Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nervenarzt ; 92(5): 433-440, 2021 May.
Artigo em Alemão | MEDLINE | ID: mdl-33502576

RESUMO

Besides cognitive behavioral therapy (CBT), psychopharmacotherapy belongs to the first-line treatment approaches for anxiety disorders according to all national and international guidelines. According to studies and meta-analyses, modern antidepressants in particular have been proven to be effective. Depending on the substance, there are approvals for panic disorder, generalized anxiety disorder and social phobia. There are also approvals for other substance groups, e.g. anticonvulsants for generalized anxiety disorder. Benzodiazepines should be used with caution in view of the risk of dependency. Although effective and well-tolerated medications are available, up to 30% of patients still do not respond or do not respond adequately to treatment. Consequently, research efforts to develop new substances are important. Based on a better understanding of the complex neurobiological mechanisms underlying anxiety disorders, a large number of substances are currently undergoing clinical trials. Modulators of current and new transmitter systems, in particular the glutamatergic and the endocannabinoid systems as well as neuropeptides, are being discussed as innovative substances. Strategies are also being investigated which, in combination with psychotherapy, aim at optimizing fear extinction memory. First studies are also underway on the use of psychedelic agents in combination with psychotherapy for anxiety.


Assuntos
Terapia Cognitivo-Comportamental , Transtorno de Pânico , Transtornos de Ansiedade/diagnóstico , Transtornos de Ansiedade/tratamento farmacológico , Extinção Psicológica , Medo , Humanos
2.
Mol Psychiatry ; 20(7): 901-12, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25510511

RESUMO

Traumatic fear memories are highly durable but also dynamic, undergoing repeated reactivation and rehearsal over time. Although overly persistent fear memories underlie anxiety disorders, such as posttraumatic stress disorder, the key neural and molecular mechanisms underlying fear memory durability remain unclear. Postsynaptic density 95 (PSD-95) is a synaptic protein regulating glutamate receptor anchoring, synaptic stability and certain types of memory. Using a loss-of-function mutant mouse lacking the guanylate kinase domain of PSD-95 (PSD-95(GK)), we analyzed the contribution of PSD-95 to fear memory formation and retrieval, and sought to identify the neural basis of PSD-95-mediated memory maintenance using ex vivo immediate-early gene mapping, in vivo neuronal recordings and viral-mediated knockdown (KD) approaches. We show that PSD-95 is dispensable for the formation and expression of recent fear memories, but essential for the formation of precise and flexible fear memories and for the maintenance of memories at remote time points. The failure of PSD-95(GK) mice to retrieve remote cued fear memory was associated with hypoactivation of the infralimbic (IL) cortex (but not the anterior cingulate cortex (ACC) or prelimbic cortex), reduced IL single-unit firing and bursting, and attenuated IL gamma and theta oscillations. Adeno-associated virus-mediated PSD-95 KD in the IL, but not the ACC, was sufficient to impair recent fear extinction and remote fear memory, and remodel IL dendritic spines. Collectively, these data identify PSD-95 in the IL as a critical mechanism supporting the durability of fear memories over time. These preclinical findings have implications for developing novel approaches to treating trauma-based anxiety disorders that target the weakening of overly persistent fear memories.


Assuntos
Córtex Cerebral/fisiologia , Medo/fisiologia , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Memória/fisiologia , Potenciais de Ação/fisiologia , Animais , Córtex Cerebral/citologia , Condicionamento Clássico/fisiologia , Sinais (Psicologia) , Espinhas Dendríticas/metabolismo , Proteína 4 Homóloga a Disks-Large , Eletrodos Implantados , Eletrochoque , Extinção Psicológica/fisiologia , Feminino , Reação de Congelamento Cataléptica/fisiologia , Ritmo Gama/fisiologia , Técnicas de Silenciamento de Genes , Guanilato Quinases/genética , Masculino , Proteínas de Membrana/genética , Camundongos Mutantes , Percepção Olfatória/fisiologia , Células Piramidais/citologia , Células Piramidais/fisiologia , Ritmo Teta/fisiologia
3.
J Neurophysiol ; 114(4): 2500-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26334021

RESUMO

Substance P (SP) is implicated in stress regulation and affective and anxiety-related behavior. Particularly high expression has been found in the main output region of the amygdala complex, the central amygdala (CE). Here we investigated the cellular mechanisms of SP in CE in vitro, taking advantage of glutamic acid decarboxylase-green fluorescent protein (GAD67-GFP) knockin mice that yield a reliable labeling of GABAergic neurons, which comprise 95% of the neuronal population in the lateral section of CE (CEl). In GFP-positive neurons within CEl, SP caused a membrane depolarization and increase in input resistance, associated with an increase in action potential firing frequency. Under voltage-clamp conditions, the SP-specific membrane current reversed at -101.5 ± 2.8 mV and displayed inwardly rectifying properties indicative of a membrane K(+) conductance. Moreover, SP responses were blocked by the neurokinin type 1 receptor (NK1R) antagonist L-822429 and mimicked by the NK1R agonist [Sar(9),Met(O2)(11)]-SP. Immunofluorescence staining confirmed localization of NK1R in GFP-positive neurons in CEl, predominantly in PKCδ-negative neurons (80%) and in few PKCδ-positive neurons (17%). Differences in SP responses were not observed between the major types of CEl neurons (late firing, regular spiking, low-threshold bursting). In addition, SP increased the frequency and amplitude of GABAergic synaptic events in CEl neurons depending on upstream spike activity. These data indicate a NK1R-mediated increase in excitability and GABAergic activity in CEl neurons, which seems to mostly involve the PKCδ-negative subpopulation. This influence can be assumed to increase reciprocal interactions between CElon and CEloff pathways, thereby boosting the medial CE (CEm) output pathway and contributing to the anxiogenic-like action of SP in the amygdala.


Assuntos
Núcleo Central da Amígdala/fisiologia , Neurônios GABAérgicos/fisiologia , Receptores da Neurocinina-1/metabolismo , Substância P/metabolismo , Animais , Núcleo Central da Amígdala/efeitos dos fármacos , Imunofluorescência , Neurônios GABAérgicos/efeitos dos fármacos , Técnicas de Introdução de Genes , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Potássio/metabolismo , Proteína Quinase C-delta/metabolismo , Técnicas de Cultura de Tecidos
5.
Neuroscience ; 158(4): 1717-30, 2009 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-19121371

RESUMO

There is growing evidence that neuropeptide Y (NPY) acting through Y1 and Y2 receptors has a prominent role in modulating anxiety- and depression-like behavior in rodents. However, a role of other Y-receptors like that of Y4 receptors in this process is poorly understood. We now investigated male Y2, Y4 single and Y2/Y4 double knockout mice in behavioral paradigms for changes in motor activity, anxiety and depression-like behavior. Motor activity was increased in Y2, Y4 and Y2/Y4 knockout mice under changing and stressful conditions, but not altered in a familiar environment. Y4 and Y2 knockout mice revealed an anxiolytic phenotype in the light/dark test, marble burying test and in stress-induced hyperthermia, and reduced depression-like behavior in the forced swim and tail suspension tests. In Y2/Y4 double knockout mice, the response in the light/dark test and in the forced swim test was further enhanced compared with Y4 and Y2 knockout mice, respectively. High levels of Y4 binding sites were observed in brain stem nuclei including nucleus of solitary tract and area postrema. Lower levels were found in the medial amygdala and hypothalamus. Peripheral administration of pancreatic polypeptide (PP) induced Y4 receptor-dependent c-Fos expression in brain stem, hypothalamus and amygdala. PP released peripherally from the pancreas in response to food intake, may act not only as a satiety signal but also modulate anxiety-related locomotion.


Assuntos
Depressão/genética , Depressão/fisiopatologia , Comportamento Exploratório/fisiologia , Atividade Motora/genética , Receptores de Neuropeptídeo Y/deficiência , Tonsila do Cerebelo/metabolismo , Animais , Autorradiografia/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Elevação dos Membros Posteriores/métodos , Hipertermia Induzida , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polipeptídeo Pancreático/farmacologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Natação/fisiologia
6.
Mol Psychiatry ; 13(11): 1028-42, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18475271

RESUMO

Hypersecretion of central corticotropin-releasing hormone (CRH) has been implicated in the pathophysiology of affective disorders. Both, basic and clinical studies suggested that disrupting CRH signaling through CRH type 1 receptors (CRH-R1) can ameliorate stress-related clinical conditions. To study the effects of CRH-R1 blockade upon CRH-elicited behavioral and neurochemical changes we created different mouse lines overexpressing CRH in distinct spatially restricted patterns. CRH overexpression in the entire central nervous system, but not when overexpressed in specific forebrain regions, resulted in stress-induced hypersecretion of stress hormones and increased active stress-coping behavior reflected by reduced immobility in the forced swim test and tail suspension test. These changes were related to acute effects of overexpressed CRH as they were normalized by CRH-R1 antagonist treatment and recapitulated the effect of stress-induced activation of the endogenous CRH system. Moreover, we identified enhanced noradrenergic activity as potential molecular mechanism underlying increased active stress-coping behavior observed in these animals. Thus, these transgenic mouse lines may serve as animal models for stress-elicited pathologies and treatments that target the central CRH system.


Assuntos
Sistema Nervoso Central/metabolismo , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Estresse Fisiológico/genética , Estresse Psicológico/genética , Adaptação Psicológica/efeitos dos fármacos , Adaptação Psicológica/fisiologia , Análise de Variância , Animais , Química Encefálica/efeitos dos fármacos , Sistema Nervoso Central/anatomia & histologia , Sistema Nervoso Central/efeitos dos fármacos , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Comportamento Exploratório , Feminino , Fenclonina/administração & dosagem , Fenclonina/análogos & derivados , Elevação dos Membros Posteriores , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Proteínas de Filamentos Intermediários/genética , Masculino , Metiltirosinas/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Nestina , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas/genética , Pirazóis/farmacologia , RNA não Traduzido , Radioimunoensaio/métodos , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/etiologia , Natação , Triazinas/farmacologia
7.
Stress ; 12(1): 58-69, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19116889

RESUMO

Chronic subordinate colony (CSC) housing has been recently validated as a murine model of chronic psychosocial stress which induces alterations of stress-related parameters including decreased body-weight gain and an increased level of anxiety in comparison with single housed control (SHC) mice. By using immunohistochemical immediate early gene (IEG) mapping we investigated whether CSC housing causes alterations in neuronal activation patterns in limbic areas including the amygdala, hippocampus, septum and the periaqueductal gray (PAG) and hypothalamic paraventricular nucleus (PVN). While CSC housing increased basal Zif-268 expression in the nucleus accumbens shell compared to SHC, IEG responses to subsequent open arm (OA) exposure were attenuated in the ventral and intermediate sub-regions of the lateral septum, parvocellular PVN and the dorsal CA3 region of the hippocampus of CSC compared with SHC mice. In contrast, a potentiated c-Fos response in CSC mice was observed in the dorsomedial PAG after OA exposure. Confirming previous findings obtained on the elevated plus-maze, an enhanced anxiety-related behavior in CSC compared with SHC mice was also observed during OA exposure. In order to investigate the appropriate control conditions for CSC housing, group housed control (GHC) mice were additionally included in the behavioral testing. Interestingly, GHC as well as CSC mice showed significantly less risk assessment/exploratory behavior during OA exposure compared with SHC mice indicating that group housing itself is stressful for mice and not an adequate control for the CSC paradigm. Overall, CSC housing is an ethologically relevant chronic psychosocial stressor which results in an elevated sensitivity to a subsequent novel, aversive challenge. However, the CSC-induced increase in anxiety-related behavior was accompanied by differences in neuronal activation, compared with SHC, in defined sub-regions of brain areas known to be involved in the processing of emotionality and stress responses.


Assuntos
Encéfalo/fisiopatologia , Predomínio Social , Estresse Psicológico/fisiopatologia , Animais , Ansiedade/psicologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Expressão Gênica , Abrigo para Animais , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-fos/metabolismo
8.
Amino Acids ; 36(1): 147-58, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18975044

RESUMO

A relationship between zinc (Zn)-deficiency and mood disorders has been suspected. Here we examined for the first time whether experimentally-induced Zn-deficiency in mice would alter depression- and anxiety-related behaviour assessed in established tests and whether these alterations would be sensitive to antidepressant treatment. Mice receiving a Zn-deficient diet (40% of daily requirement) had similar homecage and open field activity compared to normally fed mice, but displayed enhanced depression-like behaviour in both the forced swim and tail suspension tests which was reversed by chronic desipramine treatment. An anxiogenic effect of Zn-deficiency prevented by chronic desipramine and Hypericum perforatum treatment was observed in the novelty suppressed feeding test, but not in other anxiety tests performed. Zn-deficient mice showed exaggerated stress-evoked immediate-early gene expression in the amygdala which was normalised following DMI treatment. Taken together these data support the link between low Zn levels and depression-like behaviour and suggest experimentally-induced Zn deficiency as a putative model of depression in mice.


Assuntos
Antidepressivos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Depressão/metabolismo , Sistema Límbico/efeitos dos fármacos , Sistema Límbico/fisiopatologia , Zinco/deficiência , Animais , Peso Corporal/efeitos dos fármacos , Sistema Límbico/metabolismo , Sistema Límbico/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos
9.
Psychoneuroendocrinology ; 110: 104430, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31542636

RESUMO

BACKGROUND: Obesity is a risk factor for stress-related mental disorders such as post-traumatic stress disorder. The underlying mechanism through which obesity affects mental health remains poorly understood but dysregulation of the ghrelin system may be involved. Stress increases plasma ghrelin levels, which stimulates food intake as a potential stress-coping mechanism. However, diet-induced obesity induces ghrelin resistance which in turn may have deleterious effects on stress-coping. In our study, we explored whether disruption of ghrelin receptor function though high-fat diet or genetic ablation affects fear processing, anxiety-like behavior and saccharin preference in mice. METHODS: Adult male C57BL6/J mice were placed on a standard diet or high-fat diet for a total period of 8 weeks. We first established that high-fat diet exposure for 4 weeks elicits ghrelin resistance, evidenced by a blunted hyperphagic response following administration of a ghrelin receptor agonist. We then carried out an experiment in which we subjected mice to auditory fear conditioning after 4 weeks of diet exposure and evaluated effects on fear extinction, anxiety-like behavior and saccharin preference. To explore whether fear conditioning as such may influence the effect of diet exposure, we also subjected mice to auditory fear conditioning prior to diet onset and 4 weeks later we investigated auditory fear extinction, anxiety-like behavior and saccharin preference. In a final experiment, we further assessed lack of ghrelin receptor function by investigating auditory fear processing, anxiety-like behavior and saccharin preference in ghrelin receptor knockout mice and their wild-type littermates. RESULTS: High-fat diet exposure had no significant effect on auditory fear conditioning and its subsequent extinction or on anxiety-like behavior but significantly lowered saccharin preference. Similarly, ghrelin receptor knockout mice did not differ significantly from their wild-type littermates for auditory fear processing or anxiety-like behavior but showed significantly lower saccharin preference compared to wild-type littermates. CONCLUSION: Taken together, our data suggest that disruption of ghrelin receptor function per se does not affect fear or anxiety-like behavior but may decrease saccharin preference in mice.


Assuntos
Ansiedade/genética , Medo , Preferências Alimentares , Receptores de Grelina/genética , Sacarina/administração & dosagem , Animais , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Medo/psicologia , Preferências Alimentares/psicologia , Deleção de Genes , Grelina/fisiologia , Hiperfagia/genética , Hiperfagia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Grelina/fisiologia
10.
Genes Brain Behav ; 17(3): e12427, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29028150

RESUMO

The current treatment of post-traumatic stress disorder (PTSD), phobias and other anxiety disorders, remains insufficient particularly in producing long-lasting full symptom control. Dysfunctional fear processing is common in these disorders, including a deficiency in fear-inhibitory mechanisms and impairment in the ability to discriminate between safety and danger cues. Research has aimed to elucidate brain circuitries, neurotransmitters and downstream signaling pathways important in the alleviation of aberrant fear, with a specific focus on mechanisms modulating fear memory and its behavioral expression. MicroRNAs (miRNA) as "fine tuners" of gene expression at the post-transcriptional level have emerged as critical regulators of such mechanisms important in both, the generation and the inhibition of fear memories. Along these lines, abnormal expression of miRNAs has been associated with different fear-related disorders. After providing an updated overview on the involvement of miRNAs in fear learning mechanisms, we summarize and discuss in particular those studies in which the implication of miRNAs in successful inhibition of fear has been explored. For a better overview, we dissociate the different modes of fear alleviation investigated in this regard and present studies in rodents demonstrating that specific miRNAs are involved in the destabilization of fear by interfering with consolidation/reconsolidation mechanisms or that they are associated with the generation of fear extinction or safety learning. Finally, we discuss the potential of miRNAs as biomarkers and novel therapeutic targets, as well as the challenges involved in applying the discovered mechanisms in the development of improved treatments of fear- and trauma-related disorders.


Assuntos
Medo/fisiologia , Medo/psicologia , MicroRNAs/genética , Animais , Transtornos de Ansiedade/fisiopatologia , Encéfalo/fisiopatologia , Condicionamento Clássico/fisiologia , Humanos , Aprendizagem/fisiologia , Memória/fisiologia , MicroRNAs/uso terapêutico , Plasticidade Neuronal/genética , Transdução de Sinais/fisiologia , Transtornos de Estresse Pós-Traumáticos/fisiopatologia
11.
Neuroscience ; 139(3): 1005-15, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16542784

RESUMO

Ca(V)1.2 and Ca(V)1.3, are the main dihydropyridine-sensitive L-type calcium channel isoforms in the brain. To reveal the contribution of each isoform to the neuronal activation pattern elicited by the dihydropyridine L-type calcium channel activator BayK 8644, we utilized Fos expression as a marker of neuronal activation in mutant mice (Ca(V)1.2(DHP-/-) mice) expressing dihydropyridine-insensitive Ca(V)1.2 L-type calcium channels. BayK 8644-treated wildtype mice displayed intense and widespread Fos expression throughout the neuroaxis in 77 of 80 brain regions quantified. The Fos response in Ca(V)1.2(DHP-/-) mice was greatly attenuated or absent in most of these areas, suggesting that a major part of the widespread Fos induction including most cortical areas was mediated by Ca(V)1.2 L-type calcium channels. BayK 8644-induced Fos expression in Ca(V)1.2(DHP-/-) mice indicating predominantly Ca(V)1.3 L-type calcium channel-mediated activation was noted in more restricted neuronal populations (20 of 80), in particular in the central amygdala, the bed nucleus of the stria terminalis, paraventricular hypothalamic nucleus, lateral preoptic area, locus coeruleus, lateral parabrachial nucleus, central nucleus of the inferior colliculus, and nucleus of the solitary tract. Our data indicate that selective stimulation of other than Ca(V)1.2 L-type calcium channels, mostly Ca(V)1.3, causes neuronal activation in a specific set of mainly limbic, hypothalamic and brainstem areas, which are associated with functions including integration of emotion-related behavior. Hence, selective modulation of Ca(V)1.3 L-type calcium channels could represent a novel (pharmacotherapeutic) tool to influence these CNS functions.


Assuntos
Encéfalo/metabolismo , Canais de Cálcio Tipo L/metabolismo , Neurônios/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Isoformas de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo
12.
Prog Neurobiol ; 56(2): 237-67, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9760703

RESUMO

In the past 15 years the release of neurotransmitters and their metabolites in the locus coeruleus (LC) has been studied by using three approaches: microdialysis; push-pull superfusion; and voltammetry. These sophisticated techniques, which render it possible to follow the time course and magnitude of neurochemical changes in anaesthetized and conscious animals, have permitted great strides towards understanding neurotransmission in the LC. It appears that noradrenaline, known to be released in distant terminal fields, is also released in the somatodendritic area of LC neurons in response to drugs and physiological stimuli. Furthermore, determination of in vivo release enables the identification of functionally important neurotransmitter systems involved in relaying and integrating information reaching the LC via afferent neurons. As outlined in this review, the release rates of glutamate, aspartate, gamma-aminobutyric acid, glycine, 5-hydroxytryptamine and catecholamines, are modified in particular by arousing and stressful stimuli, pain, changes in cardiovascular homeostasis, as well as during opioid withdrawal or the sleep-wake-cycle. Profound interactions also occur between some of the neurotransmitters released during these situations. It appears that individual stimuli produce distinct neurochemical changes which contribute to the regulation of neuronal LC activity. Stimuli that activate LC neurons, such as pain, fall of blood pressure, noise, opiate withdrawal, do not produce a uniform response but modality-specific release patterns of excitatory and inhibitory neurotransmitters within the LC. From these studies and from existing neuroanatomical and electrophysiological data our knowledge of how neurotransmitters work in concert to regulate the functional state of LC noradrenergic perikarya in physiological and pathophysiological conditions is just emerging.


Assuntos
Locus Cerúleo/metabolismo , Neurotransmissores/metabolismo , Transmissão Sináptica/fisiologia , Animais , Humanos
13.
Transl Psychiatry ; 6(12): e974, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27922638

RESUMO

Extinction-based exposure therapy is used to treat anxiety- and trauma-related disorders; however, there is the need to improve its limited efficacy in individuals with impaired fear extinction learning and to promote greater protection against return-of-fear phenomena. Here, using 129S1/SvImJ mice, which display impaired fear extinction acquisition and extinction consolidation, we revealed that persistent and context-independent rescue of deficient fear extinction in these mice was associated with enhanced expression of dopamine-related genes, such as dopamine D1 (Drd1a) and -D2 (Drd2) receptor genes in the medial prefrontal cortex (mPFC) and amygdala, but not hippocampus. Moreover, enhanced histone acetylation was observed in the promoter of the extinction-regulated Drd2 gene in the mPFC, revealing a potential gene-regulatory mechanism. Although enhancing histone acetylation, via administering the histone deacetylase (HDAC) inhibitor MS-275, does not induce fear reduction during extinction training, it promoted enduring and context-independent rescue of deficient fear extinction consolidation/retrieval once extinction learning was initiated as shown following a mild conditioning protocol. This was associated with enhanced histone acetylation in neurons of the mPFC and amygdala. Finally, as a proof-of-principle, mimicking enhanced dopaminergic signaling by L-dopa treatment rescued deficient fear extinction and co-administration of MS-275 rendered this effect enduring and context-independent. In summary, current data reveal that combining dopaminergic and epigenetic mechanisms is a promising strategy to improve exposure-based behavior therapy in extinction-impaired individuals by initiating the formation of an enduring and context-independent fear-inhibitory memory.


Assuntos
Dopamina/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Histona Acetiltransferases/fisiologia , Transdução de Sinais/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Benzamidas/farmacologia , Terapia Combinada , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Terapia Implosiva , Levodopa/farmacologia , Masculino , Camundongos , Córtex Pré-Frontal/fisiologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
14.
Trends Pharmacol Sci ; 17(10): 356-63, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8979770

RESUMO

Biogenic amines and amino acids have been implicated in central cardiovascular homeostasis. Initially, drugs were injected into the brain and their effects on blood pressure were investigated. Other approaches allowed endogenous neurotransmitters released in the extracellular space of brain structures involved in cardiovascular regulation to be identified. As Nicolas Singewald and Athineos Philippu outline, even slight disturbances in blood pressure and/or isovolaemia lead to marked changes in the release rates of biogenic amines and amino acids in various brain structures. Blood pressure homeostasis is maintained with the participation of several brain regions and neurotransmitters which possess the same or opposing functions when released from CNS neurones.


Assuntos
Aminoácidos/fisiologia , Aminas Biogênicas/fisiologia , Fenômenos Fisiológicos Cardiovasculares , Homeostase/fisiologia , Animais , Humanos
15.
Pharmacol Ther ; 149: 150-90, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25550231

RESUMO

Pathological fear and anxiety are highly debilitating and, despite considerable advances in psychotherapy and pharmacotherapy they remain insufficiently treated in many patients with PTSD, phobias, panic and other anxiety disorders. Increasing preclinical and clinical evidence indicates that pharmacological treatments including cognitive enhancers, when given as adjuncts to psychotherapeutic approaches [cognitive behavioral therapy including extinction-based exposure therapy] enhance treatment efficacy, while using anxiolytics such as benzodiazepines as adjuncts can undermine long-term treatment success. The purpose of this review is to outline the literature showing how pharmacological interventions targeting neurotransmitter systems including serotonin, dopamine, noradrenaline, histamine, glutamate, GABA, cannabinoids, neuropeptides (oxytocin, neuropeptides Y and S, opioids) and other targets (neurotrophins BDNF and FGF2, glucocorticoids, L-type-calcium channels, epigenetic modifications) as well as their downstream signaling pathways, can augment fear extinction and strengthen extinction memory persistently in preclinical models. Particularly promising approaches are discussed in regard to their effects on specific aspects of fear extinction namely, acquisition, consolidation and retrieval, including long-term protection from return of fear (relapse) phenomena like spontaneous recovery, reinstatement and renewal of fear. We also highlight the promising translational value of the preclinial research and the clinical potential of targeting certain neurochemical systems with, for example d-cycloserine, yohimbine, cortisol, and L-DOPA. The current body of research reveals important new insights into the neurobiology and neurochemistry of fear extinction and holds significant promise for pharmacologically-augmented psychotherapy as an improved approach to treat trauma and anxiety-related disorders in a more efficient and persistent way promoting enhanced symptom remission and recovery.


Assuntos
Ansiedade/tratamento farmacológico , Extinção Psicológica/efeitos dos fármacos , Terapia Implosiva , Nootrópicos/farmacologia , Transtornos Fóbicos/tratamento farmacológico , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Transmissão Sináptica/efeitos dos fármacos , Animais , Ansiedade/terapia , Terapia Combinada , Humanos , Modelos Neurológicos , Nootrópicos/uso terapêutico , Transdução de Sinais/efeitos dos fármacos
16.
Neuropsychopharmacology ; 29(11): 2074-80, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15187982

RESUMO

Recent studies in rodents have shown that withdrawal from chronic drug abuse is associated with a significant decrease in dopamine (DA) release in mesolimbic structures, especially in the shell region of the nucleus accumbens. Since the DA system is known to play an important role in reward processes, a withdrawal-associated impairment in mesolimbic DA-mediated transmission could possibly implicate reward deficit and thus enhance vulnerability to drug craving and relapse. We have previously demonstrated that acute repetitive transcranial magnetic stimulation (rTMS) has a modulatory effect on DA release in several areas of the rat brain, including dorsal striatum, hippocampus, and nucleus accumbens shell. In the present study, we investigated the possible use of rTMS as a tool in re-establishing the dysregulated DA secretion observed during withdrawal in morphine-sensitized male Sprague-Dawley rats. Using intracerebral microdialysis, we monitored the effects of acute rTMS (20 Hz) on the intra-accumbal release-patterns of DA in freely moving animals that were subjected to a morphine sensitization scheme for a period of 8 days. We provide first evidence that acute rTMS (20 Hz) is able to increase DA concentration in the shell region of the nucleus accumbens in both control animals and morphine-sensitized rats during abstinence. The DA release in morphine-sensitized rats was significantly higher than in controls. rTMS, therefore, might gain a potential therapeutic role in the treatment of dysphoric and anhedonic states during drug withdrawal in humans.


Assuntos
Dopamina/metabolismo , Campos Eletromagnéticos , Morfina/farmacologia , Núcleo Accumbens/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Animais , Masculino , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Síndrome de Abstinência a Substâncias/terapia , Fatores de Tempo
17.
Neuropharmacology ; 46(8): 1177-83, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15111024

RESUMO

Clinically effective antidepressants are thought to exert their therapeutic effects by facilitating central monoamine neurotransmission. However, recent data showing that neurokinin-1 receptor (NK1R) antagonists have antidepressant properties in both animal and clinical studies raise the possibility that classical antidepressants may also influence NK1R expression in the brain. To test this hypothesis, rats were treated with desipramine, paroxetine, venlafaxine, tranylcypromine or vehicle for 14-42 days. NK1R binding sites and mRNA were determined in a wide variety of brain areas using in situ hybridization and quantitative receptor autoradiography. In all areas examined, the abundance of NK1R binding sites was unchanged after 14 days of treatment. None of the treatments altered the number of NK1R binding sites following 42 days treatment with the exception that an increase was found in the locus coeruleus with tranylcypromine. Taken together, we report that repeated treatment with antidepressants of different classes does not cause significant changes in NK1R expression.


Assuntos
Antidepressivos/administração & dosagem , Encéfalo/efeitos dos fármacos , Receptores da Neurocinina-1/biossíntese , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Encéfalo/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Masculino , Antagonistas dos Receptores de Neurocinina-1 , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-1/metabolismo
18.
Neuroscience ; 98(4): 759-70, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10891619

RESUMO

It is speculated that specific hindbrain transmitter pathways centred on the periaqueductal gray and locus coeruleus are an important integrative neural substrate for the expression of anxiety and the somatic symptoms and cardiovascular changes that accompany severe anxiety states, such as in panic disorder. Here we investigated the effects of various drugs, known to induce panic in humans and to be anxiogenic in animals, on Fos expression in the periaqueductal gray, locus coeruleus and other parts of the rat hindbrain. The drugs tested were the benozodiazepine inverse agonist FG-7142, the alpha(2)-adrenoceptor antagonist yohimbine, the non-selective 5-hydroxytryptamine(2C) receptor agonist m-chlorophenyl piperazine, the adenosine antagonist caffeine and the cholecystokinin analogue BOC-CCK(4). A clear-cut finding was that administration of each anxiogenic drug caused a striking region-specific pattern of Fos expression within the hindbrain. In particular, the drugs commonly increased Fos-like immunoreactivity in the periaqueductal gray and locus coeruleus. Increased Fos expression in the periaqueductal gray was specific to the rostral dorsolateral and caudal ventrolateral regions. All the anxiogenic drugs also increased Fos-like immunoreactivity in the lateral parabrachial nucleus and nucleus of the solitary tract and all but one (BOC-CCK(4)) increased Fos in the dorsal raphe nucleus. Rats habituated to the test environment and injected with saline vehicle displayed little or no Fos-like immunoreactivity in the hindbrain areas investigated. In summary, each of the anxiogenic drugs tested (FG-7142, yohimbine, m-chlorophenyl piperazine, caffeine and BOC-CCK(4)) increased Fos expression in a restricted number of hindbrain regions, including the periaqueductal gray and locus coeruleus. Previous Fos studies have found that these same regions are activated by various fearful environmental stimuli. Therefore, a specific set of hindbrain circuits may be commonly involved in the processing of anxiety-related information evoked by pharmacological and environmental manipulation. The present findings also raise the possibility that measurement of the effect of anxiogenic drugs on Fos expression might be a useful way to model hindbrain pathways activated by anxiety and possibly panic.


Assuntos
Ansiedade/metabolismo , Genes fos/fisiologia , Pânico/fisiologia , Rombencéfalo/metabolismo , Antagonistas Adrenérgicos alfa , Animais , Ansiedade/induzido quimicamente , Cafeína , Carbolinas , Estimulantes do Sistema Nervoso Central , Antagonistas GABAérgicos , Imuno-Histoquímica , Masculino , Ratos , Rombencéfalo/efeitos dos fármacos , Ioimbina
19.
Br J Pharmacol ; 123(4): 746-52, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9517395

RESUMO

1. The interactions between 5-hydroxytryptaminergic neurones and excitatory amino acid utilizing neurones were studied in the locus coeruleus of conscious, freely moving rats. The locus coeruleus was superfused with artificial cerebrospinal fluid through a push-pull cannula and 5-hydroxytryptamine (5-HT) was determined in the superfusate that was continuously collected in time periods of 10 min. 2. Superfusion of the locus coeruleus with the NMDA receptor antagonist AP5 (10 microM), kynurenic acid (1 mM), or the AMPA/kainate receptor antagonist DNQX (10 microM) reduced the 5-HT release in the locus coeruleus. 3. Superfusion with the agonists NMDA (50 microM), kainic acid (50 microM) or AMPA (10 microM) enhanced the release rate of 5-HT. AP5 (10 microM) blocked the stimulant effect of NMDA, while tetrodotoxin (1 microM) failed to influence the NMDA-induced release of 5-HT. In the presence of 10 microM DNQX, the releasing effect of 50 microM kainic acid was abolished. 4. Pain elicited by tail pinch, as well as noise-induced stress, increased the release of 5-HT. Superfusion of the locus coeruleus with 10 microM AP5 reduced the tail pinch-induced 5-HT release. AP5 (10 microM) did not affect the noise-induced release of 5-HT which was reduced, when the locus coeruleus was superfused simultaneously with this concentration of AP5 and 1 microM kynurenic acid. DNQX (10 mM) failed to influence the release of 5-HT induced by tail pinch or noise. 5. The findings suggest that 5-hydroxytryptaminergic neurones of the locus coeruleus are tonically modulated by excitatory amino acids via NMDA and AMPA/kainate receptors. The release of 5-HT elicited by tail pinch and noise is mediated to a considerable extent through endogenous excitatory amino acids acting on NMDA receptors, while AMPA/kainate receptors are not involved in this process.


Assuntos
Estimulação Acústica , Aminoácidos Excitatórios/farmacologia , Locus Cerúleo/efeitos dos fármacos , Dor , Serotonina/metabolismo , Animais , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Locus Cerúleo/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/efeitos dos fármacos , Receptores de Glutamato/metabolismo
20.
Neuroreport ; 5(14): 1709-12, 1994 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-7827313

RESUMO

The locus coeruleus (LC) of anaesthetized rats was superfused with artificial cerebrospinal fluid through a push-pull cannula and the release of the amino acids gamma-aminobutyric acid (GABA), taurine and arginine was determined in the superfusate. Increases in blood pressure (BP) induced either by intravenous infusions of noradrenaline and phenylephrine or by blood injection enhanced the release of GABA in the LC. Decreases in BP elicited by intravenous infusion of sodium nitroprusside or by haemorrhage decreased the GABA release rate. The BP changes did not influence the release rates of taurine and arginine. These findings demonstrate that GABA release in the LC is modified by cardiovascular impulses and suggest that GABAergic neurones modulate LC activity in response to disturbances in BP homeostasis.


Assuntos
Pressão Sanguínea/fisiologia , Homeostase/fisiologia , Locus Cerúleo/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Arginina/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Volume Sanguíneo/fisiologia , Hemorragia/fisiopatologia , Homeostase/efeitos dos fármacos , Locus Cerúleo/citologia , Locus Cerúleo/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Nitroprussiato/farmacologia , Norepinefrina/farmacologia , Fenilefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Taurina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA