated in a different manner to c-Fos and could be used to identify emotional learning/memory specific patterns of neural activity that are sensitive to SPS. Animals were either fear conditioned (CS-fear) or presented with CSs only (CS-only) then subjected to extinction training and testing. C-Jun was then assayed within neural substrates critical for extinction memory. Inhibited c-Jun levels in the hippocampus (Hipp) and enhanced functional connectivity between the ventromedial prefrontal cortex (vmPFC) and basolateral amygdala (BLA) during extinction training was disrupted by SPS in the CS-fear group only. As a result, these effects were specific to emotional learning/memory. SPS also disrupted inhibited Hipp c-Jun levels, enhanced BLA c-Jun levels, and altered functional connectivity among the vmPFC, BLA, and Hipp during extinction testing in SPS rats in the CS-fear and CS-only groups. As a result, these effects were not specific to emotional learning/memory. Our findings suggest that SPS disrupts neural activity specific to extinction memory, but may also disrupt the retention of fear extinction by mechanisms that do not involve emotional learning/memory.


Assuntos
Condicionamento Psicológico/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/enzimologia , Estresse Psicológico/enzimologia , Animais , Complexo Nuclear Basolateral da Amígdala/enzimologia , Complexo Nuclear Basolateral da Amígdala/patologia , Hipocampo/enzimologia , Hipocampo/patologia , Imuno-Histoquímica , Masculino , Neurônios/patologia , Córtex Pré-Frontal/enzimologia , Córtex Pré-Frontal/patologia , Ratos Sprague-Dawley , Estresse Psicológico/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA