Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cell ; 162(3): 622-34, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26232228

RESUMO

Dopamine (DA) neurons in the midbrain ventral tegmental area (VTA) integrate complex inputs to encode multiple signals that influence motivated behaviors via diverse projections. Here, we combine axon-initiated viral transduction with rabies-mediated trans-synaptic tracing and Cre-based cell-type-specific targeting to systematically map input-output relationships of VTA-DA neurons. We found that VTA-DA (and VTA-GABA) neurons receive excitatory, inhibitory, and modulatory input from diverse sources. VTA-DA neurons projecting to different forebrain regions exhibit specific biases in their input selection. VTA-DA neurons projecting to lateral and medial nucleus accumbens innervate largely non-overlapping striatal targets, with the latter also sending extensive extra-striatal axon collaterals. Using electrophysiology and behavior, we validated new circuits identified in our tracing studies, including a previously unappreciated top-down reinforcing circuit from anterior cortex to lateral nucleus accumbens via VTA-DA neurons. This study highlights the utility of our viral-genetic tracing strategies to elucidate the complex neural substrates that underlie motivated behaviors.


Assuntos
Vias Neurais , Neurônios/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Animais , Mapeamento Encefálico , Dopamina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Accumbens/metabolismo , Vírus da Raiva , Ácido gama-Aminobutírico/metabolismo
2.
PLoS Biol ; 20(10): e3001813, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36194579

RESUMO

The reduced sleep duration previously observed in Camk2b knockout mice revealed a role for Ca2+/calmodulin-dependent protein kinase II (CaMKII)ß as a sleep-promoting kinase. However, the underlying mechanism by which CaMKIIß supports sleep regulation is largely unknown. Here, we demonstrate that activation or inhibition of CaMKIIß can increase or decrease sleep duration in mice by almost 2-fold, supporting the role of CaMKIIß as a core sleep regulator in mammals. Importantly, we show that this sleep regulation depends on the kinase activity of CaMKIIß. A CaMKIIß mutant mimicking the constitutive-active (auto)phosphorylation state promotes the transition from awake state to sleep state, while mutants mimicking subsequent multisite (auto)phosphorylation states suppress the transition from sleep state to awake state. These results suggest that the phosphorylation states of CaMKIIß differently control sleep induction and maintenance processes, leading us to propose a "phosphorylation hypothesis of sleep" for the molecular control of sleep in mammals.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Cálcio , Animais , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Mamíferos/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Sono
3.
J Neurosci ; 40(20): 3981-3994, 2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32284340

RESUMO

Male animals may show alternative behaviors toward infants: attack or parenting. These behaviors are triggered by pup stimuli under the influence of the internal state, including the hormonal environment and/or social experiences. Converging data suggest that the medial preoptic area (MPOA) contributes to the behavioral selection toward the pup. However, the neural mechanisms underlying how integrated stimuli affect the MPOA-dependent behavioral selection remain unclear. Here we focus on the amygdalohippocampal area (AHi) that projects to MPOA and expresses oxytocin receptor, a hormone receptor mediating social behavior toward pups. We describe the activation of MPOA-projection AHi neurons in male mice by social contact with pups. Input mapping using the TRIO method reveals that MPOA-projection AHi neurons receive prominent inputs from several regions, including the thalamus, hypothalamus, and olfactory cortex. Electrophysiological and histologic analysis demonstrates that oxytocin modulates inhibitory synaptic responses on MPOA-projection AHi neurons. In addition, AHi forms the excitatory monosynapse to MPOA, and pharmacological activation of MPOA-projection AHi neurons enhances only aggressive behavior, but not parental behavior. Interestingly, this promoted behavior was related to social experience in male mice. Collectively, our results identified a presynaptic partner of MPOA that can integrate sensory input and hormonal state, and trigger pup-directed aggression.SIGNIFICANCE STATEMENT The medial preoptic area (MPOA) plays critical roles in parental behavior, such as motor control, motivation, and social interaction. The MPOA projects to multiple brain regions, and these projections contribute to several neural controls in parental behavior. In contrast, how inputs to MPOA are regulated by social and environmental information is poorly understood. In this study, we focus on the amygdalohippocampal area (AHi) that connects to MPOA and expresses oxytocin receptor. We demonstrate the disruption of the expression of parental behavior triggered by the activation of MPOA-projection AHi neurons. This behavior may be regulated not only by oxytocin but also by neural input from several regions.


Assuntos
Agressão/fisiologia , Tonsila do Cerebelo/fisiologia , Hipocampo/fisiologia , Vias Neurais/fisiologia , Neurônios/fisiologia , Área Pré-Óptica/fisiologia , Tonsila do Cerebelo/citologia , Animais , Mapeamento Encefálico , Fenômenos Eletrofisiológicos , Hipocampo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural , Comportamento Paterno , Área Pré-Óptica/citologia , Receptores de Ocitocina/metabolismo , Comportamento Social , Meio Social
4.
Nature ; 524(7563): 88-92, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26131933

RESUMO

Deciphering how neural circuits are anatomically organized with regard to input and output is instrumental in understanding how the brain processes information. For example, locus coeruleus noradrenaline (also known as norepinephrine) (LC-NE) neurons receive input from and send output to broad regions of the brain and spinal cord, and regulate diverse functions including arousal, attention, mood and sensory gating. However, it is unclear how LC-NE neurons divide up their brain-wide projection patterns and whether different LC-NE neurons receive differential input. Here we developed a set of viral-genetic tools to quantitatively analyse the input-output relationship of neural circuits, and applied these tools to dissect the LC-NE circuit in mice. Rabies-virus-based input mapping indicated that LC-NE neurons receive convergent synaptic input from many regions previously identified as sending axons to the locus coeruleus, as well as from newly identified presynaptic partners, including cerebellar Purkinje cells. The 'tracing the relationship between input and output' method (or TRIO method) enables trans-synaptic input tracing from specific subsets of neurons based on their projection and cell type. We found that LC-NE neurons projecting to diverse output regions receive mostly similar input. Projection-based viral labelling revealed that LC-NE neurons projecting to one output region also project to all brain regions we examined. Thus, the LC-NE circuit overall integrates information from, and broadcasts to, many brain regions, consistent with its primary role in regulating brain states. At the same time, we uncovered several levels of specificity in certain LC-NE sub-circuits. These tools for mapping output architecture and input-output relationship are applicable to other neuronal circuits and organisms. More broadly, our viral-genetic approaches provide an efficient intersectional means to target neuronal populations based on cell type and projection pattern.


Assuntos
Encéfalo/citologia , Encéfalo/metabolismo , Técnicas de Rastreamento Neuroanatômico/métodos , Neurônios/metabolismo , Neurônios/virologia , Norepinefrina/metabolismo , Vírus da Raiva/fisiologia , Animais , Axônios/fisiologia , Axônios/virologia , Encéfalo/virologia , Feminino , Locus Cerúleo/citologia , Locus Cerúleo/metabolismo , Locus Cerúleo/virologia , Masculino , Camundongos , Vias Neurais , Projetos Piloto , Células de Purkinje/fisiologia , Células de Purkinje/virologia , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Sinapses/metabolismo , Sinapses/virologia
5.
Nature ; 472(7342): 191-6, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21179085

RESUMO

In the mouse, each class of olfactory receptor neurons expressing a given odorant receptor has convergent axonal projections to two specific glomeruli in the olfactory bulb, thereby creating an odour map. However, it is unclear how this map is represented in the olfactory cortex. Here we combine rabies-virus-dependent retrograde mono-trans-synaptic labelling with genetics to control the location, number and type of 'starter' cortical neurons, from which we trace their presynaptic neurons. We find that individual cortical neurons receive input from multiple mitral cells representing broadly distributed glomeruli. Different cortical areas represent the olfactory bulb input differently. For example, the cortical amygdala preferentially receives dorsal olfactory bulb input, whereas the piriform cortex samples the whole olfactory bulb without obvious bias. These differences probably reflect different functions of these cortical areas in mediating innate odour preference or associative memory. The trans-synaptic labelling method described here should be widely applicable to mapping connections throughout the mouse nervous system.


Assuntos
Técnicas de Rastreamento Neuroanatômico , Condutos Olfatórios/citologia , Condutos Olfatórios/fisiologia , Percepção Olfatória/fisiologia , Sinapses/metabolismo , Tonsila do Cerebelo/anatomia & histologia , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Animais , Axônios/fisiologia , Viés , Mapeamento Encefálico , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Odorantes/análise , Bulbo Olfatório/anatomia & histologia , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Condutos Olfatórios/anatomia & histologia , Percepção Olfatória/genética , Neurônios Receptores Olfatórios/citologia , Neurônios Receptores Olfatórios/fisiologia , Vírus da Raiva/fisiologia , Sinapses/genética
6.
Cereb Cortex ; 26(4): 1453-1463, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25316337

RESUMO

Synapse pruning is an activity-regulated process needed for proper circuit sculpting in the developing brain. Major histocompatibility class I (MHCI) molecules are regulated by activity, but little is known about their role in the development of connectivity in cortex. Here we show that protein for 2 MHCI molecules H2-Kb and H2-Db is associated with synapses in the visual cortex. Pyramidal neurons in mice lacking H2-Kb and H2-Db (KbDb KO) have more extensive cortical connectivity than normal. Modified rabies virus tracing was used to monitor the extent of pyramidal cell connectivity: Horizontal connectivity is greater in the visual cortex of KbDb KO mice. Basal dendrites of L2/3 pyramids, where many horizontal connections terminate, are more highly branched and have elevated spine density in the KO. Furthermore, the density of axonal boutons is elevated within L2/3 of mutant mice. These increases are accompanied by elevated miniature excitatory postsynaptic current frequency, consistent with an increase in functional synapses. This functional and anatomical increase in intracortical connectivity is also associated with enhanced ocular dominance plasticity that persists into adulthood. Thus, these MHCI proteins regulate sculpting of local cortical circuits and in their absence, the excess connectivity can function as a substrate for cortical plasticity throughout life.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Genes MHC Classe I , Células Piramidais/citologia , Células Piramidais/fisiologia , Sinapses/fisiologia , Córtex Visual/citologia , Córtex Visual/fisiologia , Animais , Axônios , Espinhas Dendríticas , Potenciais Pós-Sinápticos Excitadores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal , Sinapses/genética
7.
J Neurosci ; 34(28): 9404-17, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25009272

RESUMO

The motor function of the spinal cord requires the computation of the local neuronal circuits within the same segments as well as the long-range coordination of different spinal levels. Implicated players in this process are the propriospinal neurons (PPNs) that project their axons across different levels of the spinal cord. However, their cellular, molecular, and functional properties remain unknown. Here we use a recombinant rabies virus-based method to label a specific type of long-projecting premotor PPNs in the mouse upper spinal cord that are monosynaptically connected to the motor neurons in the lumbar spinal cord. With a whole spinal cord imaging method, we find that these neurons are distributed along the entire length of the upper spinal cord with more in the lower thoracic levels. Among them, a subset of thoracic PPNs receive substantial numbers of sensory inputs, suggesting a function in coordinating the activity of trunk and hindlimb muscles. Although many PPNs in the cervical and thoracic spinal cord receive the synaptic inputs from corticospinal tract or serotonergic axons, limited bouton numbers suggested that these supraspinal inputs might not be major regulators of the PPNs in intact animals. Molecularly, these PPNs appear to be distinct from other known premotor interneurons, but some are derived from Chx10+ lineages. This study provides an anatomical basis for further exploring different functions of PPNs.


Assuntos
Neurônios Motores/citologia , Tratos Piramidais/citologia , Células Receptoras Sensoriais/citologia , Medula Espinal/citologia , Animais , Feminino , Masculino , Camundongos , Vias Neurais/citologia
8.
Proc Natl Acad Sci U S A ; 108(37): 15414-9, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21825165

RESUMO

To understand how the nervous system processes information, a map of the connections among neurons would be of great benefit. Here we describe the use of vesicular stomatitis virus (VSV) for tracing neuronal connections in vivo. We made VSV vectors that used glycoprotein (G) genes from several other viruses. The G protein from lymphocytic choriomeningitis virus endowed VSV with the ability to spread transsynaptically, specifically in an anterograde direction, whereas the rabies virus glycoprotein gave a specifically retrograde transsynaptic pattern. The use of an avian G protein fusion allowed specific targeting of cells expressing an avian receptor, which allowed a demonstration of monosynaptic anterograde tracing from defined cells. Synaptic connectivity of pairs of virally labeled cells was demonstrated by using slice cultures and electrophysiology. In vivo infections of several areas in the mouse brain led to the predicted patterns of spread for anterograde or retrograde tracers.


Assuntos
Sistema Nervoso Central/citologia , Vetores Genéticos/genética , Neurônios/metabolismo , Coloração e Rotulagem , Sinapses/metabolismo , Vesiculovirus/genética , Animais , Encéfalo/virologia , Olho/virologia , Vírus da Coriomeningite Linfocítica , Camundongos , Condutos Olfatórios/metabolismo , Condutos Olfatórios/virologia , Recombinação Genética/genética
9.
Nat Commun ; 15(1): 6054, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39025867

RESUMO

The homeostatic regulation of sleep is characterized by rebound sleep after prolonged wakefulness, but the molecular and cellular mechanisms underlying this regulation are still unknown. In this study, we show that Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent activity control of parvalbumin (PV)-expressing cortical neurons is involved in homeostatic regulation of sleep in male mice. Prolonged wakefulness enhances cortical PV-neuron activity. Chemogenetic suppression or activation of cortical PV neurons inhibits or induces rebound sleep, implying that rebound sleep is dependent on increased activity of cortical PV neurons. Furthermore, we discovered that CaMKII kinase activity boosts the activity of cortical PV neurons, and that kinase activity is important for homeostatic sleep rebound. Here, we propose that CaMKII-dependent PV-neuron activity represents negative feedback inhibition of cortical neural excitability, which serves as the distributive cortical circuits for sleep homeostatic regulation.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Córtex Cerebral , Homeostase , Neurônios , Parvalbuminas , Sono , Vigília , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Parvalbuminas/metabolismo , Masculino , Sono/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Camundongos , Vigília/fisiologia , Córtex Cerebral/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
10.
Front Neural Circuits ; 17: 1340497, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38298741

RESUMO

Parental care plays a crucial role in the physical and mental well-being of mammalian offspring. Although sexually naïve male mice, as well as certain strains of female mice, display aggression toward pups, they exhibit heightened parental caregiving behaviors as they approach the time of anticipating their offspring. In this Mini Review, I provide a concise overview of the current understanding of distinct limbic neural types and their circuits governing both aggressive and caregiving behaviors toward infant mice. Subsequently, I delve into recent advancements in the understanding of the molecular, cellular, and neural circuit mechanisms that regulate behavioral plasticity during the transition to parenthood, with a specific focus on the sex steroid hormone estrogen and neural hormone oxytocin. Additionally, I explore potential sex-related differences and highlight some critical unanswered questions that warrant further investigation.


Assuntos
Agressão , Comportamento Paterno , Humanos , Camundongos , Masculino , Animais , Feminino , Comportamento Paterno/fisiologia , Agressão/fisiologia , Ocitocina , Mamíferos
11.
Neurosci Res ; 196: 1-10, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37343600

RESUMO

In recent decades, human fathers across the globe have shown a substantial increase in their engagement in paternal caregiving behaviors. Despite the growing interest, the precise neurobiological mechanisms underlying caregiving behaviors in males remain unclear. Neurobiological studies conducted on rodents have advanced our understanding of the molecular, cellular, and circuit-level mechanisms. Typically, sexually naïve males exhibit aggression toward offspring, while fathers display parental behaviors. This drastic behavioral plasticity may be associated with changes in connections among specific regions or cell types. Recent studies have begun to describe this structural plasticity by comparing neural connections before and after fatherhood. In this Perspective, we summarize the findings from four well-studied rodent species, namely prairie voles, California mice, laboratory rats, and laboratory mice, with a view toward integrating past and current progress. We then review recent advances in the understanding of structural plasticity for parental behaviors. Finally, we discuss remaining questions that require further exploration to gain a deeper understanding of the neural mechanisms underlying paternal behaviors in males, including their possible implications for the human brain.


Assuntos
Encéfalo , Roedores , Animais , Masculino , Humanos , Encéfalo/metabolismo , Comportamento Paterno , Neurônios/metabolismo , Arvicolinae , Pais
12.
PLoS One ; 18(3): e0283152, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36930664

RESUMO

The hormone oxytocin, secreted from oxytocin neurons in the paraventricular (PVH) and supraoptic (SO) hypothalamic nuclei, promotes parturition, milk ejection, and maternal caregiving behaviors. Previous experiments with whole-body oxytocin knockout mice showed that milk ejection was the unequivocal function of oxytocin, whereas parturition and maternal behaviors were less dependent on oxytocin. Whole-body knockout, however, could induce the enhancement of expression of related gene(s), a phenomenon called genetic compensation, which may hide the actual functions of oxytocin. In addition, the relative contributions of oxytocin neurons in the PVH and SO have not been well documented. Here, we show that females with conditional knockout of oxytocin gene in both the PVH and SO undergo grossly normal parturition and maternal caregiving behaviors, while dams with a smaller number of remaining oxytocin-expressing neurons exhibit severe impairments in breastfeeding, leading to the death of their pups within 24 hours after birth. We also found that the growth of pups is normal even under oxytocin conditional knockout in PVH and SO as long as pups survive the next day of delivery, suggesting that the reduced oxytocin release affects the onset of lactation most severely. These phenotypes are largely recapitulated by SO-specific oxytocin conditional knockout, indicating the unequivocal role of oxytocin neurons in the SO in successful breastfeeding. Given that oxytocin neurons not only secrete oxytocin but also non-oxytocin neurotransmitters or neuropeptides, we further performed cell ablation of oxytocin neurons in the PVH and SO. We found that cell ablation of oxytocin neurons leads to no additional abnormalities over the oxytocin conditional knockout, suggesting that non-oxytocin ligands expressed by oxytocin neurons have negligible functions on the responses measured in this study. Collectively, our findings confirm the dispensability of oxytocin for parturition or maternal behaviors, as well as the importance of SO-derived oxytocin for breastfeeding.


Assuntos
Ocitocina , Núcleo Supraóptico , Feminino , Camundongos , Animais , Ocitocina/farmacologia , Núcleo Supraóptico/metabolismo , Neurônios/metabolismo , Hipotálamo/metabolismo , Lactação/fisiologia , Núcleo Hipotalâmico Paraventricular/metabolismo
13.
Elife ; 122023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37223988

RESUMO

Reproductive senescence is broadly observed across mammalian females, including humans, eventually leading to a loss of fertility. The pulsatile secretion of gonadotropin-releasing hormone (GnRH), which is essential for gonad function, is primarily controlled by kisspeptin neurons in the hypothalamic arcuate nucleus (ARCkiss), the pulse generator of GnRH. The pulsatility of GnRH release, as assessed by the amount of circulating gonadotropin, is markedly reduced in aged animals, suggesting that the malfunctions of ARCkiss may be responsible for reproductive aging and menopause-related disorders. However, the activity dynamics of ARCkiss during the natural transition to reproductive senescence remain unclear. Herein, we introduce chronic in vivo Ca2+ imaging of ARCkiss in female mice by fiber photometry to monitor the synchronous episodes of ARCkiss (SEskiss), a known hallmark of GnRH pulse generator activity, from the fully reproductive to acyclic phase over 1 year. During the reproductive phase, we find that not only the frequency, but also the intensities and waveforms of individual SEskiss, vary depending on the stage of the estrus cycle. During the transition to reproductive senescence, the integrity of SEskiss patterns, including the frequency and waveforms, remains mostly unchanged, whereas the intensities tend to decline. These data illuminate the temporal dynamics of ARCkiss activities in aging female mice. More generally, our findings demonstrate the utility of fiber-photometry-based chronic imaging of neuroendocrine regulators in the brain to characterize aging-associated malfunction.


Assuntos
Kisspeptinas , Neurônios , Reprodução , Animais , Feminino , Camundongos , Envelhecimento , Hormônio Liberador de Gonadotropina
14.
PLoS One ; 18(5): e0285589, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37163565

RESUMO

Breastfeeding, which is essential for the survival of mammalian infants, is critically mediated by pulsatile secretion of the pituitary hormone oxytocin from the central oxytocin neurons located in the paraventricular and supraoptic hypothalamic nuclei of mothers. Despite its importance, the molecular and neural circuit mechanisms of the milk ejection reflex remain poorly understood, in part because a mouse model to study lactation was only recently established. In our previous study, we successfully introduced fiber photometry-based chronic imaging of the pulsatile activities of oxytocin neurons during lactation. However, the necessity of Cre recombinase-based double knock-in mice substantially compromised the use of various Cre-dependent neuroscience toolkits. To overcome this obstacle, we developed a simple Cre-free method for monitoring oxytocin neurons by an adeno-associated virus vector driving GCaMP6s under a 2.6 kb mouse oxytocin mini-promoter. Using this method, we monitored calcium ion transients of oxytocin neurons in the paraventricular nucleus in wild-type C57BL/6N and ICR mothers without genetic crossing. By combining this method with video recordings of mothers and pups, we found that the pulsatile activities of oxytocin neurons require physical mother-pup contact for the milk ejection reflex. Notably, the frequencies of photometric signals were dynamically modulated by mother-pup reunions after isolation and during natural weaning stages. Collectively, the present study illuminates the temporal dynamics of pulsatile activities of oxytocin neurons in wild-type mice and provides a tool to characterize maternal oxytocin functions.


Assuntos
Lactação , Ocitocina , Feminino , Camundongos , Animais , Lactação/fisiologia , Ocitocina/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Neurônios/fisiologia , Núcleo Supraóptico/fisiologia , Núcleo Hipotalâmico Paraventricular , Mamíferos
15.
Elife ; 112022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36281647

RESUMO

Decades of studies have revealed molecular and neural circuit bases for body weight homeostasis. Neural hormone oxytocin (Oxt) has received attention in this context because it is produced by neurons in the paraventricular hypothalamic nucleus (PVH), a known output center of hypothalamic regulation of appetite. Oxt has an anorexigenic effect, as shown in human studies, and can mediate satiety signals in rodents. However, the function of Oxt signaling in the physiological regulation of appetite has remained in question, because whole-body knockout (KO) of Oxt or Oxt receptor (Oxtr) has little effect on food intake. We herein show that acute conditional KO (cKO) of Oxt selectively in the adult PVH, but not in the supraoptic nucleus, markedly increases body weight and food intake, with an elevated level of plasma triglyceride and leptin. Intraperitoneal administration of Oxt rescues the hyperphagic phenotype of the PVH Oxt cKO model. Furthermore, we show that cKO of Oxtr selectively in the posterior hypothalamic regions, especially the arcuate hypothalamic nucleus, a primary center for appetite regulations, phenocopies hyperphagic obesity. Collectively, these data reveal that Oxt signaling in the arcuate nucleus suppresses excessive food intake.


Assuntos
Leptina , Ocitocina , Humanos , Camundongos , Animais , Hiperfagia , Obesidade/genética , Núcleo Hipotalâmico Paraventricular , Peso Corporal , Hipotálamo , Hipotálamo Posterior , Triglicerídeos
16.
Neuron ; 110(12): 2009-2023.e5, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35443152

RESUMO

The adult brain can flexibly adapt behaviors to specific life-stage demands. For example, while sexually naive male mice are aggressive to the conspecific young, they start to provide caregiving to infants around the time when their own young are expected. How such behavioral plasticity is implemented at the level of neural connections remains poorly understood. Here, using viral-genetic approaches, we establish hypothalamic oxytocin neurons as the key regulators of the parental caregiving behaviors of male mice. We then use rabies-virus-mediated unbiased screening to identify excitatory neural connections originating from the lateral hypothalamus to the oxytocin neurons to be drastically strengthened when male mice become fathers. These connections are functionally relevant, as their activation suppresses pup-directed aggression in virgin males. These results demonstrate the life-stage associated, long-distance, and cell-type-specific plasticity of neural connections in the hypothalamus, the brain region that is classically assumed to be hard-wired.


Assuntos
Agressão , Ocitocina , Agressão/fisiologia , Animais , Humanos , Hipotálamo/fisiologia , Masculino , Camundongos , Neurônios/fisiologia , Pais
17.
Neuron ; 110(15): 2455-2469.e8, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35654036

RESUMO

The pheromonal information received by the vomeronasal system plays a crucial role in regulating social behaviors such as aggression in mice. Despite accumulating knowledge of the brain regions involved in aggression, the specific vomeronasal receptors and the exact neural circuits responsible for pheromone-mediated aggression remain unknown. Here, we identified one murine vomeronasal receptor, Vmn2r53, that is activated by urine from males of various strains and is responsible for evoking intermale aggression. We prepared a purified pheromonal fraction and Vmn2r53 knockout mice and applied genetic tools for neuronal activity recording, manipulation, and circuit tracing to decipher the neural mechanisms underlying Vmn2r53-mediated aggression. We found that Vmn2r53-mediated aggression is regulated by specific neuronal populations in the ventral premammillary nucleus and the ventromedial hypothalamic nucleus. Together, our results shed light on the hypothalamic regulation of male aggression mediated by a single vomeronasal receptor.


Assuntos
Agressão , Órgão Vomeronasal , Agressão/fisiologia , Animais , Hipotálamo , Masculino , Camundongos , Neurônios/fisiologia , Feromônios/fisiologia , Núcleo Hipotalâmico Ventromedial , Órgão Vomeronasal/fisiologia
18.
Curr Biol ; 32(17): 3821-3829.e6, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-35868323

RESUMO

Pulsatile release of the hormone oxytocin (OT) mediates uterine contraction during parturition and milk ejection during lactation.1-3 These pulses are generated by the unique activity patterns of the central neuroendocrine OT neurons located in the paraventricular and supraoptic hypothalamus. Classical studies have characterized putative OT neurons by in vivo extracellular recording techniques in rats and rabbits.1,4-10 Due to technical limitations, however, the identity of OT neurons in these previous studies was speculative based on their electrophysiological characteristics and axonal projection to the posterior pituitary, not on OT gene expression. To pinpoint OT neural activities among other hypothalamic neurons that project to the pituitary11,12 and make better use of cell-type-specific neuroscience toolkits,13 a mouse model needs to be developed for the studies of parturition and lactation. We herein introduce viral genetic approaches in mice to characterize the maternal activities of OT neurons by fiber photometry. A sharp photometric peak of OT neurons appeared at approximately 520 s following simultaneous suckling stimuli from three pups. The amplitude of the peaks increased as the mother mice experienced lactation, irrespective of the age of the pups, suggesting the intrinsic plasticity of maternal OT neurons. Based on a mono-synaptic input map to OT neurons, we pharmacogenetically activated the inhibitory neurons in the bed nucleus of the stria terminalis and found the suppression of the activities of OT neurons. Collectively, our study illuminates temporal dynamics in the maternal neural activities of OT neurons and identifies one of its modulatory inputs.


Assuntos
Lactação , Ocitocina , Animais , Feminino , Hipotálamo/metabolismo , Lactação/fisiologia , Camundongos , Neurônios/fisiologia , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Gravidez , Tálamo
19.
Nat Commun ; 13(1): 556, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115521

RESUMO

The vomeronasal system plays an essential role in sensing various environmental chemical cues. Here we show that mice exposed to blood and, consequently, hemoglobin results in the activation of vomeronasal sensory neurons expressing a specific vomeronasal G protein-coupled receptor, Vmn2r88, which is mediated by the interaction site, Gly17, on hemoglobin. The hemoglobin signal reaches the medial amygdala (MeA) in both male and female mice. However, it activates the dorsal part of ventromedial hypothalamus (VMHd) only in lactating female mice. As a result, in lactating mothers, hemoglobin enhances digging and rearing behavior. Manipulation of steroidogenic factor 1 (SF1)-expressing neurons in the VMHd is sufficient to induce the hemoglobin-mediated behaviors. Our results suggest that the oxygen-carrier hemoglobin plays a role as a chemosensory signal, eliciting behavioral responses in mice in a state-dependent fashion.


Assuntos
Tonsila do Cerebelo/metabolismo , Biomarcadores/sangue , Hemoglobinas/metabolismo , Células Receptoras Sensoriais/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Órgão Vomeronasal/metabolismo , Animais , Feminino , Hemoglobinas/genética , Hibridização In Situ/métodos , Lactação , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Atividade Motora/fisiologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Globinas beta/genética , Globinas beta/metabolismo
20.
Cell Rep Methods ; 1(2): 100038, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35475238

RESUMO

Recent advancements in tissue clearing technologies have offered unparalleled opportunities for researchers to explore the whole mouse brain at cellular resolution. With the expansion of this experimental technique, however, a scalable and easy-to-use computational tool is in demand to effectively analyze and integrate whole-brain mapping datasets. To that end, here we present CUBIC-Cloud, a cloud-based framework to quantify, visualize, and integrate mouse brain data. CUBIC-Cloud is a fully automated system where users can upload their whole-brain data, run analyses, and publish the results. We demonstrate the generality of CUBIC-Cloud by a variety of applications. First, we investigated the brain-wide distribution of five cell types. Second, we quantified Aß plaque deposition in Alzheimer's disease model mouse brains. Third, we reconstructed a neuronal activity profile under LPS-induced inflammation by c-Fos immunostaining. Last, we show brain-wide connectivity mapping by pseudotyped rabies virus. Together, CUBIC-Cloud provides an integrative platform to advance scalable and collaborative whole-brain mapping.


Assuntos
Doença de Alzheimer , Encéfalo , Camundongos , Animais , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Doença de Alzheimer/diagnóstico por imagem , Neurônios
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA