Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell ; 166(3): 716-728, 2016 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-27426949

RESUMEN

Fear behaviors are regulated by adaptive mechanisms that dampen their expression in the absence of danger. By studying circuits and the molecular mechanisms underlying this adaptive response, we show that cholinergic neurons of the medial habenula reduce fear memory expression through GABAB presynaptic excitation. Ablating these neurons or inactivating their GABAB receptors impairs fear extinction in mice, whereas activating the neurons or their axonal GABAB receptors reduces conditioned fear. Although considered exclusively inhibitory, here, GABAB mediates excitation by amplifying presynaptic Ca(2+) entry through Cav2.3 channels and potentiating co-release of glutamate, acetylcholine, and neurokinin B to excite interpeduncular neurons. Activating the receptors for these neurotransmitters or enhancing neurotransmission with a phosphodiesterase inhibitor reduces fear responses of both wild-type and GABAB mutant mice. We identify the role of an extra-amygdalar circuit and presynaptic GABAB receptors in fear control, suggesting that boosting neurotransmission in this pathway might ameliorate some fear disorders.


Asunto(s)
Neuronas Colinérgicas/metabolismo , Miedo/fisiología , Habénula/fisiología , Memoria/fisiología , Receptores de GABA-B/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas , Neurotransmisores/metabolismo , Transmisión Sináptica
2.
Nature ; 558(7708): 127-131, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29849148

RESUMEN

The ability of the taste system to identify a tastant (what it tastes like) enables animals to recognize and discriminate between the different basic taste qualities1,2. The valence of a tastant (whether it is appetitive or aversive) specifies its hedonic value and elicits the execution of selective behaviours. Here we examine how sweet and bitter are afforded valence versus identity in mice. We show that neurons in the sweet-responsive and bitter-responsive cortex project to topographically distinct areas of the amygdala, with strong segregation of neural projections conveying appetitive versus aversive taste signals. By manipulating selective taste inputs to the amygdala, we show that it is possible to impose positive or negative valence on a neutral water stimulus, and even to reverse the hedonic value of a sweet or bitter tastant. Remarkably, mice with silenced neurons in the amygdala no longer exhibit behaviour that reflects the valence associated with direct stimulation of the taste cortex, or with delivery of sweet and bitter chemicals. Nonetheless, these mice can still identify and discriminate between tastants, just as wild-type controls do. These results help to explain how the taste system generates stereotypic and predetermined attractive and aversive taste behaviours, and support the existence of distinct neural substrates for the discrimination of taste identity and the assignment of valence.


Asunto(s)
Amígdala del Cerebelo/citología , Amígdala del Cerebelo/fisiología , Conducta Apetitiva/fisiología , Reacción de Prevención/fisiología , Discriminación en Psicología/fisiología , Gusto/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Conducta Apetitiva/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Clozapina/análogos & derivados , Clozapina/farmacología , Discriminación en Psicología/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Gusto/efectos de los fármacos , Agua/farmacología
3.
Cereb Cortex ; 27(6): 3110-3124, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27252353

RESUMEN

Local inhibition by γ-amino butyric acid (GABA)-containing neurons is of vital importance for the operation of sensory cortices. However, the physiological response patterns of cortical GABAergic neurons are poorly understood, especially in the awake condition. Here, we utilized the recently developed optical tagging technique to specifically record GABAergic neurons in the anterior piriform cortex (aPC) in awake mice. The identified aPC GABAergic neurons were stimulated with robotic delivery of 32 distinct odorants, which covered a broad range of functional groups. We found that aPC GABAergic neurons could be divided into 4 types based on their response patterns. Type I, type II, and type III neurons displayed broad excitatory responses to test odorants with different dynamics. Type I neurons were constantly activated during odorant stimulation, whereas type II neurons were only transiently activated at the onset of odorant delivery. In addition, type III neurons displayed transient excitatory responses both at the onset and termination of odorant presentation. Interestingly, type IV neurons were broadly inhibited by most of the odorants. Taken together, aPC GABAergic neurons adopt different strategies to affect the cortical circuitry. Our results will allow for better understanding of the role of cortical GABAergic interneurons in sensory information processing.


Asunto(s)
Neuronas GABAérgicas/fisiología , Percepción Olfatoria/fisiología , Corteza Piriforme/citología , Vigilia/fisiología , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Animales , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Glutamato Descarboxilasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Luz , Ratones , Ratones Transgénicos , Inhibición Neural/fisiología , Odorantes , Optogenética , Análisis de Componente Principal , Olfato/fisiología , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(43): 17681-6, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23045693

RESUMEN

The heart peptide hormone atrial natriuretic peptide (ANP) regulates blood pressure by stimulating guanylyl cyclase-A to produce cyclic guanosine monophosphate (cGMP). ANP and guanylyl cyclase-A are also expressed in many brain areas, but their physiological functions and downstream signaling pathways remain enigmatic. Here we investigated the physiological functions of ANP signaling in the neural pathway from the medial habenula (MHb) to the interpeduncular nucleus (IPN). Biochemical assays indicate that ANP increases cGMP accumulation in the IPN of mouse brain slices. Using optogenetic stimulation and electrophysiological recordings, we show that both ANP and brain natriuretic peptide profoundly block glutamate release from MHb neurons. Pharmacological applications reveal that this blockade is mediated by phosphodiesterase 2A (PDE2A) but not by cGMP-stimulated protein kinase-G or cGMP-sensitive cyclic nucleotide-gated channels. In addition, focal infusion of ANP into the IPN enhances stress-induced analgesia, and the enhancement is prevented by PDE2A inhibitors. PDE2A is richly expressed in the axonal terminals of MHb neurons, and its activation by cGMP depletes cyclic adenosine monophosphates. The inhibitory effect of ANP on glutamate release is reversed by selectively activating protein kinase A. These results demonstrate strong presynaptic inhibition by natriuretic peptides in the brain and suggest important physiological and behavioral roles of PDE2A in modulating neurotransmitter release by negative crosstalk between cGMP-signaling and cyclic adenosine monophosphate-signaling pathways.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Péptidos Natriuréticos/farmacología , Terminales Presinápticos/enzimología , Transmisión Sináptica/efectos de los fármacos , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
5.
J Neurosci ; 33(8): 3624-32, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23426689

RESUMEN

POMC-derived melanocortins inhibit food intake. In the adult rodent brain, POMC-expressing neurons are located in the arcuate nucleus (ARC) and the nucleus tractus solitarius (NTS), but it remains unclear how POMC neurons in these two brain nuclei regulate feeding behavior and metabolism differentially. Using pharmacogenetic methods to activate or deplete neuron groups in separate brain areas, in the present study, we show that POMC neurons in the ARC and NTS suppress feeding behavior at different time scales. Neurons were activated using the DREADD (designer receptors exclusively activated by designer drugs) method. The evolved human M3-muscarinic receptor was expressed in a selective population of POMC neurons by stereotaxic infusion of Cre-recombinase-dependent, adeno-associated virus vectors into the ARC or NTS of POMC-Cre mice. After injection of the human M3-muscarinic receptor ligand clozapine-N-oxide (1 mg/kg, i.p.), acute activation of NTS POMC neurons produced an immediate inhibition of feeding behavior. In contrast, chronic stimulation was required for ARC POMC neurons to suppress food intake. Using adeno-associated virus delivery of the diphtheria toxin receptor gene, we found that diphtheria toxin-induced ablation of POMC neurons in the ARC but not the NTS, increased food intake, reduced energy expenditure, and ultimately resulted in obesity and metabolic and endocrine disorders. Our results reveal different behavioral functions of POMC neurons in the ARC and NTS, suggesting that POMC neurons regulate feeding and energy homeostasis by integrating long-term adiposity signals from the hypothalamus and short-term satiety signals from the brainstem.


Asunto(s)
Tronco Encefálico/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Inhibición Neural/fisiología , Neuronas/fisiología , Proopiomelanocortina/fisiología , Adiposidad/genética , Animales , Tronco Encefálico/virología , Dependovirus/genética , Femenino , Vectores Genéticos/administración & dosificación , Células HEK293 , Homeostasis/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Inhibición Neural/genética , Vías Nerviosas/fisiopatología , Neuronas/virología , Proopiomelanocortina/antagonistas & inhibidores
6.
Sci Bull (Beijing) ; 65(5): 389-401, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36659230

RESUMEN

Organisms must make sense of a constant stream of sensory inputs from both internal and external sources which compete for attention by determining which ones are salient. The ability to detect and respond appropriately to potentially salient stimuli in the environment is critical to all organisms. However, the neural circuits that process salience are not fully understood. Here, we identify a population of glutamatergic neurons in the ventral pallidum (VP) that play a unique role in salience processing. Using cell-type-specific fiber photometry, we find that VP glutamatergic neurons are robustly activated by a variety of aversion- and reward-related stimuli, as well as novel social and non-social stimuli. Inhibition of the VP glutamatergic neurons reduces the ability to detect salient stimuli in the environment, such as aversive cue, novel conspecific and novel object. Besides, VP glutamatergic neurons project to both the lateral habenula (LHb) and the ventral tegmental area (VTA). Together, our findings demonstrate that the VP glutamatergic neurons participate in salience processing and therefore provide a new perspective on treating several neuropsychiatric disorders, including dementia and psychosis.

7.
Neuron ; 97(4): 911-924.e5, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29398361

RESUMEN

The interactions between predator and prey represent some of the most dramatic events in nature and constitute a matter of life and death for both sides. The hypothalamus has been implicated in driving predation and evasion; however, the exact hypothalamic neural circuits underlying these behaviors remain poorly defined. Here, we demonstrate that inhibitory and excitatory projections from the mouse lateral hypothalamus (LH) to the periaqueductal gray (PAG) in the midbrain drive, respectively, predation and evasion. LH GABA neurons were activated during predation. Optogenetically stimulating PAG-projecting LH GABA neurons drove strong predatory attack, and inhibiting these cells reversibly blocked predation. In contrast, LH glutamate neurons were activated during evasion. Stimulating PAG-projecting LH glutamate neurons drove evasion and inhibiting them impeded predictive evasion. Therefore, the seemingly opposite behaviors of predation and evasion are tightly regulated by two dissociable modular command systems within a single neural projection from the LH to the PAG. VIDEO ABSTRACT.


Asunto(s)
Neuronas GABAérgicas/fisiología , Área Hipotalámica Lateral/fisiología , Sustancia Gris Periacueductal/fisiología , Conducta Predatoria/fisiología , Animales , Conducta Animal , Femenino , Masculino , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología
8.
Neuron ; 95(1): 138-152.e5, 2017 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-28625488

RESUMEN

Stress-induced hyperglycemia is a fundamental adaptive response that mobilizes energy stores in response to threats. Here, our examination of the contributions of the central catecholaminergic (CA) neuronal system to this adaptive response revealed that CA neurons in the ventrolateral medulla (VLM) control stress-induced hyperglycemia. Ablation of VLM CA neurons abolished the hyperglycemic response to both physical and psychological stress, whereas chemogenetic activation of these neurons was sufficient to induce hyperglycemia. We further found that CA neurons in the rostral VLM, but not those in the caudal VLM, cause hyperglycemia via descending projections to the spinal cord. Monosynaptic tracing experiments showed that VLM CA neurons receive direct inputs from multiple stress-responsive brain areas. Optogenetic studies identified an excitatory PVN-VLM circuit that induces hyperglycemia. This study establishes the central role of VLM CA neurons in stress-induced hyperglycemia and substantially expands our understanding of the central mechanism that controls glucose metabolism.


Asunto(s)
Glucemia/metabolismo , Catecolaminas/metabolismo , Hiperglucemia/metabolismo , Bulbo Raquídeo/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Estrés Fisiológico/fisiología , Estrés Psicológico/metabolismo , Animales , Lipopolisacáridos , Bulbo Raquídeo/fisiología , Ratones , Vías Nerviosas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo
9.
PLoS One ; 11(9): e0163500, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27684481

RESUMEN

Sleep is a ubiquitous, tightly regulated, and evolutionarily conserved behavior observed in almost all animals. Prolonged sleep deprivation can be fatal, indicating that sleep is a physiological necessity. However, little is known about its core function. To gain insight into this mystery, we used advanced quantitative proteomics technology to survey the global changes in brain protein abundance. Aiming to gain a comprehensive profile, our proteomics workflow included filter-aided sample preparation (FASP), which increased the coverage of membrane proteins; tandem mass tag (TMT) labeling, for relative quantitation; and high resolution, high mass accuracy, high throughput mass spectrometry (MS). In total, we obtained the relative abundance ratios of 9888 proteins encoded by 6070 genes. Interestingly, we observed significant enrichment for mitochondrial proteins among the differentially expressed proteins. This finding suggests that sleep deprivation strongly affects signaling pathways that govern either energy metabolism or responses to mitochondrial stress. Additionally, the differentially-expressed proteins are enriched in pathways implicated in age-dependent neurodegenerative diseases, including Parkinson's, Huntington's, and Alzheimer's, hinting at possible connections between sleep loss, mitochondrial stress, and neurodegeneration.

10.
Neuron ; 81(6): 1360-1374, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24656254

RESUMEN

The dorsal raphe nucleus (DRN) in the midbrain is a key center for serotonin (5-hydroxytryptamine; 5-HT)-expressing neurons. Serotonergic neurons in the DRN have been theorized to encode punishment by opposing the reward signaling of dopamine neurons. Here, we show that DRN neurons encode reward, but not punishment, through 5-HT and glutamate. Optogenetic stimulation of DRN Pet-1 neurons reinforces mice to explore the stimulation-coupled spatial region, shifts sucrose preference, drives optical self-stimulation, and directs sensory discrimination learning. DRN Pet-1 neurons increase their firing activity during reward tasks, and this activation can be used to rapidly change neuronal activity patterns in the cortex. Although DRN Pet-1 neurons are often associated with 5-HT, they also release glutamate, and both neurotransmitters contribute to reward signaling. These experiments demonstrate the ability of DRN neurons to organize reward behaviors and might provide insights into the underlying mechanisms of learning facilitation and anhedonia treatment.


Asunto(s)
Conducta Animal/fisiología , Ácido Glutámico/metabolismo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Recompensa , Serotonina/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Núcleos del Rafe/metabolismo
11.
Neurosci Bull ; 29(5): 517-24, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23516143

RESUMEN

Dopaminergic neurons regulate and organize numerous important behavioral processes including motor activity. Consistently, manipulation of brain dopamine concentrations changes animal activity levels. Dopamine is synthesized by several neuronal populations in the brain. This study was carried out to directly test whether selective activation of dopamine neurons in the midbrain induces hyperactivity. A pharmacogenetic approach was used to activate midbrain dopamine neurons, and behavioral assays were conducted to determine the effects on mouse activity levels. Transgenic expression of the evolved hM3Dq receptor was achieved by infusing Cre-inducible AAV viral vectors into the midbrain of DAT-Cre mice. Neurons were excited by injecting the hM3Dq ligand clozapine-N-oxide (CNO). Mouse locomotor activity was measured in an open field. The results showed that CNO selectively activated midbrain dopaminergic neurons and induced hyperactivity in a dose-dependent manner, supporting the idea that these neurons play an important role in regulating motor activity.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Hipercinesia/metabolismo , Mesencéfalo/fisiología , Actividad Motora/fisiología , Animales , Clozapina/análogos & derivados , Clozapina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Hipercinesia/fisiopatología , Inmunohistoquímica , Mesencéfalo/efectos de los fármacos , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda