Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 362
Filtrar
1.
Nature ; 583(7814): 109-114, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32528181

RESUMO

Hibernating mammals actively lower their body temperature to reduce energy expenditure when facing food scarcity1. This ability to induce a hypometabolic state has evoked great interest owing to its potential medical benefits2,3. Here we show that a hypothalamic neuronal circuit in rodents induces a long-lasting hypothermic and hypometabolic state similar to hibernation. In this state, although body temperature and levels of oxygen consumption are kept very low, the ability to regulate metabolism still remains functional, as in hibernation4. There was no obvious damage to tissues and organs or abnormalities in behaviour after recovery from this state. Our findings could enable the development of a method to induce a hibernation-like state, which would have potential applications in non-hibernating mammalian species including humans.


Assuntos
Metabolismo Energético/fisiologia , Hibernação/fisiologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Vias Neurais/citologia , Vias Neurais/fisiologia , Animais , Metabolismo Basal/fisiologia , Núcleo Hipotalâmico Dorsomedial/citologia , Núcleo Hipotalâmico Dorsomedial/fisiologia , Feminino , Neurônios GABAérgicos/metabolismo , Glutamina/metabolismo , Masculino , Camundongos , Consumo de Oxigênio/fisiologia
2.
Genes Cells ; 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38964745

RESUMO

An autism-associated gene Shank3 encodes multiple splicing isoforms, Shank3a-f. We have recently reported that Shank3a/b-knockout mice were more susceptible to kainic acid-induced seizures than wild-type mice at 4 weeks of age. Little is known, however, about how the N-terminal and ankyrin repeat domains (NT-Ank) of Shank3a/b regulate multiple molecular signals in the developing brain. To explore the functional roles of Shank3a/b, we performed a mass spectrometry-based proteomic search for proteins interacting with GFP-tagged NT-Ank. In this study, NT-Ank was predicted to form a variety of complexes with a total of 348 proteins, in which RNA-binding (n = 102), spliceosome (n = 22), and ribosome-associated molecules (n = 9) were significantly enriched. Among them, an X-linked intellectual disability-associated protein, Nono, was identified as a NT-Ank-binding protein. Coimmunoprecipitation assays validated the interaction of Shank3 with Nono in the mouse brain. In agreement with these data, the thalamus of Shank3a/b-knockout mice aberrantly expressed splicing isoforms of autism-associated genes, Nrxn1 and Eif4G1, before and after seizures with kainic acid treatment. These data indicate that Shank3 interacts with multiple RNA-binding proteins in the postnatal brain, thereby regulating the homeostatic expression of splicing isoforms for autism-associated genes after birth.

3.
J Neurosci ; 43(22): 4075-4092, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37117013

RESUMO

To understand how sleep-wakefulness cycles are regulated, it is essential to disentangle structural and functional relationships between the preoptic area (POA) and lateral hypothalamic area (LHA), since these regions play important yet opposing roles in the sleep-wakefulness regulation. GABA- and galanin (GAL)-producing neurons in the ventrolateral preoptic nucleus (VLPO) of the POA (VLPOGABA and VLPOGAL neurons) are responsible for the maintenance of sleep, while the LHA contains orexin-producing neurons (orexin neurons) that are crucial for maintenance of wakefulness. Through the use of rabies virus-mediated neural tracing combined with in situ hybridization (ISH) in male and female orexin-iCre mice, we revealed that the vesicular GABA transporter (Vgat, Slc32a1)- and galanin (Gal)-expressing neurons in the VLPO directly synapse with orexin neurons in the LHA. A majority (56.3 ± 8.1%) of all VLPO input neurons connecting to orexin neurons were double-positive for Vgat and Gal Using projection-specific rabies virus-mediated tracing in male and female Vgat-ires-Cre and Gal-Cre mice, we discovered that VLPOGABA and VLPOGAL neurons that send projections to the LHA received innervations from similarly distributed input neurons in many brain regions, with the POA and LHA being among the main upstream areas. Additionally, we found that acute optogenetic excitation of axons of VLPOGABA neurons, but not VLPOGAL neurons, in the LHA of male Vgat-ires-Cre mice induced wakefulness. This study deciphers the connectivity between the VLPO and LHA, provides a large-scale map of upstream neuronal populations of VLPO→LHA neurons, and reveals a previously uncovered function of the VLPOGABA→LHA pathway in the regulation of sleep and wakefulness.SIGNIFICANCE STATEMENT We identified neurons in the ventrolateral preoptic nucleus (VLPO) that are positive for vesicular GABA transporter (Vgat) and/or galanin (Gal) and serve as presynaptic partners of orexin-producing neurons in the lateral hypothalamic area (LHA). We depicted monosynaptic input neurons of GABA- and galanin-producing neurons in the VLPO that send projections to the LHA throughout the entire brain. Their input neurons largely overlap, suggesting that they comprise a common neuronal population. However, acute excitatory optogenetic manipulation of the VLPOGABA→LHA pathway, but not the VLPOGAL→LHA pathway, evoked wakefulness. This study shows the connectivity of major components of the sleep/wake circuitry in the hypothalamus and unveils a previously unrecognized function of the VLPOGABA→LHA pathway in sleep-wakefulness regulation. Furthermore, we suggest the existence of subpopulations of VLPOGABA neurons that innervate LHA.


Assuntos
Região Hipotalâmica Lateral , Área Pré-Óptica , Camundongos , Masculino , Feminino , Animais , Área Pré-Óptica/fisiologia , Região Hipotalâmica Lateral/fisiologia , Orexinas/metabolismo , Galanina/metabolismo , Neurônios/fisiologia , Vigília/fisiologia , Sono/fisiologia , Ácido gama-Aminobutírico/metabolismo
4.
Semin Cell Dev Biol ; 129: 40-46, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-34649805

RESUMO

Social behavior involves many processes including cognitive functions. Altered social behaviors associated with many psychiatric disorders might have alterations in the processes. Poor social environment affects development and maturation of cognitive functions that are important for social cognition, possibly introducing social stress as well as vulnerability to the stress into the developing brain. Adolescence and early adulthood have higher sensitivity to social stress, which may be linked to the onset of psychiatric disorders during this time period. Understanding social behavioral processes in detail will be crucial for elucidating mechanisms of emerging the social behavior phenotypes in psychiatric disorders and for devising therapeutic and preventive interventions to introduce the resilience for the onset of psychiatric disorders through modulation of social circuitries.


Assuntos
Transtornos Mentais , Meio Social , Encéfalo , Cognição , Humanos , Comportamento Social
5.
J Neurosci ; 41(7): 1582-1596, 2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33372061

RESUMO

During rapid eye movement (REM) sleep, anti-gravity muscle tone and bodily movements are mostly absent, because somatic motoneurons are inhibited by descending inhibitory pathways. Recent studies showed that glycine/GABA neurons in the ventromedial medulla (VMM; GlyVMM neurons) play an important role in generating muscle atonia during REM sleep (REM-atonia). However, how these REM-atonia-inducing neurons interconnect with other neuronal populations has been unknown. In the present study, we first identified a specific subpopulation of GlyVMM neurons that play an important role in induction of REM-atonia by virus vector-mediated tracing in male mice in which glycinergic neurons expressed Cre recombinase. We found these neurons receive direct synaptic input from neurons in several brain stem regions, including glutamatergic neurons in the sublaterodorsal tegmental nucleus (SLD; GluSLD neurons). Silencing this circuit by specifically expressing tetanus toxin light chain (TeTNLC) resulted in REM sleep without atonia. This manipulation also caused a marked decrease in time spent in cataplexy-like episodes (CLEs) when applied to narcoleptic orexin-ataxin-3 mice. We also showed that GlyVMM neurons play an important role in maintenance of sleep. This present study identified a population of glycinergic neurons in the VMM that are commonly involved in REM-atonia and cataplexy.SIGNIFICANCE STATEMENT We identified a population of glycinergic neurons in the ventral medulla that plays an important role in inducing muscle atonia during rapid eye movement (REM) sleep. It sends axonal projections almost exclusively to motoneurons in the spinal cord and brain stem except to those that innervate extraocular muscles, while other glycinergic neurons in the same region also send projections to other regions including monoaminergic nuclei. Furthermore, these neurons receive direct inputs from several brainstem regions including glutamatergic neurons in the sublaterodorsal tegmental nucleus (SLD). Genetic silencing of this pathway resulted in REM sleep without atonia and a decrease of cataplexy when applied to narcoleptic mice. This work identified a neural population involved in generating muscle atonia during REM sleep and cataplexy.


Assuntos
Cataplexia/fisiopatologia , Glicina/fisiologia , Bulbo/fisiologia , Músculo Esquelético/fisiologia , Neurônios/fisiologia , Sono REM/fisiologia , Animais , Ataxina-3/genética , Axônios/fisiologia , Cataplexia/genética , Eletroencefalografia , Masculino , Bulbo/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Tono Muscular/fisiologia , Músculo Esquelético/fisiopatologia , Narcolepsia/genética , Narcolepsia/fisiopatologia , Orexinas/genética , Toxina Tetânica/farmacologia
6.
Mol Psychiatry ; 26(8): 3931-3942, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33173197

RESUMO

Major mental illnesses such as schizophrenia (SZ) and bipolar disorder (BP) frequently accompany metabolic conditions, but their relationship is still unclear, in particular at the mechanistic level. We implemented an approach of "from population to neuron", combining population-based epidemiological analysis with neurobiological experiments using cell and animal models based on a hypothesis built from the epidemiological study. We characterized high-quality population data, olfactory neuronal cells biopsied from patients with SZ or BP, and healthy subjects, as well as mice genetically modified for insulin signaling. We accessed the Danish Registry and observed (1) a higher incidence of diabetes in people with SZ or BP and (2) higher incidence of major mental illnesses in people with diabetes in the same large cohort. These epidemiological data suggest the existence of common pathophysiological mediators in both diabetes and major mental illnesses. We hypothesized that molecules associated with insulin resistance might be such common mediators, and then validated the hypothesis by using two independent sets of olfactory neuronal cells biopsied from patients and healthy controls. In the first set, we confirmed an enrichment of insulin signaling-associated molecules among the genes that were significantly different between SZ patients and controls in unbiased expression profiling data. In the second set, olfactory neuronal cells from SZ and BP patients who were not pre-diabetic or diabetic showed reduced IRS2 tyrosine phosphorylation upon insulin stimulation, indicative of insulin resistance. These cells also displayed an upregulation of IRS1 protein phosphorylation at serine-312 at baseline (without insulin stimulation), further supporting the concept of insulin resistance in olfactory neuronal cells from SZ patients. Finally, Irs2 knockout mice showed an aberrant response to amphetamine, which is also observed in some patients with major mental illnesses. The bi-directional relationships between major mental illnesses and diabetes suggest that there may be common pathophysiological mediators associated with insulin resistance underlying these mental and physical conditions.


Assuntos
Transtorno Bipolar , Resistência à Insulina , Esquizofrenia , Animais , Transtorno Bipolar/genética , Humanos , Insulina , Camundongos , Neurônios , Esquizofrenia/genética
7.
Hum Mol Genet ; 27(18): 3165-3176, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29893844

RESUMO

Autophagy plays an essential role in intracellular degradation and maintenance of cellular homeostasis in all cells, including neurons. Although a recent study reported a copy number variation of Ulk2, a gene essential for initiating autophagy, associated with a case of schizophrenia (SZ), it remains to be studied whether Ulk2 dysfunction could underlie the pathophysiology of the disease. Here we show that Ulk2 heterozygous (Ulk2+/-) mice have upregulated expression of sequestosome-1/p62, an autophagy-associated stress response protein, predominantly in pyramidal neurons of the prefrontal cortex (PFC), and exhibit behavioral deficits associated with the PFC functions, including attenuated sensorimotor gating and impaired cognition. Ulk2+/- neurons showed imbalanced excitatory-inhibitory neurotransmission, due in part to selective down-modulation of gamma-aminobutyric acid (GABA)A receptor surface expression in pyramidal neurons. Genetically reducing p62 gene dosage or suppressing p62 protein levels with an autophagy-inducing agent restored the GABAA receptor surface expression and rescued the behavioral deficits in Ulk2+/- mice. Moreover, expressing a short peptide that specifically interferes with the interaction of p62 and GABAA receptor-associated protein, a protein that regulates endocytic trafficking of GABAA receptors, also restored the GABAA receptor surface expression and rescued the behavioral deficits in Ulk2+/- mice. Thus, the current study reveals a novel mechanism linking deregulated autophagy to functional disturbances of the nervous system relevant to SZ, through regulation of GABAA receptor surface presentation in pyramidal neurons.


Assuntos
Autofagia/genética , Proteínas Serina-Treonina Quinases/genética , Esquizofrenia/genética , Proteína Sequestossoma-1/genética , Animais , Variações do Número de Cópias de DNA/genética , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Peptídeos/genética , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Transporte Proteico/genética , Células Piramidais/metabolismo , Células Piramidais/patologia , Receptores de GABA-A/genética , Esquizofrenia/fisiopatologia , Transmissão Sináptica/genética
8.
Biochem Biophys Res Commun ; 521(3): 620-624, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31679691

RESUMO

Bilirubin oxidase has a post-translationally formed covalent-bond between the imidazole ring of His398 coordinated to type I copper and the indole ring of Trp396 located in the outer-coordination sphere. We performed point mutations at Trp396 with Ala, Thr, Phe, and Tyr with the aim of elucidating the role of the imidazole-indole moiety found only in bilirubin oxidase. The result showed shifts in the redox potential of type I copper towards negative direction by > 100 mV and decreases in cathodic current in electrochemistry, whereas optical and magnetic properties of type I copper were not affected or sparingly affected. Along with the conspicuous changes in redox properties enzymatic activities of the Trp396 mutants were prominently decreased. Further, chemical modification of the Trp residues with N-bromosuccinimide and photo-induced formylations of bilirubin oxidase exerted more pronounced effects on both redox properties and enzymatic activities compared to the Trp396 mutants. All these results unequivocally indicate that the covalent-bond formed between Trp396 and His398 plays a crucial role to enhance enzymatic activities of bilirubin oxidase by shifting the redox potential of type I Cu towards positive direction and also by functioning as the effective pathway of electron transport.


Assuntos
Cobre/química , Hypocreales/enzimologia , Imidazóis/química , Indóis/química , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/química , Hypocreales/química , Hypocreales/genética , Modelos Moleculares , Oxirredução , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Mutação Puntual , Conformação Proteica , Processamento de Proteína Pós-Traducional
9.
Addict Biol ; 25(1): e12723, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30734456

RESUMO

In drug addiction, environmental stimuli previously associated with cocaine use readily elicit cocaine-associated memories, which persist long after abstinence and trigger cocaine craving and consumption. Although previous studies suggest that the medial prefrontal cortex (mPFC) is involved in the expression of cocaine-addictive behaviors, it remains unclear whether excitatory and inhibitory neurons in the mPFC are causally related to the formation and retrieval of cocaine-associated memories. To address this issue, we used the designer receptors exclusively activated by designer drugs (DREADD) technology combined with a cocaine-induced conditioned place preference (CPP) paradigm. We suppressed mPFC neuronal activity in a cell-type- and timing-dependent manner. C57BL/6J wild-type mice received bilateral intra-mPFC infusion of an adeno-associated virus (AAV) expressing inhibitory DREADD (hM4Di) under the control of CaMKII promotor to selectively suppress mPFC pyramidal neurons. GAD67-Cre mice received bilateral intra-mPFC infusion of a Cre-dependent AAV expressing hM4Di to specifically silence GABAergic neurons. Chemogenetic suppression of mPFC pyramidal neurons significantly attenuated both the acquisition and expression of cocaine CPP, while suppression of mPFC GABAergic neurons affected neither the acquisition nor expression of cocaine CPP. Moreover, chemogenetic inhibition of mPFC glutamatergic neurons did not affect the acquisition and expression of lithium chloride-induced conditioned place aversion. These results suggest that the activation of glutamatergic, but not GABAergic, neurons in the mPFC mediates both the formation and retrieval of cocaine-associated memories.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Cocaína/farmacologia , Neurônios GABAérgicos/efeitos dos fármacos , Memória/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiopatologia , Animais , Modelos Animais de Doenças , Inibidores da Captação de Dopamina/farmacologia , Eletrofisiologia , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL
10.
Proc Natl Acad Sci U S A ; 114(17): E3526-E3535, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28396432

RESUMO

Narcolepsy is a sleep disorder caused by the loss of orexin (hypocretin)-producing neurons and marked by excessive daytime sleepiness and a sudden weakening of muscle tone, or cataplexy, often triggered by strong emotions. In a mouse model for narcolepsy, we previously demonstrated that serotonin neurons of the dorsal raphe nucleus (DRN) mediate the suppression of cataplexy-like episodes (CLEs) by orexin neurons. Using an optogenetic tool, in this paper we show that the acute activation of DRN serotonin neuron terminals in the amygdala, but not in nuclei involved in regulating rapid eye-movement sleep and atonia, suppressed CLEs. Not only did stimulating serotonin nerve terminals reduce amygdala activity, but the chemogenetic inhibition of the amygdala using designer receptors exclusively activated by designer drugs also drastically decreased CLEs, whereas chemogenetic activation increased them. Moreover, the optogenetic inhibition of serotonin nerve terminals in the amygdala blocked the anticataplectic effects of orexin signaling in DRN serotonin neurons. Taken together, the results suggest that DRN serotonin neurons, as a downstream target of orexin neurons, inhibit cataplexy by reducing the activity of amygdala as a center for emotional processing.


Assuntos
Tonsila do Cerebelo , Catalepsia , Núcleo Dorsal da Rafe , Neurônios Serotoninérgicos/metabolismo , Transdução de Sinais , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Tonsila do Cerebelo/fisiopatologia , Animais , Catalepsia/genética , Catalepsia/metabolismo , Catalepsia/patologia , Catalepsia/fisiopatologia , Núcleo Dorsal da Rafe/metabolismo , Núcleo Dorsal da Rafe/patologia , Núcleo Dorsal da Rafe/fisiopatologia , Movimentos Oculares , Masculino , Camundongos , Camundongos Knockout , Neurônios Serotoninérgicos/patologia , Serotonina/metabolismo
11.
Proc Natl Acad Sci U S A ; 114(22): 5731-5736, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28507129

RESUMO

Narcolepsy-cataplexy is a debilitating disorder of sleep/wakefulness caused by a loss of orexin-producing neurons in the lateroposterior hypothalamus. Genetic or pharmacologic orexin replacement ameliorates symptoms in mouse models of narcolepsy-cataplexy. We have recently discovered a potent, nonpeptide OX2R-selective agonist, YNT-185. This study validates the pharmacological activity of this compound in OX2R-transfected cells and in OX2R-expressing neurons in brain slice preparations. Intraperitoneal, and intracerebroventricular, administration of YNT-185 suppressed cataplexy-like episodes in orexin knockout and orexin neuron-ablated mice, but not in orexin receptor-deficient mice. Peripherally administered YNT-185 also promotes wakefulness without affecting body temperature in wild-type mice. Further, there was no immediate rebound sleep after YNT-185 administration in active phase in wild-type and orexin-deficient mice. No desensitization was observed after repeated administration of YNT-185 with respect to the suppression of cataplexy-like episodes. These results provide a proof-of-concept for a mechanistic therapy of narcolepsy-cataplexy by OX2R agonists.


Assuntos
Compostos de Anilina/farmacologia , Benzamidas/farmacologia , Cataplexia/tratamento farmacológico , Narcolepsia/tratamento farmacológico , Receptores de Orexina/agonistas , Orexinas/metabolismo , Transtornos do Sono do Ritmo Circadiano/tratamento farmacológico , Promotores da Vigília/uso terapêutico , Vigília/efeitos dos fármacos , Compostos de Anilina/química , Animais , Benzamidas/química , Modelos Animais de Doenças , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Orexina/genética , Orexinas/genética , Técnicas de Patch-Clamp , Sono/efeitos dos fármacos
12.
J Neurosci ; 38(28): 6366-6378, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29915137

RESUMO

The hypothalamus plays an important role in the regulation of sleep/wakefulness states. While the ventrolateral preoptic nucleus (VLPO) plays a critical role in the initiation and maintenance of sleep, the lateral posterior part of the hypothalamus contains neuronal populations implicated in maintenance of arousal, including orexin-producing neurons (orexin neurons) in the lateral hypothalamic area (LHA) and histaminergic neurons in the tuberomammillary nucleus (TMN). During a search for neurons that make direct synaptic contact with histidine decarboxylase-positive (HDC+), histaminergic neurons (HDC neurons) in the TMN and orexin neurons in the LHA of male mice, we found that these arousal-related neurons are heavily innervated by GABAergic neurons in the preoptic area including the VLPO. We further characterized GABAergic neurons electrophysiologically in the VLPO (GABAVLPO neurons) that make direct synaptic contact with these hypothalamic arousal-related neurons. These neurons (GABAVLPO→HDC or GABAVLPO→orexin neurons) were both potently inhibited by noradrenaline and serotonin, showing typical electrophysiological characteristics of sleep-promoting neurons in the VLPO. This work provides direct evidence of monosynaptic connectivity between GABAVLPO neurons and hypothalamic arousal neurons and identifies the effects of monoamines on these neuronal pathways.SIGNIFICANCE STATEMENT Rabies-virus-mediated tracing of input neurons of two hypothalamic arousal-related neuron populations, histaminergic and orexinergic neurons, showed that they receive similar distributions of input neurons in a variety of brain areas, with rich innervation by GABAergic neurons in the preoptic area, including the ventrolateral preoptic area (VLPO), a region known to play an important role in the initiation and maintenance of sleep. Electrophysiological experiments found that GABAergic neurons in the VLPO (GABAVLPO neurons) that make direct input to orexin or histaminergic neurons are potently inhibited by noradrenaline and serotonin, suggesting that these monoamines disinhibit histamine and orexin neurons. This work demonstrated functional and structural interactions between GABAVLPO neurons and hypothalamic arousal-related neurons.


Assuntos
Nível de Alerta/fisiologia , Neurônios GABAérgicos/fisiologia , Região Hipotalâmica Lateral/fisiologia , Área Pré-Óptica/fisiologia , Sono/fisiologia , Animais , Neurônios GABAérgicos/citologia , Região Hipotalâmica Lateral/citologia , Masculino , Camundongos , Camundongos Transgênicos , Vias Neurais/citologia , Vias Neurais/metabolismo , Norepinefrina/metabolismo , Área Pré-Óptica/citologia , Serotonina/metabolismo
13.
J Neurosci ; 38(47): 10080-10092, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30282729

RESUMO

Sleep-wake behavior is controlled by a wide range of neuronal populations in the mammalian brain. Although the ventral midbrain/pons (VMP) area is suggested to participate in sleep-wake regulation, the neuronal mechanisms have remained unclear. Here, we found that nonspecific cell ablation or selective ablation of GABAergic neurons by expressing diphtheria toxin fragment A in the VMP in male mice induced a large increase in wakefulness that lasted at least 4 weeks. In contrast, selective ablation of dopaminergic neurons in the VMP had little effect on wakefulness. Chemogenetic inhibition of VMP GABAergic neurons also markedly increased wakefulness. The wake-promoting effect of the VMP GABAergic neuron ablation or inhibition was attenuated to varying degrees by the administration of dopamine D1 or D2/3 receptor antagonists and abolished by the administration of both antagonists together. In contrast, chemogenetic activation of VMP GABAergic neurons very strongly increased slow-wave sleep and reduced wakefulness. These findings suggest that VMP GABAergic neurons regulate dopaminergic actions in the sleep-wake behavior of mice.SIGNIFICANCE STATEMENT Current understanding of the neuronal mechanisms and populations that regulate sleep-wake behavior is incomplete. Here, we identified a GABAergic ventral midbrain/pons area that is necessary for controlling the daily amount of sleep and wakefulness in mice. We also found that these inhibitory neurons control wakefulness by suppressing dopaminergic systems. Surprisingly, activation of these neurons strongly induced slow-wave sleep while suppressing wakefulness. Our study reveals a new brain mechanism critical for sleep-wake regulation.


Assuntos
Neurônios GABAérgicos/fisiologia , Mesencéfalo/fisiologia , Ponte/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Antagonistas de Dopamina/farmacologia , Eletroencefalografia/métodos , Neurônios GABAérgicos/efeitos dos fármacos , Masculino , Mesencéfalo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ponte/efeitos dos fármacos , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos
14.
Semin Cell Dev Biol ; 129: 1-2, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35654664

Assuntos
Cognição , Emoções
15.
Neurobiol Dis ; 131: 104322, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30423472

RESUMO

Cognitive functions including social cognition improve significantly during adolescence, the time period during which the brain typically handles a large volume of incoming information from the outside environment. Processing information and responding to environmental challenges allow the prefrontal cortex, a brain region important for cognition, to mature further and establish self-identity, social skills, and other cognitive abilities, thus helping individuals to function in society. People with vulnerable circuitries predisposed by either genetic or early environmental insults, may not be able to deal with social situations appropriately, and develop network dysfunction that may lead to the onset of schizophrenia, which often occurs during this period. Populations with higher risk for developing schizophrenia present "prodromal" phenotypes, including cognitive deficits, even before the onset of the disorder. Modulating circuit plasticity when the prefrontal cortex is particularly vulnerable allows us to support the development of cognitive functions in such populations and prevent them from transitioning into full-blown schizophrenia. For this approach to be successful, we need to conduct both human and animal studies side by side to better understand the neurobiology underlying the disorder, especially changes that occur over the disease trajectory that may be clinically relevant. By taking a multidisciplinary approach, there is a hope for precision medicine for schizophrenia in the future.


Assuntos
Cognição/fisiologia , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/fisiopatologia , Adolescente , Animais , Humanos , Sintomas Prodrômicos , Comportamento Social
16.
Biochem Biophys Res Commun ; 517(3): 520-524, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31376934

RESUMO

Simultaneous imaging and manipulation of a genetically defined neuronal population can provide a causal link between its activity and function. Here, we designed a miniaturized microscope (or 'miniscope') that allows fluorescence imaging and optogenetic manipulation at the cellular level in freely behaving animals. This miniscope has an integrated optical connector that accepts any combination of external light sources, allowing flexibility in the choice of sensors and manipulators. Moreover, due to its simple structure and use of open source software, the miniscope is easy to build and modify. Using this miniscope, we demonstrate the optogenetic silencing of hippocampal CA1 neurons using two laser light sources-one stimulating a calcium sensor (i.e., jGCaAMP7c) and the other serving as an optogenetic silencer (i.e., Jaws). This new miniscope can contribute to efforts to determine causal relationships between neuronal network dynamics and animal behavior.


Assuntos
Região CA1 Hipocampal/metabolismo , Microscopia/instrumentação , Rede Nervosa/metabolismo , Neuroimagem/métodos , Neurônios/metabolismo , Optogenética/métodos , Animais , Comportamento Animal/fisiologia , Região CA1 Hipocampal/ultraestrutura , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Injeções Intraventriculares , Luz , Camundongos , Microscopia/métodos , Rede Nervosa/ultraestrutura , Neuroimagem/instrumentação , Neurônios/ultraestrutura , Imagem Óptica/instrumentação , Imagem Óptica/métodos , Optogenética/instrumentação , Rodopsina/genética , Rodopsina/metabolismo
17.
Nat Rev Neurosci ; 15(11): 719-31, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25301357

RESUMO

Wakefulness and vigilance levels are required for maintaining purposeful activities and motivated behaviours, which are often triggered by sensory information conveying external cues. An increasing body of work has suggested that orexins (also known as hypocretins)--a pair of neuropeptides that are crucial for maintaining wakefulness - are also involved in the regulation of motivated behaviours, including feeding, emotional behaviour and reward seeking, and that these functions are mediated by two subtypes of orexin receptors. Autonomic and endocrine responses, which accompany these motivated behaviours, are also influenced by the orexin system. Orexin-producing neurons act as a hub that links information about the internal and external environments of an animal to vigilance levels and internal bodily functions to support various motivated behaviours.


Assuntos
Emoções/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Motivação/fisiologia , Neuropeptídeos/fisiologia , Vigília/fisiologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Comportamento Alimentar , Humanos , Vias Neurais/fisiologia , Neurônios/fisiologia , Orexinas , Recompensa
18.
Stem Cells ; 36(7): 969-976, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29484772

RESUMO

Neural stem and progenitor cells continue to generate new neurons in particular regions of the brain during adulthood. One of these neurogenic regions is the dentate gyrus (DG) of the hippocampus, which plays an important role in cognition and emotion. By exploiting this innate neuronal regeneration mechanism in the DG, new technologies have the potential to promote resistance to or recovery from brain dysfunction or degeneration. However, a deeper understanding of how adult DG neurogenesis is regulated by factors such as sleep and epigenetic modifications of gene expression could lead to further breakthroughs in the clinical application of neural stem and progenitor cells. In this review, we discuss the functions of adult-born DG neurons, describe the epigenetic regulation of adult DG neurogenesis, identify overlaps in how sleep and epigenetic modifications impact adult DG neurogenesis and memory consolidation, and suggest ways of using sleep or epigenetic interventions as therapies for neurodegenerative and psychiatric disorders. By knitting together separate strands of the literature, we hope to trigger new insights into how the functions of adult-generated neurons are directed by interactions between sleep-related neural processes and epigenetic mechanisms to facilitate novel approaches to preventing and treating brain disorders such as depression, post-traumatic stress disorder, and Alzheimer's disease. Stem Cells 2018;36:969-976.


Assuntos
Cognição/fisiologia , Emoções/fisiologia , Epigênese Genética/genética , Hipocampo/fisiopatologia , Neurogênese/genética , Sono/genética , Animais , Humanos , Camundongos
19.
Proc Natl Acad Sci U S A ; 113(21): 6023-8, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27140610

RESUMO

Neuropeptide B (NPB) and neuropeptide W (NPW) are endogenous neuropeptide ligands for the G protein-coupled receptors NPBWR1 and NPBWR2. Here we report that the majority of NPW neurons in the mesolimbic region possess tyrosine hydroxylase immunoreactivity, indicating that a small subset of dopaminergic neurons coexpress NPW. These NPW-containing neurons densely and exclusively innervate two limbic system nuclei in adult mouse brain: the lateral bed nucleus of the stria terminalis and the lateral part of the central amygdala nucleus (CeAL). In the CeAL of wild-type mice, restraint stress resulted in an inhibition of cellular activity, but this stress-induced inhibition was attenuated in the CeAL neurons of NPW(-/-) mice. Moreover, the response of NPW(-/-) mice to either formalin-induced pain stimuli or a live rat (i.e., a potential predator) was abnormal only when they were placed in a novel environment: The mice failed to show the normal species-specific self-protective and aversive reactions. In contrast, the behavior of NPW(-/-) mice in a habituated environment was indistinguishable from that of wild-type mice. These results indicate that the NPW/NPBWR1 system could play a critical role in the gating of stressful stimuli during exposure to novel environments.


Assuntos
Aprendizagem da Esquiva , Comportamento Animal , Núcleo Central da Amígdala/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neuropeptídeos/metabolismo , Dor/metabolismo , Animais , Núcleo Central da Amígdala/patologia , Neurônios Dopaminérgicos/patologia , Camundongos , Camundongos Knockout , Neuropeptídeos/genética , Dor/genética , Dor/patologia , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
20.
J Neurosci ; 37(30): 7164-7176, 2017 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-28642284

RESUMO

Emotionally salient situations usually trigger arousal along with autonomic and neuroendocrine reactions. To determine whether the extended amygdala plays a role in sleep-wakefulness regulation, we examined the effects of optogenetic and pharmacogenetic excitation of GABAergic neurons in the bed nucleus of the stria terminalis (GABABNST neurons). Acute optogenetic excitation of these cells during nonrapid eye movement (NREM) sleep resulted in an immediate state transition to wakefulness, whereas stimulation during REM sleep showed no effect on sleep-wakefulness states in male mice. An anterograde tracing study suggested GABABNST neurons send axonal projections to several brain regions implicated in arousal, including the preoptic area, lateral hypothalamus, periaqueductal gray, deep mesencephalic nucleus, and parabrachial nucleus. A dual orexin receptor antagonist, DORA-22, did not affect the optogenetic transition from NREM sleep to wakefulness. Chemogenetic excitation of GABABNST neurons evoked a sustained wakefulness state, but this arousal effect was markedly attenuated by DORA-22. These observations suggest that GABABNST neurons play an important role in transition from NREM sleep to wakefulness without the function of orexin neurons, but prolonged excitation of these cells mobilizes the orexin system to sustain wakefulness.SIGNIFICANCE STATEMENT We examined the role of the bed nucleus of the stria terminalis (BNST) in the regulation of wakefulness. Optogenetic excitation of GABAergic neurons in the BNST (GABABNST neurons) during nonrapid eye movement (NREM) sleep in mice resulted in immediate transition to a wakefulness state without function of orexins. Prolonged excitation of GABABNST neurons by a chemogenetic method evoked a longer-lasting, sustained wakefulness state, which was abolished by preadministration of a dual orexin receptor antagonist, DORA-22. This study revealed a role of the BNST GABAergic system in sleep-wakefulness control, especially in shifting animals' behavioral states from NREM sleep to wakefulness, and provides an important insight into the pathophysiology of insomnia and the role of orexin in arousal regulation.


Assuntos
Potenciais de Ação/fisiologia , Nível de Alerta/fisiologia , Neurônios GABAérgicos/fisiologia , Núcleos Septais/fisiologia , Fases do Sono/fisiologia , Vigília/fisiologia , Animais , Comportamento Animal/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA