Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroimage ; 289: 120542, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38369167

RESUMO

MRI-guided neuro interventions require rapid, accurate, and reproducible segmentation of anatomical brain structures for identification of targets during surgical procedures and post-surgical evaluation of intervention efficiency. Segmentation algorithms must be validated and cleared for clinical use. This work introduces a methodology for shape-constrained deformable brain segmentation, describes the quantitative validation used for its clinical clearance, and presents a comparison with manual expert segmentation and FreeSurfer, an open source software for neuroimaging data analysis. ClearPoint Maestro is software for fully-automatic brain segmentation from T1-weighted MRI that combines a shape-constrained deformable brain model with voxel-wise tissue segmentation within the cerebral hemispheres and the cerebellum. The performance of the segmentation was validated in terms of accuracy and reproducibility. Segmentation accuracy was evaluated with respect to training data and independently traced ground truth. Segmentation reproducibility was quantified and compared with manual expert segmentation and FreeSurfer. Quantitative reproducibility analysis indicates superior performance compared to both manual expert segmentation and FreeSurfer. The shape-constrained methodology results in accurate and highly reproducible segmentation. Inherent point based-correspondence provides consistent target identification ideal for MRI-guided neuro interventions.


Assuntos
Algoritmos , Software , Humanos , Reprodutibilidade dos Testes , Encéfalo/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Processamento de Imagem Assistida por Computador/métodos
2.
Neuropharmacology ; 148: 40-49, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30557566

RESUMO

Brain-derived neurotrophic factor (BDNF) is known to promote fear learning as well as avoidant behavioral responses to chronic social defeat stress, but, conversely, this peptide can also have antidepressant effects and can reduce depressant-like symptoms such as social avoidance. The purpose of this study was to use a variety of approaches to determine whether BDNF acting on tropomyosin receptor kinase B (TrkB) promotes or prevents avoidant phenotypes in hamsters and mice that have experienced acute social defeat stress. We utilized systemic and brain region-dependent manipulation of BDNF signaling before or immediately following social defeat stress in Syrian hamsters, TrkBF616A knock-in mice, and C57Bl/6J mice and measured the subsequent behavioral response to a novel opponent. Systemic TrkB receptor agonists reduced, and TrkB receptor antagonists enhanced, behavioral responses to social defeat in hamsters and mice. In the neural circuit that we have shown mediates defeat-induced behavioral responses, BDNF in the basolateral amygdala, but not the nucleus accumbens, also reduced social avoidant phenotypes. Conversely, knockdown in the basolateral amygdala of TrkB signaling in TrkBF616A mice enhanced defeat-induced social avoidance. These data demonstrate that systemic administration of BDNF-TrkB drugs at the time of social defeat alters the behavioral response to the defeat stressor. These drugs appear to act, at least in part, in the basolateral amygdala and not the nucleus accumbens. These findings were generalizable to two rodent species with very different social structures and, within mice, to a variety of strains providing converging evidence that BDNF-TrkB signaling reduces anxiety- and depression-like symptoms following short-term social stress.


Assuntos
Aprendizagem da Esquiva/fisiologia , Complexo Nuclear Basolateral da Amígdala/fisiologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Glicoproteínas de Membrana/fisiologia , Núcleo Accumbens/fisiologia , Proteínas Tirosina Quinases/fisiologia , Comportamento Social , Estresse Psicológico/psicologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Cricetinae , Técnicas de Introdução de Genes , Técnicas de Silenciamento de Genes , Masculino , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Camundongos , Microinjeções , Núcleo Accumbens/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética
3.
Biol Psychiatry ; 83(3): 284-295, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29054677

RESUMO

BACKGROUND: Molecular mechanisms underlying psychological sequelae of exposure to stressful experiences, such as posttraumatic stress disorder (PTSD) and depression, are not well understood. METHODS: Using convergent evidence from animal and human transcriptomic and genomic studies, we aimed to identify genetic mechanisms underlying depression and anxiety after traumatic experiences. RESULTS: From a transcriptome-wide analysis in mice, we found the Ppm1f gene to be differentially expressed in the amygdala and medial prefrontal cortex (mPFC) a week after immobilization stress. Next, we found that PPM1F messenger RNA levels in human blood were downregulated in cases with symptoms of comorbid PTSD and depression and consistently in cases with anxiety symptoms in a separate human dataset. Furthermore, we showed that a genetic variant of PPM1F, rs17759843, was associated with comorbid PTSD and depression and with PPM1F expression in both human brain and blood. Given prior reported mechanistic links between PPM1F and CAMK2 (CAMKII), we examined blood messenger RNA level of CAMK2G in humans and found it to be lower in cases with comorbid PTSD and depression. We also found that PPM1F protein levels and colocalization with CAMK2G were altered in amygdala and mPFC of male mice. Additionally, we found that a systemic dose of corticosterone blocked the depressive-like phenotype elicited by stress in female mice. Lastly, corticosterone rescued the anxiety-like phenotype and messenger RNA levels of Ppm1f in amygdala and mPFC in male mice and in mPFC of female mice. CONCLUSIONS: Taken together, our data suggest a mechanistic pathway involving PPM1F and CAMK2G in stress- and trauma-related manifestation of anxiety and depression across species.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Transtorno Depressivo/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Córtex Pré-Frontal/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Estresse Psicológico/metabolismo , Animais , Ansiedade/epidemiologia , Comportamento Animal/fisiologia , Comorbidade , Transtorno Depressivo/epidemiologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos de Estresse Pós-Traumáticos/epidemiologia , Estresse Psicológico/epidemiologia , Transcriptoma/genética
4.
Physiol Behav ; 177: 264-269, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28511867

RESUMO

Although Syrian hamsters are thought to be naturally solitary, recent evidence from our laboratory demonstrates that hamsters may actually prefer social contact. Hamsters increase their preference for a location associated with an agonistic encounter regardless of whether they have "won" or "lost". It has also been reported that social housing as well as exposure to intermittent social defeat or to a brief footshock stressor increase food intake and body mass in hamsters. By contrast, it has also been suggested that housing hamsters in social isolation causes anxiety-induced anorexia and reductions in body mass selectively in females. The purpose of this study was to determine the physiological consequences of housing hamsters in social isolation versus in social groups. Male and female hamsters were housed singly or in stable groups of 5 for 4weeks after which they were weighed and trunk blood was collected. In addition, fat pads and thymus and adrenal glands were extracted and weighed. Serum and fecal cortisol were measured using an enzyme-linked immunoassay. Housing condition had no effect on serum or fecal cortisol, but socially housed hamsters displayed modest thymus gland involution. Socially housed females weighed more than did any other group, and socially housed females and males had more fat than did socially isolated hamsters. No wounding or tissue damage occurred in grouped hamsters. Overall, these data suggest that Syrian hamsters tolerate both stable social housing and social isolation in the laboratory although social housing is associated with some alteration in stress-related and bioenergetic measures.


Assuntos
Abrigo para Animais , Hidrocortisona/metabolismo , Isolamento Social/psicologia , Estresse Psicológico/metabolismo , Adaptação Psicológica , Adiposidade , Glândulas Suprarrenais/anatomia & histologia , Animais , Peso Corporal , Dominação-Subordinação , Feminino , Masculino , Mesocricetus , Tamanho do Órgão , Caracteres Sexuais , Timo/anatomia & histologia
5.
Psychoneuroendocrinology ; 75: 100-109, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27810703

RESUMO

Histone acetylation has emerged as a critical factor regulating learning and memory both during and after exposure to stressful stimuli. There are drugs that we now know affect histone acetylation that are already in use in clinical populations. The current study uses these drugs to examine the consequences of acutely increasing or decreasing histone acetylation during exposure to social stress. Using an acute model of social defeat in Syrian hamsters, we systemically and site-specifically administered drugs that alter histone acetylation and measured subsequent behavior and immediate-early gene activity. We found that systemic administration of a histone deacetylase inhibitor enhances social stress-induced behavioral responses in males and females. We also found that systemic administration completely blocks defeat-induced neuronal activation, as measured by Fos-immunoreactivity, in the infralimbic cortex, but not in the amygdala, after a mild social defeat stressor. Lastly, we demonstrated that site-specific administration of histone deacetylase inhibitors in the infralimbic region of the prefrontal cortex, but not in the basolateral amygdala, mimics the systemic effect. Conversely, decreasing acetylation by inhibiting histone acetyltransferases in the infralimbic cortex reduces behavioral responses to defeat. This is the first demonstration that acute pharmacological manipulation of histone acetylation during social defeat alters subsequent behavioral responses in both males and females. These results reveal that even systemic administration of drugs that alter histone acetylation can significantly alter behavioral responses to social stress and highlight the importance of the infralimbic cortex in mediating this effect.


Assuntos
Acetiltransferases/antagonistas & inibidores , Comportamento Animal/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Comportamento Social , Estresse Psicológico/fisiopatologia , Animais , Cricetinae , Feminino , Masculino , Mesocricetus
6.
Prog Mol Biol Transl Sci ; 122: 169-92, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24484701

RESUMO

Brain-derived neurotrophic factor (BDNF) and its single transmembrane receptor, tropomysin-related kinase B (TrkB), are essential for adult synaptic plasticity and the formation of memories. However, there are regional and task-dependent differences underlying differential mechanisms of BDNF-TrkB function in the formation of these memories. Additionally, the BDNF pathway has been implicated in several psychiatric disorders including posttraumatic stress disorder, phobia, and panic disorder. Gaining a better understanding of this pathway and the neurobiology of memory through fundamental research may be helpful to identify effective prevention and treatment approaches both for diseases of memory deficit as well as in cases of enhanced aversive memory, such as in anxiety disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Regulação da Expressão Gênica , Memória/fisiologia , Transtornos Mentais/metabolismo , Transtornos Mentais/patologia , Plasticidade Neuronal/fisiologia , Receptor trkB/metabolismo , Adulto , Fator Neurotrófico Derivado do Encéfalo/genética , Humanos , Transtornos Mentais/genética , Receptor trkB/genética , Transdução de Sinais
7.
Biol Psychiatry ; 75(11): 864-72, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24094510

RESUMO

BACKGROUND: The current effective treatment options for posttraumatic stress disorder (PTSD) are limited, and therefore the need to explore new treatment strategies is critical. Pharmacological inhibition of the renin-angiotensin system is a common approach to treat hypertension, and emerging evidence highlights the importance of this pathway in stress and anxiety. A recent clinical study from our laboratory provides evidence supporting a role for the renin-angiotensin system in the regulation of the stress response in patients diagnosed with PTSD. METHODS: With an animal model of PTSD and the selective angiotensin receptor type 1 (AT1) antagonist losartan, we investigated the acute and long-term effects of AT1 receptor inhibition on fear memory and baseline anxiety. After losartan treatment, we performed classical Pavlovian fear conditioning pairing auditory cues with footshocks and examined extinction behavior, gene expression changes in the brain, as well as neuroendocrine and cardiovascular responses. RESULTS: After cued fear conditioning, both acute and 2-week administration of losartan enhanced the consolidation of extinction memory but had no effect on fear acquisition, baseline anxiety, blood pressure, and neuroendocrine stress measures. Gene expression changes in the brain were also altered in mice treated with losartan for 2 weeks, in particular reduced amygdala AT1 receptor and bed nucleus of the stria terminalis c-Fos messenger RNA levels. CONCLUSIONS: These data suggest that AT1 receptor antagonism enhances the extinction of fear memory and therefore might be a beneficial therapy for PTSD patients who have impairments in extinction of aversive memories.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Losartan/farmacologia , Memória/efeitos dos fármacos , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Animais , Modelos Animais de Doenças , Losartan/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico
8.
J Neurosci ; 33(25): 10396-404, 2013 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23785152

RESUMO

Research has identified distinct neuronal circuits within the basolateral amygdala (BLA) that differentially mediate fear expression versus inhibition; however, molecular markers of these populations remain unknown. Here we examine whether optogenetic activation of a cellular subpopulation, which may correlate with the physiologically identified extinction neurons in the BLA, would differentially support fear conditioning versus fear inhibition/extinction. We first molecularly characterized Thy1-channelrhodopsin-2 (Thy1-ChR2-EYFP)-expressing neurons as a subpopulation of glutamatergic pyramidal neurons within the BLA. Optogenetic stimulation of these neurons inhibited a subpopulation of medial central amygdala neurons and shunted excitation from the lateral amygdala. Brief activation of these neurons during fear training disrupted later fear memory in male mice. Optogenetic activation during unreinforced stimulus exposure enhanced extinction retention, but had no effect on fear expression, locomotion, or open-field behavior. Together, these data suggest that the Thy1-expressing subpopulation of BLA pyramidal neurons provide an important molecular and pharmacological target for inhibiting fear and enhancing extinction and for furthering our understanding of the molecular mechanisms of fear processing.


Assuntos
Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Inibição Psicológica , Neurônios/fisiologia , Antígenos Thy-1/fisiologia , Tonsila do Cerebelo/citologia , Animais , Channelrhodopsins , Condicionamento Psicológico , Sinais (Psicologia) , Extinção Psicológica , Ácido Glutâmico/fisiologia , Imuno-Histoquímica , Lasers , Locomoção/fisiologia , Camundongos , Atividade Motora/fisiologia , Técnicas de Patch-Clamp , Estimulação Luminosa , Antígenos Thy-1/genética
9.
Neuroimage ; 63(2): 800-11, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22796992

RESUMO

BACKGROUND: High throughput, brain-wide analysis of neural circuit connectivity is needed to understand brain function across species. Combining such tractography techniques with small animal models will allow more rapid integration of systems neuroscience with molecular genetic, behavioral, and cellular approaches. METHODS: We collected DTI and T2 scans on 3 series of 6 fixed mouse brains ex vivo in a 9.4 Tesla magnet. The DTI analysis of ten mouse brains focused on comparing prelimbic (PL) and Infralimbic (IL) probabilistic tractography. To validate the DTI results a preliminary set of 24 additional mice were injected with BDA into the IL and PL. The DTI results and preliminary BDA results were also compared to previously published rat connectivity. RESULTS: We focused our analyses on the connectivity of the mouse prelimbic (PL) vs. infralimbic (IL) cortices. We demonstrated that this DTI analysis is consistent across scanned mice, with prior analyses of rat IL/PL connectivity, and with mouse PL and IL projections using the BDA tracer. CONCLUSIONS: High-throughput ex vivo DTI imaging in the mouse delineated both common and differential connectivity of the IL and PL cortex. The scanning methodology provided a balance of tissue contrast, signal-to-noise ratio, resolution and throughput. Our results are largely consistent with previously published anterograde staining techniques in rats, and the preliminary tracer study of the mouse IL and PL provided here.


Assuntos
Encéfalo/anatomia & histologia , Imagem de Tensor de Difusão/métodos , Vias Neurais/anatomia & histologia , Animais , Ensaios de Triagem em Larga Escala , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Endogâmicos C57BL
10.
Physiol Behav ; 107(5): 699-710, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-22429904

RESUMO

The neural circuitry of fear likely underlies anxiety and fear-related disorders such as specific and social phobia, panic disorder, and posttraumatic stress disorder. The primary pharmacological treatments currently utilized for these disorders include benzodiazepines, which act on the GABAergic receptor system, and antidepressants, which modulate the monamine systems. However, recent work on the regulation of fear neural circuitry suggests that specific neuropeptide modulation of this system is of critical importance. Recent reviews have examined the roles of the hypothalamic-pituitary-adrenal axis neuropeptides as well as the roles of neurotrophic factors in regulating fear. The present review, instead, will focus on three neuropeptide systems which have received less attention in recent years but which are clearly involved in regulating fear and its extinction. The endogenous opioid system, particularly activating the µ opioid receptors, has been demonstrated to regulate fear expression and extinction, possibly through functioning as an error signal within the ventrolateral periaqueductal gray to mark unreinforced conditioned stimuli. The cholecystokinin (CCK) system initially led to much excitement through its potential role in panic disorder. More recent work in the CCK neuropeptide pathway suggests that it may act in concordance with the endogenous cannabinoid system in the modulation of fear inhibition and extinction. Finally, older as well as very recent data suggests that neuropeptide Y (NPY) may play a very interesting role in counteracting stress effects, enhancing extinction, and enhancing resilience in fear and stress preclinical models. Future work in understanding the mechanisms of neuropeptide functioning, particularly within well-known behavioral circuits, are likely to provide fascinating new clues into the understanding of fear behavior as well as suggesting novel therapeutics for treating disorders of anxiety and fear dysregulation.


Assuntos
Ansiedade/fisiopatologia , Colecistocinina/fisiologia , Medo/fisiologia , Neuropeptídeo Y/fisiologia , Peptídeos Opioides/fisiologia , Animais , Encéfalo/fisiologia , Humanos
11.
Proc Natl Acad Sci U S A ; 107(47): 20529-34, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21059919

RESUMO

Individuals often eat calorically dense, highly palatable "comfort" foods during stress for stress relief. This article demonstrates that palatable food intake (limited intake of sucrose drink) reduces neuroendocrine, cardiovascular, and behavioral responses to stress in rats. Artificially sweetened (saccharin) drink reproduces the stress dampening, whereas oral intragastric gavage of sucrose is without effect. Together, these results suggest that the palatable/rewarding properties of sucrose are necessary and sufficient for stress dampening. In support of this finding, another type of natural reward (sexual activity) similarly reduces stress responses. Ibotenate lesions of the basolateral amygdala (BLA) prevent stress dampening by sucrose, suggesting that neural activity in the BLA is necessary for the effect. Moreover, sucrose intake increases mRNA and protein expression in the BLA for numerous genes linked with functional and/or structural plasticity. Lastly, stress dampening by sucrose is persistent, which is consistent with long-term changes in neural activity after synaptic remodeling. Thus, natural rewards, such as palatable foods, provide a general means of stress reduction, likely via structural and/or functional plasticity in the BLA. These findings provide a clearer understanding of the motivation for consuming palatable foods during times of stress and influence therapeutic strategies for the prevention and/or treatment of obesity and other stress-related disorders.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Comportamento Animal/fisiologia , Prazer/fisiologia , Estresse Psicológico/fisiopatologia , Sacarose/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Análise de Variância , Animais , Fenômenos Fisiológicos Cardiovasculares , Hormônios/sangue , Masculino , Análise em Microsséries , Ratos , Estresse Psicológico/tratamento farmacológico , Telemetria
12.
Proc Natl Acad Sci U S A ; 107(6): 2675-80, 2010 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-20133801

RESUMO

In the medial prefrontal cortex, the prelimbic area is emerging as a major modulator of fear behavior, but the mechanisms remain unclear. Using a selective neocortical knockout mouse, virally mediated prelimbic cortical-specific gene deletion, and pharmacological rescue with a TrkB agonist, we examined the role of a primary candidate mechanism, BDNF, in conditioned fear. We found consistently robust deficits in consolidation of cued fear but no effects on acquisition, expression of unlearned fear, sensorimotor function, and spatial learning. This deficit in learned fear in the BDNF knockout mice was rescued with systemic administration of a TrkB receptor agonist, 7,8-dihydroxyflavone. These data indicate that prelimbic BDNF is critical for consolidation of learned fear memories, but it is not required for innate fear or extinction of fear. Moreover, use of site-specific, inducible BDNF deletions shows a powerful mechanism that may further our understanding of the pathophysiology of fear-related disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Medo/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Córtex Cerebral/metabolismo , Condicionamento Psicológico/efeitos dos fármacos , Extinção Psicológica , Medo/efeitos dos fármacos , Medo/psicologia , Feminino , Flavonas/farmacologia , Hibridização In Situ , Aprendizagem/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Knockout , Neocórtex/metabolismo , Receptor trkB/agonistas , Receptor trkB/fisiologia
13.
Curr Top Behav Neurosci ; 2: 279-99, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21309114

RESUMO

Improved efficacy in the treatment of posttraumatic stress disorder (PTSD) and other anxiety disorders is urgently needed. Traditional anxiety treatments of hypnosis and psychodynamic therapy may be of some help, but uncontrolled studies lead to inconclusive results on the efficacy of these treatment techniques. There is a larger literature supporting the efficacy of cognitive-behavioral procedures with PTSD, including prolonged exposure therapy, eye movement desensitization and reprocessing, and anxiety management techniques. The cutting-edge technology of virtual reality-based exposure therapy for PTSD is particularly exciting. To further build on effective psychosocial treatments, current pharmacological augmentation approaches to emotional learning are being combined with psychotherapy. In particular, D-cycloserine, a partial NMDA agonist, has shown to be effective in facilitating the exposure/extinction therapy to improve the efficacy of treating anxiety disorders, and may guide the way for new pharmacological enhancements of behavioral therapy.


Assuntos
Terapia Cognitivo-Comportamental , Ciclosserina/uso terapêutico , Dessensibilização Psicológica , Extinção Psicológica , Hipnose , Transtornos de Estresse Pós-Traumáticos/terapia , Animais , Transtornos de Ansiedade/terapia , Terapia Cognitivo-Comportamental/métodos , Terapia Combinada/métodos , Dessensibilização Psicológica/métodos , Dessensibilização e Reprocessamento através dos Movimentos Oculares , Humanos , Terapia Implosiva , Transtorno Obsessivo-Compulsivo/terapia , Transtorno de Pânico/terapia , Transtornos Fóbicos/terapia , Receptores de N-Metil-D-Aspartato/agonistas , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Transtornos de Estresse Pós-Traumáticos/psicologia , Resultado do Tratamento
14.
J Neurosci ; 28(49): 13106-11, 2008 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-19052201

RESUMO

Olfactory learning in humans leads to enhanced perceptual discrimination of odor cues. Examining mouse models of both aversive and appetitive conditioning, we demonstrate a mechanism which may underlie this adult learning phenomenon. Topographically unique spatial wiring of the olfactory system allowed us to demonstrate that emotional learning of odor cues alters the primary sensory representation within the nose and brain of adult mice. Transgenic mice labeled at the M71 odorant receptor (specifically activated by the odorant acetophenone) were behaviorally trained with olfactory-dependent fear conditioning or conditioned place preference using acetophenone. Odor-trained mice had larger M71-specific glomeruli and an increase in M71-specific sensory neurons within the nose compared with mice that were untrained, trained to a non-M71 activating odorant, or had nonassociative pairings of acetophenone. These data indicate that the primary sensory neuron population and its projections may remain plastic in adults, providing a structural mechanism for learning-enhanced olfactory sensitivity and discrimination.


Assuntos
Aprendizagem/fisiologia , Plasticidade Neuronal/fisiologia , Condutos Olfatórios/fisiologia , Neurônios Receptores Olfatórios/fisiologia , Olfato/fisiologia , Acetofenonas/farmacologia , Animais , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Sinais (Psicologia) , Emoções/fisiologia , Medo/fisiologia , Masculino , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Condutos Olfatórios/efeitos dos fármacos , Neurônios Receptores Olfatórios/efeitos dos fármacos , Receptores Odorantes/efeitos dos fármacos , Receptores Odorantes/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia
15.
Psychoneuroendocrinology ; 33(5): 659-69, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18378095

RESUMO

The bed nucleus of the stria terminalis (BST) plays a prominent role in brain integration of acute responses to stressful stimuli. This study tests the hypothesis that the BST plays a complementary role in regulation of physiological changes associated with chronic stress exposure. Male Sprague-Dawley rats received bilateral ibotenate lesions or sham lesions of the posterior medial region of the BST (BSTpm), an area known to be involved in inhibition of HPA axis responses to acute stress. Chronic stress was induced by 14-day exposure to twice daily stressors in an unpredictable sequence (chronic variable stress, CVS). In the morning after the end of CVS, stressed and non-stressed controls were exposed to a novel restraint stress challenge. As previously documented, CVS caused adrenal hypertrophy, thymic involution, and attenuated body weight gain. None of these endpoints were affected by BSTpm lesions. Chronic stress exposure facilitated plasma corticosterone responses to the novel restraint stress and elevated CRH mRNA. Lesions of the BSTpm increased novel stressor-induced plasma ACTH and corticosterone secretion and enhanced c-fos mRNA induction in the paraventricular nucleus of the hypothalamus (PVN). In addition, lesion of the BSTpm resulted in an additive increase in CVS-induced facilitation of corticosterone responses and PVN CRH expression. Collectively these data confirm that the BSTpm markedly inhibits HPA responses to acute stress, but do not strongly support an additional role for this region in limiting HPA axis responses to chronic drive. The data further suggest that acute versus chronic stress integration are subserved by different brain circuitry.


Assuntos
Glândulas Suprarrenais/patologia , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Núcleos Septais/metabolismo , Estresse Psicológico/metabolismo , Doença Aguda , Hormônio Adrenocorticotrópico/sangue , Animais , Peso Corporal , Doença Crônica , Corticosterona/sangue , Agonistas de Aminoácidos Excitatórios , Hipertrofia/psicologia , Ácido Ibotênico , Imuno-Histoquímica , Hibridização In Situ , Masculino , Tamanho do Órgão , Proteínas Proto-Oncogênicas c-fos/metabolismo , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Núcleos Septais/patologia , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/patologia , Aumento de Peso
16.
Endocrinology ; 149(2): 818-26, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18039788

RESUMO

The anteroventral region of the bed nucleus of the stria terminalis (BST) stimulates hypothalamic-pituitary-adrenocortical (HPA) axis responses to acute stress. However, the role of the anterior BST nuclei in chronic drive of the HPA axis has yet to be established. Therefore, this study tests the role of the anteroventral BST in physiological responses to chronic drive, using a chronic variable stress (CVS) model. Male Sprague-Dawley rats received either bilateral ibotenate lesions, targeting the anteroventral BST, or vehicle injection into the same region. Half of the lesion and control rats were exposed to a 14-d CVS paradigm consisting of twice-daily exposure to unpredictable, alternating stressors. The remaining rats were nonhandled control animals that remained in home cages. On the morning after the end of CVS exposure, all rats were exposed to a novel restraint stress challenge. CVS induced attenuated body weight gain, adrenal hypertrophy, thymic involution, and enhanced CRH mRNA in hypophysiotrophic neurons of the hypothalamic paraventricular nucleus, none of which were affected by anteroventral BST lesions. In the absence of CVS, lesions attenuated the plasma corticosterone and paraventricular nucleus c-fos mRNA responses to the acute restraint stress. In contrast, lesions of the anteroventral BST elevated plasma ACTH and corticosterone responses to novel restraint in the rats previously exposed to CVS. These data suggest that the anterior BST plays very different roles in integrating acute stimulation and chronic drive of the HPA axis, perhaps mediated by chronic stress-induced recruitment of distinct BST cell groups or functional reorganization of stress-integrative circuits.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Núcleos Septais/fisiologia , Estresse Fisiológico/fisiopatologia , Doença Aguda , Hormônio Adrenocorticotrópico/sangue , Animais , Arginina Vasopressina/genética , Peso Corporal/fisiologia , Doença Crônica , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Sistema Hipotálamo-Hipofisário/citologia , Masculino , Vias Neurais , Tamanho do Órgão/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/citologia , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Núcleos Septais/citologia
17.
J Neurosci ; 27(8): 2025-34, 2007 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-17314298

RESUMO

Limbic and cortical neurocircuits profoundly influence hypothalamic-pituitary-adrenal (HPA) axis responses to stress yet have little or no direct projections to the hypothalamic paraventricular nucleus (PVN). Numerous lines of evidence suggest that the bed nucleus of the stria terminalis (BST) is well positioned to relay limbic information to the PVN. The BST comprises multiple anatomically distinct nuclei, of which some are known to receive direct limbic and/or cortical input and to heavily innervate the PVN. Our studies test the hypothesis that subregions of the BST differentially regulate HPA axis responses to acute stress. Male Sprague Dawley rats received bilateral ibotenate lesions, targeting either the principal nucleus in the posterior BST or the dorsomedial/fusiform nuclei in the anteroventral BST. Posterior BST lesions elevated plasma ACTH and corticosterone in response to acute restraint stress, increased stress-induced PVN c-fos mRNA, and elevated PVN corticotropin-releasing hormone (CRH) and parvocellular arginine vasopressin (AVP) mRNA expression relative to sham-lesion animals. In contrast, anterior BST lesions attenuated the plasma corticosterone response and decreased c-fos mRNA induction in the PVN but did not affect CRH and parvocellular AVP mRNA expression in the PVN. These data suggest that posterior BST nuclei are involved in inhibition of the HPA axis, whereas the anteroventral BST nuclei are involved in HPA axis excitation. The results indicate that the BST contains functional subdomains that play different roles in integrating and processing limbic information in response to stress and further suggest that excitatory as well as inhibitory limbic information is funneled through these important cell groups.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiopatologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Núcleos Septais/fisiopatologia , Estresse Fisiológico/fisiopatologia , Doença Aguda , Glândulas Suprarrenais/patologia , Hormônio Adrenocorticotrópico/sangue , Animais , Peso Corporal , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Glutamato Descarboxilase/genética , Isoenzimas/genética , Sistema Límbico/fisiologia , Masculino , Tamanho do Órgão , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Restrição Física , Núcleos Septais/metabolismo , Estresse Fisiológico/sangue , Estresse Fisiológico/metabolismo , Estresse Fisiológico/patologia , Timo/patologia , Vasopressinas/genética
18.
Brain Res ; 1138: 1-9, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17276416

RESUMO

Development of the hypothalamo-pituitary-adrenocortical (HPA) axis is marked by a diminution in stress responsiveness early in the postnatal period (days 4-14 in the rat). This 'stress hyporesponsive period' (SHRP) is thought to be at least in part centrally mediated. To investigate central mechanisms underlying the SHRP, this study assessed expression of glutamic acid decarboxylase (GAD) 67 in key stress-regulatory regions in the forebrain following acute stress with or without prior maternal deprivation. This isoform of GAD is known to be induced by stress in the adult and is believed to be a major contributor to production of the inhibitory neurotransmitter GABA under stimulated conditions. Expression of GAD67 mRNA was increased in the hippocampus, central amygdala and dorsomedial hypothalamus in pups tested early in the SHRP (day 6) or after its conclusion (day 18). In contrast, restraint caused a down-regulation of GAD67 mRNA in these structures when tested later in the SHRP (day 12). GAD67 mRNA expression was not affected by prior maternal deprivation in these regions. Reduced GABA production in the hippocampus (interneurons) is consistent with enhanced HPA axis inhibition, whereas reduced amygdalar expression predicts impaired stress excitation. Expression of GAD67 mRNA in the bed nucleus of the stria terminalis (BST) was minimally affected by acute restraint or maternal deprivation during the SHRP. However, older animals showed down-regulation of basal expression following maternal deprivation and substantial GAD67 mRNA up-regulation in both deprived and non-deprived groups following acute restraint. In contrast, non-responsiveness of the BST during the SHRP suggests either that BST GABA circuits are not actively engaged by stressors during this period or that circuits regulating BST GAD67 production are not yet in place. Overall, the data implicate forebrain GABA circuits in inhibition of HPA axis activity during the SHRP.


Assuntos
Glutamato Descarboxilase/genética , Hipotálamo/fisiopatologia , Isoenzimas/genética , Sistema Límbico/fisiopatologia , Prosencéfalo/fisiopatologia , RNA Mensageiro/metabolismo , Estresse Fisiológico/fisiopatologia , Ácido gama-Aminobutírico/metabolismo , Envelhecimento/metabolismo , Animais , Feminino , Hipotálamo/enzimologia , Sistema Límbico/enzimologia , Masculino , Privação Materna , Inibição Neural , Vias Neurais/fisiopatologia , Prosencéfalo/enzimologia , Prosencéfalo/metabolismo , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Restrição Física , Núcleos Septais/fisiopatologia , Estresse Fisiológico/etiologia , Estresse Fisiológico/metabolismo , Distribuição Tecidual , Regulação para Cima
19.
Endocrinology ; 148(4): 1823-34, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17204558

RESUMO

Stress can promote palatable food intake, and consumption of palatable foods may dampen psychological and physiological responses to stress. Here we develop a rat model of daily limited sweetened drink intake to further examine the linkage between consumption of preferred foods and hypothalamic-pituitary-adrenocortical axis responses to acute and chronic stress. Adult male rats with free access to water were given additional twice-daily access to 4 ml sucrose (30%), saccharin (0.1%; a noncaloric sweetener), or water. After 14 d of training, rats readily learned to drink sucrose and saccharin solutions. Half the rats were then given chronic variable stress (CVS) for 14 d immediately after each drink exposure; the remaining rats (nonhandled controls) consumed their appropriate drinking solution at the same time. On the morning after CVS, responses to a novel restraint stress were assessed in all rats. Multiple indices of chronic stress adaptation were effectively altered by CVS. Sucrose consumption decreased the plasma corticosterone response to restraint stress in CVS rats and nonhandled controls; these reductions were less pronounced in rats drinking saccharin. Sucrose or saccharin consumption decreased CRH mRNA expression in the paraventricular nucleus of the hypothalamus. Moreover, sucrose attenuated restraint-induced c-fos mRNA expression in the basolateral amygdala, infralimbic cortex, and claustrum. These data suggest that limited consumption of sweetened drink attenuates hypothalamic-pituitary-adrenocortical axis stress responses, and calories contribute but are not necessary for this effect. Collectively the results support the hypothesis that the intake of palatable substances represents an endogenous mechanism to dampen physiological stress responses.


Assuntos
Ingestão de Líquidos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Estresse Psicológico/fisiopatologia , Sacarose/farmacologia , Adaptação Psicológica , Hormônio Adrenocorticotrópico/sangue , Animais , Ritmo Circadiano , Ingestão de Alimentos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Long-Evans , Estresse Psicológico/metabolismo
20.
Am J Physiol Endocrinol Metab ; 292(4): E1173-82, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17179393

RESUMO

It is well established that estrogens markedly enhance the glucocorticoid response to acute stress in females. However, the precise mechanism responsible for this regulation is poorly understood. Here, we tested whether estrogens enhance the activation of the paraventricular nucleus (PVN) of the hypothalamus by measuring stress-induced c-fos mRNA expression in the PVN of restraint-stressed ovariectomized (OVX) rats treated with physiologically relevant doses of estradiol (E(2)), the major female estrogen. As expected, E(2) enhanced plasma corticosterone responses to restraint in OVX females. However, E(2) markedly attenuated the stress-induced c-fos gene expression in the PVN and inhibited plasma ACTH responses in these animals. Furthermore, E(2)-inhibitory effects were mimicked by progesterone (P) alone or in combination with E(2). Interestingly, the suppressive central effects of both E(2) and P were apparently independent of basal paraventricular corticotropin-releasing hormone (CRH) transcription, since these ovarian steroids did not significantly affect PVN CRH mRNA expression in unstressed rats. These unexpected findings suggested that E(2) promotes glucocorticoid hypersecretion in females by additional peripheral (i.e., adrenal) mechanisms. Indeed, E(2) markedly enhanced plasma corticosterone responses and adrenal corticosterone content in dexamethasone-blocked OVX rats challenged with varying doses of exogenous ACTH. These results suggest that enhanced adrenal sensitive to ACTH is an important physiological mechanism mediating E(2)-related glucocorticoid hypersecretion in stressed females.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/metabolismo , Estradiol/farmacologia , Fatores Sexuais , Estresse Fisiológico/fisiopatologia , Hormônio Adrenocorticotrópico/farmacologia , Animais , Corticosterona/sangue , Relação Dose-Resposta a Droga , Implantes de Medicamento , Sinergismo Farmacológico , Estradiol/administração & dosagem , Estradiol/sangue , Feminino , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Injeções Subcutâneas , Aprendizagem em Labirinto , Ovariectomia , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Progesterona/sangue , Progesterona/farmacologia , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Restrição Física , Estresse Fisiológico/etiologia , Estresse Fisiológico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...