Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(26): E2428-36, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23754384

RESUMEN

Traumatic events can engender persistent excessive fear responses to trauma reminders that may return even after successful treatment. Extinction, the laboratory analog of behavior therapy, does not erase conditioned fear memories but generates competing, fear-inhibitory "extinction memories" that, however, are tied to the context in which extinction occurred. Accordingly, a dominance of fear over extinction memory expression--and, thus, return of fear--is often observed if extinguished fear stimuli are encountered outside the extinction (therapy) context. We show that postextinction administration of the dopamine precursor L-dopa makes extinction memories context-independent, thus strongly reducing the return of fear in both mice and humans. Reduced fear is accompanied by decreased amygdala and enhanced ventromedial prefrontal cortex activation in both species. In humans, ventromedial prefrontal cortex activity is predicted by enhanced resting-state functional coupling of the area with the dopaminergic midbrain during the postextinction consolidation phase. Our data suggest that dopamine-dependent boosting of extinction memory consolidation is a promising avenue to improving anxiety therapy.


Asunto(s)
Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Levodopa/administración & dosificación , Memoria/efectos de los fármacos , Adulto , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Extinción Psicológica/fisiología , Miedo/fisiología , Humanos , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología
2.
Lab Anim (NY) ; 53(3): 67-79, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38438748

RESUMEN

Although biomedical research is experiencing a data explosion, the accumulation of vast quantities of data alone does not guarantee a primary objective for science: building upon existing knowledge. Data collected that lack appropriate metadata cannot be fully interrogated or integrated into new research projects, leading to wasted resources and missed opportunities for data repurposing. This issue is particularly acute for research using animals, where concerns regarding data reproducibility and ensuring animal welfare are paramount. Here, to address this problem, we propose a minimal metadata set (MNMS) designed to enable the repurposing of in vivo data. MNMS aligns with an existing validated guideline for reporting in vivo data (ARRIVE 2.0) and contributes to making in vivo data FAIR-compliant. Scenarios where MNMS should be implemented in diverse research environments are presented, highlighting opportunities and challenges for data repurposing at different scales. We conclude with a 'call for action' to key stakeholders in biomedical research to adopt and apply MNMS to accelerate both the advancement of knowledge and the betterment of animal welfare.


Asunto(s)
Investigación Biomédica , Metadatos , Animales , Reproducibilidad de los Resultados , Bienestar del Animal
3.
Int J Neuropsychopharmacol ; 14(10): 1341-55, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21320392

RESUMEN

Increasing evidence suggests that specific physiological measures may serve as biomarkers for successful treatment to alleviate symptoms of pathological anxiety. Studies of autonomic function investigating parameters such as heart rate (HR), HR variability and blood pressure (BP) indicated that HR variability is consistently reduced in anxious patients, whereas HR and BP data show inconsistent results. Therefore, HR and HR variability were measured under various emotionally challenging conditions in a mouse model of high innate anxiety (high anxiety behaviour; HAB) vs. control normal anxiety-like behaviour (NAB) mice. Baseline HR, HR variability and activity did not differ between mouse lines. However, after cued Pavlovian fear conditioning, both elevated tachycardia and increased fear responses were observed in HAB mice compared to NAB mice upon re-exposure to the conditioning stimulus serving as the emotional stressor. When retention of conditioned fear was tested in the home cage, HAB mice again displayed higher fear responses than NAB mice, while the HR responses were similar. Conversely, in both experimental settings HAB mice consistently exhibited reduced HR variability. Repeated administration of the anxiolytic NK1 receptor antagonist L-822429 lowered the conditioned fear response and shifted HR dynamics in HAB mice to a more regular pattern, similar to that in NAB mice. Additional receiver-operating characteristic (ROC) analysis demonstrated the high specificity and sensitivity of HR variability to distinguish between normal and high anxiety trait. These findings indicate that assessment of autonomic response in addition to freezing might be a useful indicator of the efficacy of novel anxiolytic treatments.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Sistema Nervioso Autónomo/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Corazón/inervación , Piperidinas/farmacología , Taquicardia/tratamiento farmacológico , Animales , Ansiedad/etiología , Ansiedad/fisiopatología , Ansiedad/psicología , Sistema Nervioso Autónomo/fisiopatología , Ritmo Circadiano , Condicionamiento Psicológico/efectos de los fármacos , Señales (Psicología) , Modelos Animales de Enfermedad , Electrocardiografía Ambulatoria , Electrochoque , Miedo/efectos de los fármacos , Femenino , Ratones , Actividad Motora/efectos de los fármacos , Antagonistas del Receptor de Neuroquinina-1 , Ruido , Curva ROC , Taquicardia/etiología , Taquicardia/fisiopatología , Taquicardia/psicología , Telemetría , Factores de Tiempo , Grabación en Video
4.
Front Behav Neurosci ; 15: 758274, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35242017

RESUMEN

In drug discovery and development, traditional assessment of human patients and preclinical subjects occurs at limited time points in potentially stressful surroundings (i.e., the clinic or a test arena), which can impact data quality and welfare. However, recent advances in remote digital monitoring technologies enable the assessment of human patients and preclinical subjects across multiple time points in familiar surroundings. The ability to monitor a patient throughout disease progression provides an opportunity for more relevant and efficient diagnosis as well as improved assessment of drug efficacy and safety. In preclinical in vivo animal models, these digital technologies allow for continuous, longitudinal, and non-invasive monitoring in the home environment. This manuscript provides an overview of digital monitoring technologies for use in preclinical studies including their history and evolution, current engagement through use cases, and impact of digital biomarkers (DBs) on drug discovery and the 3Rs. We also discuss barriers to implementation and strategies to overcome them. Finally, we address data consistency and technology standards from the perspective of technology providers, end-users, and subject matter experts. Overall, this review establishes an improved understanding of the value and implementation of digital biomarker (DB) technologies in preclinical research.

5.
Front Behav Neurosci ; 14: 575434, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33192366

RESUMEN

Animal models of neurodegenerative and neuropsychiatric disorders require extensive behavioral phenotyping. Currently, this presents several caveats and the most important are: (i) rodents are nocturnal animals, but mostly tested during the light period; (ii) the conventional behavioral experiments take into consideration only a snapshot of a rich behavioral repertoire; and (iii) environmental factors, as well as experimenter influence, are often underestimated. Consequently, serious concerns have been expressed regarding the reproducibility of research findings on the one hand, and appropriate welfare of the animals (based on the principle of 3Rs-reduce, refine and replace) on the other hand. To address these problems and improve behavioral phenotyping in general, several solutions have been proposed and developed. Undisturbed, 24/7 home-cage monitoring (HCM) is gaining increased attention and popularity as demonstrating the potential to substitute or complement the conventional phenotyping methods by providing valuable data for identifying the behavioral patterns that may have been missed otherwise. In this review, we will briefly describe the different technologies used for HCM systems. Thereafter, based on our experience, we will focus on two systems, IntelliCage (NewBehavior AG and TSE-systems) and Digital Ventilated Cage (DVC®, Tecniplast)-how they have been developed and applied during recent years. Additionally, we will touch upon the importance of the environmental/experimenter artifacts and propose alternative suggestions for performing phenotyping experiments based on the published evidence. We will discuss how the integration of telemetry systems for deriving certain physiological parameters can help to complement the description of the animal model to offer better translation to human studies. Ultimately, we will discuss how such HCM data can be statistically interpreted and analyzed.

6.
Eur J Neurosci ; 28(11): 2299-309, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19019199

RESUMEN

The impaired extinction of acquired fear is a core symptom of anxiety disorders, such as post-traumatic stress disorder, phobias or panic disorder, and is known to be particularly resistant to existing pharmacotherapy. We provide here evidence that a similar relationship between trait anxiety and resistance to extinction of fear memory can be mimicked in a psychopathologic animal model. Wistar rat lines selectively bred for high (HAB) or low (LAB) anxiety-related behaviour were tested in a classical cued fear conditioning task utilizing freezing responses as a measure of fear. Fear acquisition was similar in both lines. In the extinction trial, however, HAB rats showed a marked deficit in the attenuation of freezing responses to repeated auditory conditioned stimulus presentations as compared with LAB rats, which exhibited rapid extinction. To gain information concerning the putatively altered neuronal processing associated with the differential behavioural response between HAB and LAB rats, c-Fos expression was investigated in the main prefrontal-amygdala pathways important for cued fear extinction. HAB compared to LAB rats showed an attenuated c-Fos response to repeated conditioned stimulus presentations in infralimbic and cingulate cortices, as well as in the lateral amygdala, but facilitated the c-Fos response in the medial part of the central amygdala. In conclusion, the present results support the notion that impaired extinction in high anxiety rats is accompanied by an aberrant activation profile in extinction-relevant prefrontal-amygdala circuits. Thus, HAB rats may represent a clinically relevant model to study the mechanisms and potential targets to accelerate delayed extinction processes in subjects with enhanced trait anxiety.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Trastornos de Ansiedad/fisiopatología , Extinción Psicológica/fisiología , Miedo/fisiología , Aprendizaje/fisiología , Corteza Prefrontal/fisiopatología , Animales , Trastornos de Ansiedad/genética , Biomarcadores/análisis , Biomarcadores/metabolismo , Condicionamiento Psicológico/fisiología , Señales (Psicología) , Modelos Animales de Enfermedad , Giro del Cíngulo/fisiopatología , Masculino , Vías Nerviosas/fisiopatología , Neuronas/metabolismo , Pruebas Neuropsicológicas , Proteínas Proto-Oncogénicas c-fos/análisis , Proteínas Proto-Oncogénicas c-fos/metabolismo , Carácter Cuantitativo Heredable , Ratas
9.
Neuropsychopharmacology ; 39(9): 2211-20, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24663011

RESUMEN

An imbalance of the gamma-aminobutyric acid (GABA) system is considered a major neurobiological pathomechanism of anxiety, and the amygdala is a key brain region involved. Reduced GABA levels have been found in anxiety patients, and genetic variations of glutamic acid decarboxylase (GAD), the rate-limiting enzyme of GABA synthesis, have been associated with anxiety phenotypes in both humans and mice. These findings prompted us to hypothesize that a deficiency of GAD65, the GAD isoform controlling the availability of GABA as a transmitter, affects synaptic transmission and plasticity in the lateral amygdala (LA), and thereby interferes with fear responsiveness. Results indicate that genetically determined GAD65 deficiency in mice is associated with (1) increased synaptic length and release at GABAergic connections, (2) impaired efficacy of GABAergic synaptic transmission and plasticity, and (3) reduced spillover of GABA to presynaptic GABAB receptors, resulting in a loss of the associative nature of long-term synaptic plasticity at cortical inputs to LA principal neurons. (4) In addition, training with high shock intensities in wild-type mice mimicked the phenotype of GAD65 deficiency at both the behavioral and synaptic level, indicated by generalization of conditioned fear and a loss of the associative nature of synaptic plasticity in the LA. In conclusion, GAD65 is required for efficient GABAergic synaptic transmission and plasticity, and for maintaining extracellular GABA at a level needed for associative plasticity at cortical inputs in the LA, which, if disturbed, results in an impairment of the cue specificity of conditioned fear responses typifying anxiety disorders.


Asunto(s)
Amígdala del Cerebelo/enzimología , Miedo/fisiología , Generalización Psicológica/fisiología , Glutamato Descarboxilasa/metabolismo , Transmisión Sináptica/fisiología , Ácido gamma-Aminobutírico/metabolismo , Amígdala del Cerebelo/citología , Animales , Aprendizaje por Asociación/fisiología , Condicionamiento Psicológico/fisiología , Electrochoque , Espacio Extracelular/metabolismo , Glutamato Descarboxilasa/genética , Ácido Glutámico/metabolismo , Interneuronas/citología , Interneuronas/fisiología , Potenciación a Largo Plazo/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/fisiología , Receptores de GABA-B/metabolismo
10.
Neuropsychopharmacology ; 38(7): 1234-44, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23325324

RESUMEN

Increasing evidence suggests that high-frequency deep brain stimulation of the nucleus accumbens (NAcb-DBS) may represent a novel therapeutic strategy for individuals suffering from treatment-resistant depression, although the underlying mechanisms of action remain largely unknown. In this study, using a unique mouse model of enhanced depression- and anxiety-like behavior (HAB), we investigated behavioral and neurobiological effects of NAcb-DBS. HAB mice either underwent chronic treatment with one of three different selective serotonin reuptake inhibitors (SSRIs) or received NAcb-DBS for 1 h per day for 7 consecutive days. Animals were tested in established paradigms revealing depression- and anxiety-related behaviors. The enhanced depression-like behavior of HAB mice was not influenced by chronic SSRI treatment. In contrast, repeated, but not single, NAcb-DBS induced robust antidepressant and anxiolytic responses in HAB animals, while these behaviors remained unaffected in normal depression/anxiety animals (NAB), suggesting a preferential effect of NAcb-DBS on pathophysiologically deranged systems. NAcb-DBS caused a modulation of challenge-induced activity in various stress- and depression-related brain regions, including an increase in c-Fos expression in the dentate gyrus of the hippocampus and enhanced hippocampal neurogenesis in HABs. Taken together, these findings show that the normalization of the pathophysiologically enhanced, SSRI-insensitive depression-like behavior by repeated NAcb-DBS was associated with the reversal of reported aberrant brain activity and impaired adult neurogenesis in HAB mice, indicating that NAcb-DBS affects neuronal activity as well as plasticity in a defined, mood-associated network. Thus, HAB mice may represent a clinically relevant model for elucidating the neurobiological correlates of NAcb-DBS.


Asunto(s)
Ansiedad/terapia , Conducta Animal/fisiología , Estimulación Encefálica Profunda , Depresión/terapia , Núcleo Accumbens/fisiología , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Giro Dentado/fisiología , Depresión/tratamiento farmacológico , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos , Neurogénesis/fisiología
11.
Exp Neurol ; 247: 531-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23399889

RESUMEN

Multiple system atrophy (MSA) is a fatal, rapidly progressive neurodegenerative disease with limited symptomatic treatment options. Discrimination of MSA from other degenerative disorders crucially depends on the presence of early and severe cardiovascular autonomic failure (CAF). We have previously shown that neuropathologic lesions in the central autonomic nuclei similar to the human disease are present in transgenic MSA mice generated by targeted oligodendroglial overexpression of α-syn using the PLP promoter. We here explore whether such lesions result in abnormalities of heart rate variability (HRV) and circadian rhythmicity which are typically impaired in MSA patients. HRV analysis was performed in five month old transgenic PLP-α-syn (tg) MSA mice and age-matched wild type controls. Decreased HRV and alterations in the circadian rhythmicity were detected in the tg MSA group. The number of choline-acetyltransferase-immunoreactive neurons in the nucleus ambiguus was significantly decreased in the tg group, whereas the levels of arginine-vasopressin neurons in the suprachiasmatic and paraventricular nucleus were not affected. Our finding of impaired HRV and circadian rhythmicity in tg MSA mice associated with degeneration of the nucleus ambiguus suggests that a cardinal non-motor feature of human MSA can be reproduced in the mouse model strengthening its role as a valuable testbed for studying selective vulnerability and assessing translational therapies.


Asunto(s)
Anomalías Cardiovasculares/etiología , Atrofia de Múltiples Sistemas/complicaciones , Oligodendroglía/patología , alfa-Sinucleína/metabolismo , Análisis de Varianza , Animales , Arginina Vasopresina/metabolismo , Temperatura Corporal/genética , Anomalías Cardiovasculares/genética , Colina O-Acetiltransferasa/metabolismo , Modelos Animales de Enfermedad , Electrocardiografía , Electroencefalografía , Frecuencia Cardíaca/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/genética , Atrofia de Múltiples Sistemas/genética , Proteína Proteolipídica de la Mielina/genética , Núcleo Accumbens/metabolismo , Núcleo Accumbens/patología , Oligodendroglía/metabolismo , Factores de Tiempo , alfa-Sinucleína/genética
12.
Neuropsychopharmacology ; 37(6): 1534-47, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22334122

RESUMEN

Mood and anxiety disorders develop in some but not all individuals following exposure to stress and psychological trauma. However, the factors underlying individual differences in risk and resilience for these disorders, including genetic variation, remain to be determined. Isogenic inbred mouse strains provide a valuable approach to elucidating these factors. Here, we performed a comprehensive examination of the extinction-impaired 129S1/SvImJ (S1) inbred mouse strain for multiple behavioral, autonomic, neuroendocrine, and corticolimbic neuronal morphology phenotypes. We found that S1 exhibited fear overgeneralization to ambiguous contexts and cues, impaired context extinction and impaired safety learning, relative to the (good-extinguishing) C57BL/6J (B6) strain. Fear overgeneralization and impaired extinction was rescued by treatment with the front-line anxiety medication fluoxetine. Telemetric measurement of electrocardiogram signals demonstrated autonomic disturbances in S1 including poor recovery of fear-induced suppression of heart rate variability. S1 with a history of chronic restraint stress displayed an attenuated corticosterone (CORT) response to a novel, swim stressor. Conversely, previously stress-naive S1 showed exaggerated CORT responses to acute restraint stress or extinction training, insensitivity to dexamethasone challenge, and reduced hippocampal CA3 glucocorticoid receptor mRNA, suggesting downregulation of negative feedback control of the hypothalamic-pituitary-adrenal axis. Analysis of neuronal morphology in key neural nodes within the fear and extinction circuit revealed enlarged dendritic arbors in basolateral amygdala neurons in S1, but normal infralimbic cortex and prelimbic cortex dendritic arborization. Collectively, these data provide convergent support for the utility of the S1 strain as a tractable model for elucidating the neural, molecular and genetic basis of persistent, excessive fear.


Asunto(s)
Amígdala del Cerebelo/patología , Trastornos de Ansiedad/complicaciones , Trastornos de Ansiedad/patología , Enfermedades del Sistema Nervioso Autónomo/etiología , Dendritas/patología , Enfermedades del Sistema Endocrino/etiología , Extinción Psicológica/fisiología , Miedo/fisiología , Inhibición Psicológica , Análisis de Varianza , Animales , Antidepresivos de Segunda Generación/uso terapéutico , Trastornos de Ansiedad/tratamiento farmacológico , Reacción de Prevención/efectos de los fármacos , Corticosterona/sangre , Discriminación en Psicología , Modelos Animales de Enfermedad , Electrocardiografía , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Fluoxetina/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos , ARN Mensajero/metabolismo , Receptores de Glucocorticoides , Telemetría
13.
PLoS One ; 6(2): e16849, 2011 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-21386891

RESUMEN

The propensity to develop an anxiety disorder is thought to be determined by genetic and environmental factors. Here we investigated the relationship between a genetic predisposition to trait anxiety and experience-based learned fear in a psychopathological mouse model. Male CD-1 mice selectively bred for either high (HAB), or normal (NAB) anxiety-related behaviour on the elevated plus maze were subjected to classical fear conditioning. During conditioning both mouse lines showed increased fear responses as assessed by freezing behaviour. However, 24 h later, HAB mice displayed more pronounced conditioned responses to both a contextual or cued stimulus when compared with NAB mice. Interestingly, 6 h and already 1 h after fear conditioning, freezing levels were high in HAB mice but not in NAB mice. These results suggest that trait anxiety determines stronger fear memory and/or a weaker ability to inhibit fear responses in the HAB line. The enhanced fear response of HAB mice was attenuated by treatment with either the α(2,3,5)-subunit selective benzodiazepine partial agonist L-838,417, corticosterone or the selective neurokinin-1 receptor antagonist L-822,429. Overall, the HAB mouse line may represent an interesting model (i) for identifying biological factors underlying misguided conditioned fear responses and (ii) for studying novel anxiolytic pharmacotherapies for patients with fear-associated disorders, including post-traumatic stress disorder and phobias.


Asunto(s)
Ansiolíticos/uso terapéutico , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/patología , Modelos Animales de Enfermedad , Miedo/fisiología , Ratones , Animales , Trastornos de Ansiedad/complicaciones , Trastornos de Ansiedad/psicología , Conducta Animal , Corticosterona/uso terapéutico , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Emoción Expresada/fisiología , Fluorobencenos/uso terapéutico , Masculino , Antagonistas del Receptor de Neuroquinina-1 , Trastornos Fóbicos/complicaciones , Trastornos Fóbicos/tratamiento farmacológico , Trastornos Fóbicos/patología , Condicionamiento Físico Animal/fisiología , Condicionamiento Físico Animal/psicología , Piperidinas/uso terapéutico , Psicopatología , Triazoles/uso terapéutico , Regulación hacia Arriba
14.
Neuropsychopharmacology ; 36(3): 652-63, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20980994

RESUMEN

The cannabinoid receptor type 1 (CB1) and the central nucleus of the amygdala (CeA) are both known to have crucial roles in the processing of fear and anxiety, whereby they appear to be especially involved in the control of fear states. However, in contrast to many other brain regions including the cortical subregions of the amygdala, the existence of CB1 in the CeA remains enigmatic. In this study we show that CB1 is expressed in the CeA of mice and that CB1 in the CeA mediates short-term synaptic plasticity, namely depolarization-induced suppression of excitation (DSE) and inhibition (DSI). Moreover, the CB1 antagonist AM251 increased both excitatory and inhibitory postsynaptic responses in CeA neurons. Local application of AM251 in the CeA in vivo resulted in an acutely increased fear response in an auditory fear conditioning paradigm. Upon application of AM251 in the basolateral nucleus of the amygdala (BLA) in an otherwise identical protocol, no such acute behavioral effects were detected, but CB1 blockade resulted in increased fear responses during tone exposures on the subsequent days. Moreover, we observed that the efficacy of DSE and DSI in the CeA was increased on the day following fear conditioning, indicating that a single tone-shock pairing resulted in changes in endocannabinoid signaling in the CeA. Taken together, our data show the existence of CB1 proteins in the CeA, and their critical role for ensuring short-term adaptation of responses to fearful events, thereby suggesting a potential therapeutic target to accompany habituation-based therapies of post-traumatic symptoms.


Asunto(s)
Adaptación Psicológica/fisiología , Amígdala del Cerebelo/fisiología , Moduladores de Receptores de Cannabinoides/metabolismo , Condicionamiento Psicológico/fisiología , Endocannabinoides , Miedo/fisiología , Transducción de Señal/fisiología , Potenciales de Acción/efectos de los fármacos , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Conducta Animal , Moduladores de Receptores de Cannabinoides/antagonistas & inhibidores , Condicionamiento Psicológico/efectos de los fármacos , Estimulación Eléctrica/métodos , Antagonistas de Aminoácidos Excitadores/farmacología , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Miedo/efectos de los fármacos , Antagonistas del GABA/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Técnicas In Vitro , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácidos Fosfínicos/farmacología , Piperidinas/farmacología , Propanolaminas/farmacología , Pirazoles/farmacología , Piridazinas/farmacología , Quinoxalinas/farmacología , Receptor Cannabinoide CB1/deficiencia , Células Receptoras Sensoriales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Valina/análogos & derivados , Valina/farmacología
15.
Neuropsychopharmacology ; 34(3): 775-85, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18800067

RESUMEN

Stress and anxiety are mainly regulated by amygdala and hypothalamic circuitries involving several neurotransmitter systems and providing physiological responses to peripheral organs via the hypothalamic-pituitary-adrenal axis and other pathways. The role of endogenous opioid peptides in this process is largely unknown. Here we show for the first time that anxiolytic parameters of explorative behavior in mice lacking prodynorphin were increased 2-4-fold in the open field, the elevated plus maze and the light-dark test. Consistent with this, treatment of wild-type mice with selective kappa-opioid receptor antagonists GNTI or norbinaltorphimine showed the same effects. Furthermore, treatment of prodynorphin knockout animals with U-50488H, a selective kappa-opioid receptor agonist, fully reversed their anxiolytic phenotype. These behavioral data are supported by an approximal 30% reduction in corticotropin-releasing hormone (CRH) mRNA expression in the hypothalamic paraventricular nucleus and central amygdala and an accompanying 30-40% decrease in corticosterone serum levels in prodynorphin knockout mice. Although stress-induced increases in corticosterone levels were attenuated in prodynorphin knockout mice, they were associated with minor increases in depression-like behavior in the tail suspension and forced swim tests. Taken together, our data suggest a pronounced impact of endogenous prodynorphin-derived peptides on anxiety, but not stress coping ability and that these effects are mediated via kappa-opioid receptors. The delay in the behavioral response to kappa-opioid receptor agonists and antagonist treatment suggests an indirect control level for the action of dynorphin, probably by modulating the expression of CRH or neuropeptide Y, and subsequently influencing behavior.


Asunto(s)
Ansiedad/metabolismo , Corticosterona , Encefalinas/fisiología , Neuropéptidos/metabolismo , Precursores de Proteínas/fisiología , Receptores Opioides kappa , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/fisiopatología , Tronco Encefálico/metabolismo , Corticosterona/sangre , Hormona Liberadora de Corticotropina/metabolismo , Dinorfinas/fisiología , Encefalinas/genética , Encefalinas/metabolismo , Conducta Exploratoria , Femenino , Guanidinas , Hipotálamo/metabolismo , Masculino , Aprendizaje por Laberinto , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/metabolismo , Morfinanos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Neuropéptido Y/fisiología , Neuropéptidos/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Núcleos del Rafe/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Estrés Psicológico/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA