Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-39000204

RESUMO

Fear conditioning evokes a physiologic release of glucocorticoids that assists learning. As a cochaperone in the glucocorticoid receptor complex, FKBP51 modulates stress-induced glucocorticoid signaling and may influence conditioned fear responses. This study combines molecular and behavioral approaches to examine whether locally reducing FKBP51 expression in the ventral hippocampus is sufficient to affect fear-related behaviors. We hypothesized that reducing FKBP51 expression in the VH would increase glucocorticoid signaling to alter auditory fear conditioning. Adult male rats were injected with an adeno-associated virus (AAV) vector expressing short hairpin - RNAs (shRNA) targeting FKBP5 into the ventral hippocampus to reduce FKBP5 levels or a control AAV. Infusion of FKBP5-shRNA into the ventral hippocampus decreased auditory fear acquisition and recall. Although animals injected with FKBP5-shRNA showed less freezing during extinction recall, the difference was due to a reduced fear recall rather than improved extinction. Reducing ventral hippocampus FKBP51 did not affect exploratory behavior in either the open field test or the elevated zero maze test but did increase passive behavior in the forced swim test, suggesting that the reduction in auditory fear recall was not due to more active responses to acute stress. Furthermore, lower ventral hippocampus FKBP51 levels did not alter corticosterone release in response to restraint stress, suggesting that the reduced fear recall was not due to lower corticosterone release. Our findings suggest FKBP51 in the ventral hippocampus plays a selective role in modulating fear-learning processes and passive behavioral responses to acute stress rather than hypothalamic-pituitary-adrenal axis reactivity or exploratory responses.


Assuntos
Medo , Hipocampo , Proteínas de Ligação a Tacrolimo , Animais , Masculino , Medo/fisiologia , Proteínas de Ligação a Tacrolimo/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Hipocampo/metabolismo , Ratos , Corticosterona/metabolismo , Corticosterona/sangue , Ratos Sprague-Dawley , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/genética , Receptores de Glucocorticoides/metabolismo , Extinção Psicológica/fisiologia
2.
Brain Behav Immun ; 110: 107-118, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36822379

RESUMO

Clinical evidence has linked increased peripheral pro-inflammatory cytokines with post-traumatic stress disorder (PTSD) symptoms. However, whether inflammation contributes to or is a consequence of PTSD is still unclear. Previous research shows that stress can activate purinergic P2X7 receptors (P2X7Rs) on microglia to induce inflammation and behavioral changes. In this investigation, we examined whether P2X7Rs contribute to the development of PTSD-like behaviors induced by single prolonged stress (SPS) exposure in rats. Consistent with the literature, exposing adult male and female rats to SPS produced a PTSD-like phenotype of impaired fear extinction and extinction of cue-induced center avoidance one week after exposure. Next, we examined if inflammation precedes the behavioral manifestations. Three days after SPS exposure, increased inflammatory cytokines were found in the blood and hippocampal microglia showed increased expression of the P2X7R, IL-1ß, and TNF-α, suggesting increased peripheral and central inflammation before the onset of impaired fear extinction. In addition, SPS-exposed animals with impaired fear extinction recall also had more Iba1-positive microglia expressing the P2X7R in the ventral hippocampus. To determine whether P2X7Rs contribute to the PTSD-related behaviors induced by SPS exposure, we gave ICV infusions of the P2X7R antagonist, A-438079, for one week starting the day of SPS exposure. Blocking P2X7Rs prevented the SPS-induced impaired fear extinction and extinction of cue-induced center avoidance in male and female rats, suggesting that SPS activates P2X7Rs which increase inflammation to produce a PTSD-like phenotype.


Assuntos
Transtornos de Estresse Pós-Traumáticos , Ratos , Masculino , Feminino , Animais , Transtornos de Estresse Pós-Traumáticos/metabolismo , Extinção Psicológica/fisiologia , Medo/fisiologia , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Inflamação/metabolismo , Citocinas/metabolismo , Estresse Psicológico
3.
Adv Physiol Educ ; 47(4): 919-929, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37823189

RESUMO

Biomedical graduate students receive intensive training in their scientific area of interest yet need additional skills for successful scientific careers. Our aim was to promote team building, improve collaborations and enhance communication skills. An off-site yearly retreat was organized for the graduate students in our NIH-funded Research Initiative for Scientific Enhancement (RISE) graduate training program. Retreat themes were addressed through short presentations, case studies, live podcasts, webinars, focus groups, role-play, and breakout sessions with various team building exercises to practice communication skills and identify abilities, knowledge, values, and behaviors. Trainees gave short presentations and served as discussion leaders on topics related to the central theme. Expert guest speakers participated in discussion sessions with the trainees. Trainees evaluated the retreats at the end. A total of 48 trainees, 12 RISE Program faculty and staff, and 26 external speakers from industry, academia, media/journalism, the arts, psychology, and holistic medical fields participated over 9 years. The overall average benefit of the in-person retreats was rated 4.80 on a Likert scale of 1-5 by trainees. Trainees particularly enjoyed the informal interactions with program faculty, staff, and fellow trainees. They appreciated the opportunity to learn soft skills, such as interpersonal communication, conflict resolution, and leadership. Two additional retreats conducted virtually because of the COVID-19 pandemic were perceived as less beneficial. We conclude that off-site interactive retreats are a valuable tool for enhancing soft skills and a sense of team identity in a biomedical sciences graduate program, while covering important issues related to scientific careers.NEW & NOTEWORTHY Off-site interactive science-related retreats are a valuable tool for enhancing soft skills and sense of team identity in a biomedical sciences graduate program, while covering important issues related to pursuing a career in science. There are many perceived benefits, so we encourage other training programs to include a similar type of regular activity in students' training with the goal of improving trainee well-being and supporting their academic and research productivity.


Assuntos
Docentes , Pandemias , Humanos , Educação de Pós-Graduação em Medicina , Estudantes , Comunicação
4.
Neurobiol Learn Mem ; 169: 107117, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31765801

RESUMO

In rodents, the anterior cingulate (ACC), prelimbic (PL), and infralimbic cortex (IL) comprise the medial prefrontal cortex (mPFC). Through extensive connections with cortical and subcortical structures, the mPFC plays a key modulatory role in the neuronal circuits underlying associative fear and reward learning. In this article, we have compiled the evidence that associative learning induces plasticity in both the intrinsic and synaptic excitability of mPFC neurons to modulate conditioned fear and cocaine seeking behavior. The literature highlights the accumulating evidence that plasticity in the intrinsic excitability of mPFC neurons represents a major cellular mechanism that interacts with synaptic changes to alter the impact of the mPFC on fear and reward circuits.


Assuntos
Potenciais de Ação , Aprendizagem por Associação/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Condicionamento Clássico/fisiologia , Comportamento de Procura de Droga/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Humanos , Recompensa
5.
J Neuroinflammation ; 16(1): 262, 2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31829243

RESUMO

BACKGROUND: HIV-1-associated neurocognitive disorders (HAND) progression is related to continued inflammation despite undetectable viral loads and may be caused by early viral proteins expressed by latently infected cells. Astrocytes represent an HIV reservoir in the brain where the early viral neurotoxin negative factor (Nef) is produced. We previously demonstrated that astrocytic expression of Nef in the hippocampus of rats causes inflammation, macrophage infiltration, and memory impairment. Since these processes are affected by TGFß signaling pathways, and TGFß-1 is found at higher levels in the central nervous system of HIV-1+ individuals and is released by astrocytes, we hypothesized a role for TGFß-1 in our model of Nef neurotoxicity. METHODS: To test this hypothesis, we compared cytokine gene expression by cultured astrocytes expressing Nef or green fluorescent protein. To determine the role of Nef and a TGFßRI inhibitor on memory and learning, we infused astrocytes expressing Nef into the hippocampus of rats and then treated them daily with an oral dose of SD208 (10 mg/kg) or placebo for 7 days. During this time, locomotor activity was recorded in an open field and spatial learning tested in the novel location recognition paradigm. Postmortem tissue analyses of inflammatory and signaling molecules were conducted using immunohistochemistry and immunofluorescence. RESULTS: TGFß-1 was induced in cultures expressing Nef at 24 h followed by CCL2 induction which was prevented by blocking TGFßRI with SD208 (competitive inhibitor). Interestingly, Nef seems to change the TGFßRI localization as suggested by the distribution of the immunoreactivity. Nef caused a deficit in spatial learning that was recovered upon co-administration of SD208. Brain tissue from Nef-treated rats given SD208 showed reduced CCL2, phospho-SMAD2, cluster of differentiation 163 (CD163), and GFAP immunoreactivity compared to the placebo group. CONCLUSIONS: Consistent with our previous findings, rats treated with Nef showed deficits in spatial learning and memory in the novel location recognition task. In contrast, rats treated with Nef + SD208 showed better spatial learning suggesting that Nef disrupts memory formation in a TGFß-1-dependent manner. The TGFßRI inhibitor further reduced the induction of inflammation by Nef which was concomitant with decreased TGFß signaling. Our findings suggest that TGFß-1 signaling is an intriguing target to reduce neuroHIV.


Assuntos
Encéfalo/metabolismo , Quimiocina CCL2/biossíntese , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Receptor do Fator de Crescimento Transformador beta Tipo I/biossíntese , Aprendizagem Espacial/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/biossíntese , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/genética , Técnicas de Cocultura , Masculino , Pteridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Aprendizagem Espacial/efeitos dos fármacos , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
6.
Learn Mem ; 24(4): 145-152, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28298552

RESUMO

The factors influencing resiliency to the development of post-traumatic stress disorder (PTSD) remain to be elucidated. Clinical studies associate PTSD with polymorphisms of the FK506 binding protein 5 (FKBP5). However, it is unclear whether changes in FKBP5 expression alone could produce resiliency or susceptibility to PTSD-like symptoms. In this study, we used rats as an animal model to examine whether FKBP5 in the infralimbic (IL) or prelimbic (PL) medial prefrontal cortex regulates fear conditioning or extinction. First, we examined FKBP5 expression in IL and PL during fear conditioning or extinction. In contrast to the stable expression of FKBP5 seen in PL, FKBP5 expression in IL increased after fear conditioning and remained elevated even after extinction suggesting that IL FKBP5 levels may modulate fear conditioning or extinction. Consistent with this possibility, reducing basal FKBP5 expression via local infusion of FKBP5-shRNA into IL reduced fear conditioning. Furthermore, reducing IL FKBP5, after consolidation of the fear memory, enhanced extinction memory indicating that IL FKBP5 opposed formation of the extinction memory. Our findings demonstrate that lowering FKBP5 expression in IL is sufficient to both reduce fear acquisition and enhance extinction, and suggest that lower expression of FKBP5 in the ventral medial prefrontal cortex could contribute to resiliency to PTSD.


Assuntos
Condicionamento Psicológico/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Regulação da Expressão Gênica/fisiologia , Córtex Pré-Frontal/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Estimulação Acústica/efeitos adversos , Análise de Variância , Animais , Aprendizagem da Esquiva/fisiologia , Comportamento Exploratório/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Aprendizagem em Labirinto/fisiologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas de Ligação a Tacrolimo/genética , Fatores de Tempo
7.
J Neurosci ; 35(36): 12394-403, 2015 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-26354908

RESUMO

Adolescent rats are prone to impaired fear extinction, suggesting that mechanistic differences in extinction could exist in adolescent and adult rats. Since the infralimbic cortex (IL) is critical for fear extinction, we used PCR array technology to identify gene expression changes in IL induced by fear extinction in adolescent rats. Interestingly, the ephrin type B receptor 2 (EphB2), a tyrosine kinase receptor associated with synaptic development, was downregulated in IL after fear extinction. Consistent with the PCR array results, EphB2 levels of mRNA and protein were reduced in IL after fear extinction compared with fear conditioning, suggesting that EphB2 signaling in IL regulates fear extinction memory in adolescents. Finally, reducing EphB2 synthesis in IL with shRNA accelerated fear extinction learning in adolescent rats, but not in adult rats. These findings identify EphB2 in IL as a key regulator of fear extinction during adolescence, perhaps due to the increase in synaptic remodeling occurring during this developmental phase.


Assuntos
Tonsila do Cerebelo/metabolismo , Extinção Psicológica , Medo , Córtex Pré-Frontal/metabolismo , Receptor EphB2/metabolismo , Tonsila do Cerebelo/crescimento & desenvolvimento , Tonsila do Cerebelo/fisiologia , Animais , Masculino , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor EphB2/genética
8.
Neurobiol Learn Mem ; 130: 77-82, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26860438

RESUMO

Patients with posttraumatic stress disorder (PTSD) show hypo-active ventromedial prefrontal cortices (vmPFC) that correlate with their impaired ability to discriminate between safe and dangerous contexts and cues. Previously, we found that auditory fear conditioning depresses the excitability of neurons populating the homologous structure in rodents, the infralimbic cortex (IL). However, it is undetermined if IL depression was mediated by the cued or contextual information. The objective of this study was to examine whether contextual information was sufficient to depress IL neuronal excitability. After exposing rats to context-alone, pseudoconditioning, or contextual fear conditioning, we used whole-cell current-clamp recordings to examine the excitability of IL neurons in prefrontal brain slices. We found that contextual fear conditioning reduced IL neuronal firing in response to depolarizing current steps. In addition, neurons from contextual fear conditioned animals showed increased slow afterhyperpolarization potentials (sAHPs). Moreover, the observed changes in IL excitability correlated with contextual fear expression, suggesting that IL depression may contribute to the encoding of contextual fear.


Assuntos
Condicionamento Clássico/fisiologia , Medo/fisiologia , Sistema Límbico/fisiologia , Potenciais de Ação/fisiologia , Animais , Extinção Psicológica/fisiologia , Masculino , Neurônios/fisiologia , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley
9.
Brain Behav Immun ; 52: 169-177, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26520214

RESUMO

Patients with post-traumatic stress disorder (PTSD) tend to show signs of a relatively increased inflammatory state suggesting that activation of the immune system may contribute to the development of PTSD. In the present study, we tested whether activation of the innate immune system can disrupt acquisition or recall of auditory fear extinction using an animal model of PTSD. Male adolescent rats received auditory fear conditioning in context A. The next day, an intraperitoneal injection of lipopolysaccharide (LPS; 100 µg/kg) prior to auditory fear extinction in context B impaired acquisition and recall of extinction. LPS (100 µg/kg) given after extinction training did not impair extinction recall suggesting that LPS did not affect consolidation of extinction. In contrast to cued fear extinction, contextual fear extinction was not affected by prior injection of LPS (100 µg/kg). Although LPS also reduced locomotion, we could dissociate the effects of LPS on extinction and locomotion by using a lower dose of LPS (50 µg/kg) which impaired locomotion without affecting extinction. In addition, 15 h after an injection of 250 µg/kg LPS in adult rats, extinction learning and recall were impaired without affecting locomotion. A sub-chronic treatment with candesartan, an angiotensin II type 1 receptor blocker, prevented the LPS-induced impairment of extinction in adult rats. Our results demonstrate that activation of the innate immune system can disrupt auditory fear extinction in adolescent and adult animals. These findings also provide direction for clinical studies of novel treatments that modulate the innate immune system for stress-related disorders like PTSD.


Assuntos
Benzimidazóis/farmacologia , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Tetrazóis/farmacologia , Animais , Compostos de Bifenilo , Condicionamento Psicológico , Sinais (Psicologia) , Imunidade Inata/imunologia , Lipopolissacarídeos/farmacologia , Masculino , Rememoração Mental , Modelos Animais , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Transtornos de Estresse Pós-Traumáticos/imunologia , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/imunologia
10.
J Neurosci ; 33(17): 7184-93, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23616528

RESUMO

Studies suggest that plasticity in the infralimbic prefrontal cortex (IL) in rodents and its homolog in humans is necessary for inhibition of fear during the recall of fear extinction. The recall of extinction is impaired by locally blocking metabotropic glutamate receptor type 5 (mGluR5) activation in IL during extinction training. This finding suggests that mGluR5 stimulation may lead to IL plasticity needed for fear extinction. To test this hypothesis, we recorded AMPA and NMDA currents, AMPA receptor (AMPAR) rectification, and intrinsic excitability in IL pyramidal neurons in slices from trained rats using whole-cell patch-clamp recording. We observed that fear extinction increases the AMPA/NMDA ratio, consistent with insertion of AMPARs into IL synapses. In addition, extinction training increased inward rectification, suggesting that extinction induces the insertion of calcium-permeable (GluA2-lacking) AMPARs into IL synapses. Consistent with this, selectively blocking calcium-permeable AMPARs with Naspm reduced the AMPA EPSCs in IL neurons to a larger degree after extinction. Extinction-induced changes in AMPA/NMDA ratio, rectification, and intrinsic excitability were blocked with an mGluR5 antagonist. These findings suggest that mGluR5 activation leads to consolidation of fear extinction by regulating the intrinsic excitability of IL neurons and modifying the composition of AMPARs in IL synapses. Therefore, impaired mGluR5 activity in IL synapses could be one factor that causes inappropriate modulation of fear expression leading to anxiety disorders.


Assuntos
Extinção Psicológica/fisiologia , Sistema Límbico/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Sinapses/fisiologia , Animais , Antagonistas de Aminoácidos Excitatórios/farmacologia , Extinção Psicológica/efeitos dos fármacos , Medo , Sistema Límbico/efeitos dos fármacos , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Sinapses/efeitos dos fármacos
11.
J Neurovirol ; 20(4): 315-31, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24867611

RESUMO

HIV-1 infection can lead to neurocognitive impairment collectively known as HIV-associated neurocognitive disorders (HAND). Although combined antiretroviral treatment (cART) has significantly ameliorated HIV's morbidity and mortality, persistent neuroinflammation and neurocognitive dysfunction continue. This review focuses on the current clinical and molecular evidence of the viral and host factors that influence glutamate-mediated neurotoxicity and neuropathogenesis as an important underlying mechanism during the course of HAND development. In addition, discusses potential pharmacological strategies targeting the glutamatergic system that may help prevent and improve neurological outcomes in HIV-1-infected subjects.


Assuntos
Complexo AIDS Demência/metabolismo , Ácido Glutâmico/metabolismo , Animais , HIV-1 , Humanos
12.
Neurobiol Dis ; 49: 128-36, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22926191

RESUMO

Despite the widespread use of antiretroviral therapy that effectively limits viral replication, memory impairment remains a dilemma for HIV infected people. In the CNS, HIV infection of astrocytes leads to the production of the HIV-1 Nef protein without viral replication. Post mortem studies have found Nef expression in hippocampal astrocytes of people with HIV associated dementia suggesting that astrocytic Nef may contribute to HIV associated cognitive impairment even when viral replication is suppressed. To test whether astrocytic expression of Nef is sufficient to induce cognitive deficits, we examined the effect of implanting primary rat astrocytes expressing Nef into the hippocampus on spatial and recognition memory. Rats implanted unilaterally with astrocytes expressing Nef showed impaired novel location and novel object recognition in comparison with controls implanted with astrocytes expressing green fluorescent protein (GFP). This impairment was correlated with an increase in chemokine ligand 2 (CCL2) expression and the infiltration of peripheral macrophages into the hippocampus at the site of injection. Furthermore, the Nef exposed rats exhibited a bilateral loss of CA3 neurons. These results suggest that Nef protein expressed by the implanted astrocytes activates the immune system leading to neuronal damage and spatial and recognition memory deficits. Therefore, the continued expression of Nef by astrocytes in the absence of viral replication has the potential to contribute to HIV associated cognitive impairment.


Assuntos
Astrócitos/metabolismo , Hipocampo/fisiopatologia , Transtornos da Memória/fisiopatologia , Reconhecimento Psicológico/fisiologia , Memória Espacial/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Animais , Ansiedade/patologia , Ansiedade/fisiopatologia , Transplante de Tecido Encefálico , Células Cultivadas , Quimiocina CCL2/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/patologia , Imuno-Histoquímica , Deficiências da Aprendizagem/patologia , Deficiências da Aprendizagem/fisiopatologia , Macrófagos/fisiologia , Masculino , Transtornos da Memória/patologia , Atividade Motora/fisiologia , Neurônios/patologia , Neurônios/fisiologia , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Transfecção , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
13.
Behav Brain Res ; 454: 114639, 2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37652238

RESUMO

Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that not only entails alterations in fear behavior and anxiety but also includes neuroendocrine dysfunctions involving the hypothalamic pituitary adrenal (HPA) axis and the renin-angiotensin system. Recent preclinical studies demonstrate that activation of the angiotensin type 1 receptor (AT1R) in the paraventricular region of the hypothalamus (PVR) promotes anxiety-like behaviors and enables microglia proliferation. An increase in microglia and anxiety-like behavior also occurs in the PTSD animal model single-prolonged stress (SPS). In the present study, we tested whether AT1Rs contribute to the effects of SPS on behavior and microglia in brain structures important for HPA axis regulation and fear behavior. To test this, male and female animals were exposed to SPS and then given the oral AT1R antagonist candesartan beginning one week later. Candesartan did not alter auditory fear conditioning or extinction in SPS-exposed male or female animals. However, we found that the male animals exposed to SPS showed increased anxiety-like behavior, which was reversed by candesartan. In contrast, neither SPS nor candesartan altered anxiety-like behavior in the female animals. At the molecular level, SPS increased the cellular expression of AT1Rs in the PVR of male animals and candesartan reversed this effect, whereas AT1Rs in the PVR of females were unaltered by either SPS or candesartan. Iba1-expressing microglia increased in the PVR after SPS exposure and was reversed by candesartan in both sexes suggesting that SPS stimulates AT1Rs to increase microglia in the PVR. Collectively, these results suggest that the contribution of AT1Rs to the molecular and behavioral effects of SPS is sex-dependent.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II , Sistema Hipotálamo-Hipofisário , Feminino , Masculino , Animais , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Sistema Hipófise-Suprarrenal , Benzimidazóis/farmacologia
14.
Cereb Cortex ; 21(3): 727-35, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20705895

RESUMO

Consolidation of fear extinction involves enhancement of N-methyl D aspartate (NMDA) receptor-dependent bursting in the infralimbic region (IL) of the medial prefrontal cortex (mPFC). Previous studies have shown that systemic blockade of metabotropic glutamate receptor type 5 (mGluR5) reduces bursting in the mPFC and mGluR5 agonists enhance NMDA receptor currents in vitro, suggesting that mGluR5 activation in IL may contribute to fear extinction. In the current study, rats injected with the mGluR5 antagonist 2-methyl-6-(phenylethyl)-pyridine (MPEP) systemically, or intra-IL, prior to extinction exhibited normal within-session extinction, but were impaired in their ability to recall extinction the following day. To directly determine whether mGluR5 stimulation enhances the burst firing of IL neurons, we used patch-clamp electrophysiology in prefrontal slices. The mGluR5 agonist, (RS)-2-chloro-5-hydroxyphenylglycine (CHPG), increased intrinsic bursting in IL neurons. Increased bursting was correlated with a reduction in the slow after hyperpolarizing potential and was prevented by coapplication of MPEP. CHPG did not increase NMDA currents, suggesting that an NMDA receptor-independent enhancement of IL bursting via stimulation of mGluR5 receptors contributes to fear extinction. Therefore, the mGluR5 receptor could be a suitable target for pharmacological adjuncts to extinction-based therapies for anxiety disorders.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Sistema Límbico/fisiologia , Memória/fisiologia , Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Masculino , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5
15.
Front Behav Neurosci ; 16: 1014767, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699653

RESUMO

Single prolonged stress (SPS) is a preclinical rodent model for studying post-traumatic stress disorder (PTSD)-like behaviors. Previously we found that increased expression of the microglial marker Iba-1 in the ventral hippocampus after SPS exposure was associated with impaired fear extinction, suggesting that microglial activity contributed to the SPS-induced behavioral changes. To test this, we examined whether reducing microglia with the colony-stimulating factor 1 receptor blocker, PLX3397, in the diet would prevent the SPS-induced extinction impairment. Male rats exposed to SPS showed enhanced fear acquisition and impaired fear extinction memory. Adding PLX3397 to the diet prevented these behavioral changes. In contrast, PLX3397 did not prevent SPS from impairing fear extinction memory in the female rats. Despite the sex-dependent behavioral effects, we found a reduced number and area fraction of Iba-1+ microglia in both male and female rats suggesting that PLX3397 had similar effects on microglia in both sexes. Altogether, these results suggest that microglia contribute to the behavioral changes induced by SPS in male but not female rats.

16.
Front Mol Biosci ; 9: 859760, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35601829

RESUMO

Clinical and preclinical studies suggest that increases in long-chain ceramides in blood may contribute to the development of depressive-like behavior. However, which factors contribute to these increases and whether the increases are sufficient to induce depressive-like behaviors is unclear. To begin to address this issue, we examined the effects of high fat diet (HFD) and short-term unpredictable (STU) stress on long-chain ceramides in the serum of male and female rats. We found that brief exposure to HFD or unpredictable stress was sufficient to induce selective increases in the serum concentrations of long-chain ceramides, associated with depression in people. Furthermore, combined exposure to HFD and unpredictable stress caused a synergistic increase in C16:0, C16:1, and C18:0 ceramides in both sexes and C18:1 and C24:1 in males. However, the increased peripheral long-chain ceramides were not associated with increases in depressive-like behaviors suggesting that increases in serum long-chain ceramides may not be associated with the development of depressive-like behaviors in rodents.

17.
Front Behav Neurosci ; 16: 899627, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36090653

RESUMO

Increased long-chain C20:0 ceramides have been found in the serum of patients with depression. Moreover, ceramides are linked with increased microglia reactivity and inflammatory cytokine production, which are associated with depression. Since ceramides can readily cross the blood brain barrier, peripheral C20:0 ceramides could enter the brain, activate microglia, and induce depressive-like behavior. In this study, we determined whether localized infusion of C20:0 ceramides into the ventral hippocampus (VH) of rats is sufficient to activate microglia and induce depressive-like behaviors. Adult male and female rats received infusions of C20:0 ceramides or vehicle solution every other day for 2 weeks. After the third infusion, C20:0-infused animals showed reduced sucrose preference suggesting anhedonia-like behavior. In contrast, infusions of C20:0 ceramides did not affect immobility in the forced swim test or sucrose grooming suggesting that the behavioral effects of ceramides are task dependent. Furthermore, C20:0-infusions did not increase Iba-1 + microglia or inflammatory markers in the VH suggesting that localized increases in C20:0 ceramides in the VH are sufficient to induce anhedonia-like behavior without microglia activation.

18.
J Neurosci ; 30(37): 12379-86, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844133

RESUMO

Growing evidence indicates that the activity of infralimbic prefrontal cortex (IL) is critical for inhibiting inappropriate fear responses following extinction learning. Recently, we showed that fear conditioning and extinction alter the intrinsic excitability and bursting of IL pyramidal neurons in brain slices. IL neurons from Sprague Dawley rats expressing high fear had lower intrinsic excitability and bursting than those from rats expressing low fear, suggesting that regulating the intrinsic excitability and bursting of IL neurons would modulate fear expression. To test this, we combined patch-clamp electrophysiology, auditory fear conditioning, and IL infusions of M-type K(+) channel modulators. Patch-clamp recordings from IL neurons showed that the M-type K(+) channel blocker, XE-991, increased the number of spikes evoked by a depolarizing pulse and reduced the first interspike interval indicating enhanced bursting. To test whether pharmacological enhancement of IL excitability and bursting reduces fear expression and facilitates extinction, fear-conditioned rats were infused with XE-991 into IL before extinction training. XE-infused rats showed reduced freezing and facilitated extinction compared to vehicle-infused rats. The following day, recall of extinction memory was enhanced. Reducing IL excitability and bursting with the M-type K(+) channel agonist, flupirtine, had the opposite effect. Flupirtine reduced IL spike count and bursting in brain slices. Fear-conditioned rats infused with flupirtine into IL before extinction showed significantly higher levels of freezing, indicating that stimulation of M-channels enhanced fear expression. Our findings suggest that the intrinsic excitability and bursting of IL neurons regulate fear expression even before extinction.


Assuntos
Potenciais de Ação/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Canais de Potássio KCNQ/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Estimulação Acústica , Potenciais de Ação/efeitos dos fármacos , Aminopiridinas/farmacologia , Analgésicos/farmacologia , Animais , Antracenos/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Canais de Potássio KCNQ/agonistas , Canais de Potássio KCNQ/antagonistas & inibidores , Masculino , Memória/efeitos dos fármacos , Memória/fisiologia , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Córtex Pré-Frontal/citologia , Ratos , Ratos Sprague-Dawley
19.
Front Synaptic Neurosci ; 13: 695964, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335223

RESUMO

Although the infralimbic cortex (IL) is not thought to play a role in fear acquisition, recent experiments found evidence that synaptic plasticity is occurring at ventral hippocampal (vHPC) synapses in IL during auditory fear acquisition as measured by changes in the N-methyl-D-aspartate (NMDA) receptor-mediated currents in male rats. These electrophysiological data suggest that fear conditioning changes the expression of NMDA receptors on vHPC-to-IL synapses. To further evaluate the plasticity of NMDA receptors at this specific synapse, we injected AAV particles expressing channelrhodopsin-EYFP into the vHPC of male and female rats to label vHPC projections with EYFP. To test for NMDA receptor changes in vHPC-to-IL synapses after fear learning, we used fluorescence-activated cell sorting (FACS) to quantify synaptosomes isolated from IL tissue punches that were positive for EYFP and the obligatory GluN1 subunit. More EYFP+/GluN1+ synaptosomes with greater average expression of GluN1 were isolated from male rats exposed to auditory fear conditioning (AFC) than those exposed to context and tones only or to contextual fear conditioning (CFC), suggesting that AFC increased NMDA receptor expression in males. In a second experiment, we found that pairing the tones and shocks was required to induce the molecular changes and that fear extinction did not reverse the changes. In contrast, females showed similar levels of EYFP+/GluN1+ synaptosomes in all behavioral groups. These findings suggest that AFC induces synaptic plasticity of NMDA receptors in the vHPC-to-IL projection in males, while female rats rely on different synaptic mechanisms.

20.
Artigo em Inglês | MEDLINE | ID: mdl-33562262

RESUMO

The Research Centers in Minority Institutions (RCMI) Program was congressionally mandated in 1985 to build research capacity at institutions that currently and historically recruit, train, and award doctorate degrees in the health professions and health-related sciences, primarily to individuals from underrepresented and minority populations. RCMI grantees share similar infrastructure needs and institutional goals. Of particular importance is the professional development of multidisciplinary teams of academic and community scholars (the "workforce") and the harnessing of the heterogeneity of thought (the "thinkforce") to reduce health disparities. The purpose of this report is to summarize the presentations and discussion at the RCMI Investigator Development Core (IDC) Workshop, held in conjunction with the RCMI Program National Conference in Bethesda, Maryland, in December 2019. The RCMI IDC Directors provided information about their professional development activities and Pilot Projects Programs and discussed barriers identified by new and early-stage investigators that limit effective career development, as well as potential solutions to overcome such obstacles. This report also proposes potential alignments of professional development activities, targeted goals and common metrics to track productivity and success.


Assuntos
Pesquisa Biomédica , Grupos Minoritários , Humanos , Maryland , Pesquisadores , Recursos Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa