Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
PLoS Comput Biol ; 18(6): e1010225, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35653331

RESUMEN

[This corrects the article DOI: 10.1371/journal.pcbi.1009316.].

2.
PLoS Comput Biol ; 17(8): e1009316, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34432801

RESUMEN

A salient feature of mammalian sleep is the alternation between rapid eye movement (REM) and non-REM (NREM) sleep. However, how these two sleep stages influence each other and thereby regulate the timing of REM sleep episodes is still largely unresolved. Here, we developed a statistical model that specifies the relationship between REM and subsequent NREM sleep to quantify how REM sleep affects the following NREM sleep duration and its electrophysiological features in mice. We show that a lognormal mixture model well describes how the preceding REM sleep duration influences the amount of NREM sleep till the next REM sleep episode. The model supports the existence of two different types of sleep cycles: Short cycles form closely interspaced sequences of REM sleep episodes, whereas during long cycles, REM sleep is first followed by an interval of NREM sleep during which transitions to REM sleep are extremely unlikely. This refractory period is characterized by low power in the theta and sigma range of the electroencephalogram (EEG), low spindle rate and frequent microarousals, and its duration proportionally increases with the preceding REM sleep duration. Using our model, we estimated the propensity for REM sleep at the transition from NREM to REM sleep and found that entering REM sleep with higher propensity resulted in longer REM sleep episodes with reduced EEG power. Compared with the light phase, the buildup of REM sleep propensity was slower during the dark phase. Our data-driven modeling approach uncovered basic principles underlying the timing and duration of REM sleep episodes in mice and provides a flexible framework to describe the ultradian regulation of REM sleep in health and disease.


Asunto(s)
Ritmo Circadiano , Probabilidad , Sueño REM , Animales , Electroencefalografía , Ratones
3.
Glia ; 64(3): 350-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26462610

RESUMEN

Myelination in corpus callosum plays important role for normal brain functions by transferring neurological information between various brain regions. However, the factors controlling expression of myelin genes in myelination are poorly understood. Here, CXXC5, a recently identified protein with CXXC-type zinc finger DNA binding motif, was characterized as a transcriptional activator of major myelin genes. We identified expression of CXXC5 expression was increased by Wnt/ß-catenin signaling. CXXC5 specifically expressed in the white matter induced expression of myelin genes through the direct binding of CXXC DNA-binding motif of CXXC5 on the MBP promoter. During the differentiation of neural stem cells (NSCs) of CXXC5(-/-) mice, the expressions of myelin genes were simultaneously reduced. The CXXC5(-/-) mice exhibited severely reduction of myelin genes expression in corpus callosum as well as abnormalities in myelin structure. The disrupted structural integrity of myelin in the CXXC5(-/-) mice resulted in reduced electrical conduction amplitudes at corpus callosum. These findings indicate that the regulation of myelin genes expression by CXXC5 is important for forming myelin structure involved with axonal electrical signal transfer in the corpus callosum.


Asunto(s)
Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/fisiología , Potenciales de Acción/genética , Animales , Animales Recién Nacidos , Axones/metabolismo , Axones/ultraestructura , Células Cultivadas , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/metabolismo , Proteínas de Unión al ADN , Embrión de Mamíferos , Proteína Ácida Fibrilar de la Glía/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Transgénicos , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/genética , Conducción Nerviosa/genética , Células-Madre Neurales , Oligodendroglía/ultraestructura , Factores de Transcripción , Vía de Señalización Wnt/genética , Proteína Wnt3A/farmacología , beta Catenina/metabolismo
4.
Paediatr Anaesth ; 25(10): 1033-45, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26095314

RESUMEN

OBJECTIVE: To examine whether neonatal exposure to sevoflurane induces autism-like behaviors in mice. BACKGROUND: There are continuing reports regarding the potential negative effects of anesthesia on the developing brain. Recently, several studies suggest that neurotoxicity caused by anesthesia may lead to neurodevelopmental impairments. However, unlike reports focusing on learning and memory, there are only a few animal studies focusing on neurodevelopmental disorders after general anesthesia. Therefore, we have focused on autism, a representative neurodevelopmental disorder. METHODS: Neonatal mice (P6-7) were exposed to a titrated dose of sevoflurane for 6 h. Apoptosis was evaluated by assessing the expression level of cleaved (activated) caspase-3. Autism-like behaviors, general activity, anxiety level, and long-term memory were evaluated with multiple behavioral assays. RESULTS: Western blotting confirmed that neonatal exposure to sevoflurane increased the expression level of activated caspase-3, indicative of apoptosis. Mice exposed to sevoflurane also showed impaired long-term memory in fear tests. However, sevoflurane-exposed mice did not exhibit autism-like features in all of the following assays: social interaction (three-chamber test, caged social interaction), social communication (ultrasonic vocalization test), or repetitive behavior (self-grooming test, digging). There were also no differences in general activity (open field test, home cage activity) and anxiety (open field test, light-dark box) after sevoflurane exposure. CONCLUSIONS: Our results confirm previous studies that neonatal sevoflurane exposure causes neurodegeneration and long-term memory impairment in mice. However, sevoflurane did not induce autism-like features. Our study suggests that mice are more vulnerable to long-term memory deficits than autism-like behaviors after exposure to sevoflurane.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Trastorno Autístico , Memoria a Largo Plazo/efectos de los fármacos , Éteres Metílicos/efectos adversos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Western Blotting , Caspasa 3/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Sevoflurano
5.
Curr Biol ; 34(10): 2247-2255.e5, 2024 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-38714199

RESUMEN

Rapid eye movement (REM) sleep is known to facilitate fear extinction and play a protective role against fearful memories.1,2 Consequently, disruption of REM sleep after a traumatic event may increase the risk for developing PTSD.3,4 However, the underlying mechanisms by which REM sleep promotes extinction of aversive memories remain largely unknown. The infralimbic cortex (IL) is a key brain structure for the consolidation of extinction memory.5 Using calcium imaging, we found in mice that most IL pyramidal neurons are intensively activated during REM sleep. Optogenetically suppressing the IL specifically during REM sleep within a 4-h window after auditory-cued fear conditioning impaired extinction memory consolidation. In contrast, REM-specific IL inhibition after extinction learning did not affect the extinction memory. Whole-cell patch-clamp recordings demonstrated that inactivating IL neurons during REM sleep depresses their excitability. Together, our findings suggest that REM sleep after fear conditioning facilitates fear extinction by enhancing IL excitability and highlight the importance of REM sleep in the aftermath of traumatic events for protecting against traumatic memories.


Asunto(s)
Extinción Psicológica , Miedo , Sueño REM , Animales , Miedo/fisiología , Sueño REM/fisiología , Ratones , Extinción Psicológica/fisiología , Masculino , Ratones Endogámicos C57BL , Memoria/fisiología , Consolidación de la Memoria/fisiología , Condicionamiento Clásico/fisiología , Células Piramidales/fisiología
6.
bioRxiv ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-37662417

RESUMEN

Rapid-eye-movement sleep (REMs) is characterized by activated electroencephalogram (EEG) and muscle atonia, accompanied by vivid dreams. REMs is homeostatically regulated, ensuring that any loss of REMs is compensated by a subsequent increase in its amount. However, the neural mechanisms underlying the homeostatic control of REMs are largely unknown. Here, we show that GABAergic neurons in the preoptic area of the hypothalamus projecting to the tuberomammillary nucleus (POAGAD2→TMN neurons) are crucial for the homeostatic regulation of REMs. POAGAD2→TMN neurons are most active during REMs, and inhibiting them specifically decreases REMs. REMs restriction leads to an increased number and amplitude of calcium transients in POAGAD2→TMN neurons, reflecting the accumulation of REMs pressure. Inhibiting POAGAD2→TMN neurons during REMs restriction blocked the subsequent rebound of REMs. Our findings reveal a hypothalamic circuit whose activity mirrors the buildup of homeostatic REMs pressure during restriction and that is required for the ensuing rebound in REMs.

7.
Elife ; 122024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884573

RESUMEN

Rapid eye movement sleep (REMs) is characterized by activated electroencephalogram (EEG) and muscle atonia, accompanied by vivid dreams. REMs is homeostatically regulated, ensuring that any loss of REMs is compensated by a subsequent increase in its amount. However, the neural mechanisms underlying the homeostatic control of REMs are largely unknown. Here, we show that GABAergic neurons in the preoptic area of the hypothalamus projecting to the tuberomammillary nucleus (POAGAD2→TMN neurons) are crucial for the homeostatic regulation of REMs in mice. POAGAD2→TMN neurons are most active during REMs, and inhibiting them specifically decreases REMs. REMs restriction leads to an increased number and amplitude of calcium transients in POAGAD2→TMN neurons, reflecting the accumulation of REMs pressure. Inhibiting POAGAD2→TMN neurons during REMs restriction blocked the subsequent rebound of REMs. Our findings reveal a hypothalamic circuit whose activity mirrors the buildup of homeostatic REMs pressure during restriction and that is required for the ensuing rebound in REMs.


Asunto(s)
Neuronas GABAérgicas , Homeostasis , Área Preóptica , Sueño REM , Animales , Área Preóptica/fisiología , Sueño REM/fisiología , Ratones , Neuronas GABAérgicas/fisiología , Masculino , Electroencefalografía , Área Hipotalámica Lateral/fisiología
8.
bioRxiv ; 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38234815

RESUMEN

Sleep disturbances are prevalent in children with autism spectrum disorder (ASD) and have a major impact on the quality of life. Strikingly, sleep problems are positively correlated with the severity of ASD symptoms, such as memory impairment. However, the neural mechanisms underlying sleep disturbances and cognitive deficits in ASD are largely unexplored. Here, we show that non-rapid eye movement sleep (NREMs) is highly fragmented in the 16p11.2 deletion mouse model of ASD. The degree of sleep fragmentation is reflected in an increased number of calcium transients in the activity of locus coeruleus noradrenergic (LC-NE) neurons during NREMs. Exposure to a novel environment further exacerbates sleep disturbances in 16p11.2 deletion mice by fragmenting NREMs and decreasing rapid eye movement sleep (REMs). In contrast, optogenetic inhibition of LC-NE neurons and pharmacological blockade of noradrenergic transmission using clonidine reverse sleep fragmentation. Furthermore, inhibiting LC-NE neurons restores memory. Rabies-mediated unbiased screening of presynaptic neurons reveals altered connectivity of LC-NE neurons with sleep- and memory regulatory brain regions in 16p11.2 deletion mice. Our findings demonstrate that heightened activity of LC-NE neurons and altered brain-wide connectivity underlies sleep fragmentation in 16p11.2 deletion mice and identify a crucial role of the LC-NE system in regulating sleep stability and memory in ASD.

9.
Res Sq ; 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38559267

RESUMEN

Sleep disturbances are prevalent in children with autism spectrum disorder (ASD) and have a major impact on the quality of life. Strikingly, sleep problems are positively correlated with the severity of ASD symptoms, such as memory impairment. However, the neural mechanisms underlying sleep disturbances and cognitive deficits in ASD are largely unexplored. Here, we show that non-rapid eye movement sleep (NREMs) is highly fragmented in the 16p11.2 deletion mouse model of ASD. The degree of sleep fragmentation is reflected in an increased number of calcium transients in the activity of locus coeruleus noradrenergic (LC-NE) neurons during NREMs. Exposure to a novel environment further exacerbates sleep disturbances in 16p11.2 deletion mice by fragmenting NREMs and decreasing rapid eye movement sleep (REMs). In contrast, optogenetic inhibition of LC-NE neurons and pharmacological blockade of noradrenergic transmission using clonidine reverse sleep fragmentation. Furthermore, inhibiting LC-NE neurons restores memory. Rabies-mediated unbiased screening of presynaptic neurons reveals altered connectivity of LC-NE neurons with sleep- and memory regulatory brain regions in 16p11.2 deletion mice. Our findings demonstrate that heightened activity of LC-NE neurons and altered brain-wide connectivity underlies sleep fragmentation in 16p11.2 deletion mice and identify a crucial role of the LC-NE system in regulating sleep stability and memory in ASD.

10.
Nat Neurosci ; 26(10): 1820-1832, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37735498

RESUMEN

Rapid eye movement (REM) sleep is accompanied by intense cortical activity, underlying its wake-like electroencephalogram. The neural activity inducing REM sleep is thought to originate from subcortical circuits in brainstem and hypothalamus. However, whether cortical neurons can also trigger REM sleep has remained unknown. Here we show in mice that the medial prefrontal cortex (mPFC) strongly promotes REM sleep. Bidirectional optogenetic manipulations demonstrate that excitatory mPFC neurons promote REM sleep through their projections to the lateral hypothalamus and regulate phasic events, reflected in accelerated electroencephalogram theta oscillations and increased eye movement density during REM sleep. Calcium imaging reveals that the majority of lateral hypothalamus-projecting mPFC neurons are maximally activated during REM sleep and a subpopulation is recruited during phasic theta accelerations. Our results delineate a cortico-hypothalamic circuit for the top-down control of REM sleep and identify a critical role of the mPFC in regulating phasic events during REM sleep.


Asunto(s)
Neuronas , Sueño REM , Ratones , Animales , Sueño REM/fisiología , Neuronas/fisiología , Hipotálamo/fisiología , Corteza Prefrontal/fisiología , Área Hipotalámica Lateral , Sueño/fisiología
11.
Res Sq ; 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37886570

RESUMEN

Rapid-eye-movement (REM) sleep is accompanied by intense cortical activity, underlying its wake-like electroencephalogram (EEG). The neural activity inducing REM sleep is thought to originate from subcortical circuits in brainstem and hypothalamus. However, whether cortical neurons can also trigger REM sleep has remained unknown. Here, we show in mice that the medial prefrontal cortex (mPFC) strongly promotes REM sleep. Bidirectional optogenetic manipulations demonstrate that excitatory mPFC neurons promote REM sleep through their projections to the lateral hypothalamus (LH) and regulate phasic events, reflected in accelerated EEG theta oscillations and increased eye-movement density during REM sleep. Calcium imaging reveals that the majority of LH-projecting mPFC neurons are maximally activated during REM sleep and a subpopulation is recruited during phasic theta accelerations. Our results delineate a cortico-hypothalamic circuit for the top-down control of REM sleep and identify a critical role of the mPFC in regulating phasic events during REM sleep.

12.
Mol Brain ; 14(1): 92, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34127022

RESUMEN

Functional lateralization of the prefrontal cortex has been implicated in stress and emotional disorders, yet underlying gene expression changes remains unknown. Here, we report molecular signatures lateralized by chronic social defeats between the two medial prefrontal cortices (mPFCs). Stressed mice show 526 asymmetrically expressed genes between the mPFCs. This cortical asymmetry selectively occurs in stressed mice with depressed social activity, but not in resilient mice with normal behavior. We have isolated highly asymmetric genes including connective tissue growth factor (CTGF), a molecule that modulates wound healing at the periphery. Knockdown of CTGF gene in the right mPFC by shRNA led to a stress-resistant behavioral phenotype. Overexpression of CTGF in the right mPFC using viral transduction induces social avoidance while the left mPFC thereof prevent stress-induced social avoidance. Our study provides a molecular window into the mechanism of stress-induced socioemotional disorders, which can pave the way for new interventions by targeting cortical asymmetry.


Asunto(s)
Corteza Prefrontal/patología , Estrés Psicológico/patología , Animales , Reacción de Prevención , Enfermedad Crónica , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Anotación de Secuencia Molecular , Resiliencia Psicológica , Derrota Social , Estrés Psicológico/genética
13.
Nat Neurosci ; 21(6): 895, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29515215

RESUMEN

In the version of this article initially published, a sentence in the fifth paragraph of the Results read, "Immunohistochemistry revealed that VGLUT2+ MPA neurons rarely expressed CaMKIIα, which is a putative marker for subcortical glutamatergic neurons." It should have read, "Immunohistochemistry revealed that CaMKIIα+ MPA neurons rarely expressed VGLUT2, which is a putative marker for subcortical glutamatergic neurons." The error has been corrected in the HTML and PDF versions of the article. In the supplementary information originally posted online, the wrong version of Supplementary Fig. 1 was posted and some of the supplementary videos were interchanged. In the corrected Supplementary Fig. 1, the top right subpanel was added and the original Supplementary Fig. 1a was divided into 1a and 1b, with subsequent panels incremented accordingly. The legend was changed from "a. Schematic illustrating electrical lesioning of the rat anterior hypothalamus. Electrical lesion areas (gray) in five representative brain sections are depicted. Scale bar, 1 mm" to "a. Repetitive electrical stimulations of the anterior hypothalamus using bipolar electrodes (Left) caused a lesion at the hypothalamic area (middle, marked by asterisk) successfully in 7 rats (Right, overlapped images of brain sections located from the bregma -0.24 mm). Scale bar, 1 mm. b. Electrical lesion areas (gray) in five representative brain sections from anterior to posterior are depicted." The errors have been corrected online.

14.
Nat Neurosci ; 21(3): 364-372, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29379117

RESUMEN

As animals forage, they must obtain useful targets by orchestrating appropriate actions that range from searching to chasing, biting and carrying. Here, we reveal that neurons positive for the α subunit of Ca2+/calmodulin-dependent kinase II (CaMKIIα) in the medial preoptic area (MPA) that send projections to the ventral periaqueductal gray (vPAG) mediate these target-directed actions in mice. During photostimulation of the MPA-vPAG circuit, mice vigorously engaged with 3D objects and chased moving objects. When exposed to a cricket, they hunted down the prey and bit it to kill. By applying a head-mounted object control with timely photostimulation of the MPA-vPAG circuit, we found that MPA-vPAG circuit-induced actions occurred only when the target was detected within the binocular visual field. Using this device, we successfully guided mice to navigate specified routes. Our study explains how the brain yields a strong motivation to acquire a target object along the continuum of hunting behavior.


Asunto(s)
Conducta Predatoria/fisiología , Área Preóptica/fisiología , Animales , Conducta Alimentaria , Gryllidae , Masculino , Ratones , Ratones Endogámicos C57BL , Motivación/fisiología , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuronas/fisiología , Sustancia Gris Periacueductal/citología , Sustancia Gris Periacueductal/fisiología , Estimulación Luminosa , Área Preóptica/citología , Ratas , Ratas Long-Evans , Conducta Sexual Animal/fisiología , Visión Binocular/fisiología , Campos Visuales/fisiología
15.
Sci Rep ; 7(1): 5363, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28706238

RESUMEN

The infralimbic cortex (IL) is known to facilitate the formation of extinction memory through reciprocal interactions with the amygdala, which produces fear responses such as freezing. Thus, whether presynaptic input from the amygdala and post-synaptic output of IL neurons are functionally dissociated in extinction memory formation remains unclear. Here, we demonstrated that photostimulation of IL inputs from BLA did not change freezing responses to conditioned stimuli (CS) during training, but did facilitate extinction memory, measured as a reduction in freezing responses to the CS 1 day later. On the other hand, photostimulation of somata of IL neurons induced an immediate reduction in freezing to CS, but this did not affect extinction memory tested the next day. These results provide in vivo evidence for IL-dependent facilitation of extinction memory without post-synaptic modulation of freezing circuits.


Asunto(s)
Respuesta al Choque por Frío , Extinción Psicológica , Miedo , Memoria , Corteza Prefrontal/fisiología , Animales , Condicionamiento Clásico , Masculino , Ratones Endogámicos C57BL
16.
Sci Rep ; 5: 13342, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26302668

RESUMEN

When subjected to stress, some individuals develop maladaptive symptoms whereas others retain normal behavior. The medial prefrontal cortex (mPFC) is known to control these adaptive responses to stress. Here, we show that mPFC neurons in the left hemisphere control stress effects on social behavior. Mice made socially avoidant by the stress of chronic social defeats showed depressed neural activity in the left mPFC. Photoactivation of these neurons reversed social avoidance and restored social activity. Despite social defeats, resilient mice with normal sociability showed normal firing rates in the left mPFC; however, photoinhibition of these neurons induced social avoidance. The same photomodulation administered to the right mPFC caused no significant effects. These results explain how stressed individuals develop maladaptive behaviors through left cortical depression, as reported in mood and anxiety disorders.


Asunto(s)
Dominancia Cerebral , Red Nerviosa/fisiopatología , Corteza Prefrontal/fisiopatología , Trastorno de la Conducta Social/fisiopatología , Conducta Social , Estrés Psicológico/fisiopatología , Adaptación Fisiológica , Animales , Reacción de Prevención , Conducta Animal , Masculino , Ratones , Ratones Endogámicos C57BL , Trastorno de la Conducta Social/complicaciones , Estrés Psicológico/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA