Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cell ; 186(6): 1099-1100, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36863338

ABSTRACT

Fighting is an intense experience not only for the executors but also for the observers. In the current issue of Cell, Yang et al. identified hypothalamic aggression mirror neurons, activated during both physical fighting and witnessing a fight, possibly representing a neural mechanism for understanding social experiences in other minds.


Subject(s)
Aggression , Mirror Neurons , Aggression/physiology
2.
Cell ; 174(2): 481-496.e19, 2018 07 12.
Article in English | MEDLINE | ID: mdl-30007419

ABSTRACT

Dopamine (DA) is a central monoamine neurotransmitter involved in many physiological and pathological processes. A longstanding yet largely unmet goal is to measure DA changes reliably and specifically with high spatiotemporal precision, particularly in animals executing complex behaviors. Here, we report the development of genetically encoded GPCR-activation-based-DA (GRABDA) sensors that enable these measurements. In response to extracellular DA, GRABDA sensors exhibit large fluorescence increases (ΔF/F0 ∼90%) with subcellular resolution, subsecond kinetics, nanomolar to submicromolar affinities, and excellent molecular specificity. GRABDA sensors can resolve a single-electrical-stimulus-evoked DA release in mouse brain slices and detect endogenous DA release in living flies, fish, and mice. In freely behaving mice, GRABDA sensors readily report optogenetically elicited nigrostriatal DA release and depict dynamic mesoaccumbens DA signaling during Pavlovian conditioning or during sexual behaviors. Thus, GRABDA sensors enable spatiotemporally precise measurements of DA dynamics in a variety of model organisms while exhibiting complex behaviors.


Subject(s)
Dopamine/analysis , Drosophila/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Behavior, Animal , Dopamine/metabolism , Female , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , Optogenetics/methods , Receptors, G-Protein-Coupled/genetics , TRPV Cation Channels/genetics , Zebrafish Proteins/genetics
3.
Nature ; 626(7998): 347-356, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267576

ABSTRACT

To survive in a complex social group, one needs to know who to approach and, more importantly, who to avoid. In mice, a single defeat causes the losing mouse to stay away from the winner for weeks1. Here through a series of functional manipulation and recording experiments, we identify oxytocin neurons in the retrochiasmatic supraoptic nucleus (SOROXT) and oxytocin-receptor-expressing cells in the anterior subdivision of the ventromedial hypothalamus, ventrolateral part (aVMHvlOXTR) as a key circuit motif for defeat-induced social avoidance. Before defeat, aVMHvlOXTR cells minimally respond to aggressor cues. During defeat, aVMHvlOXTR cells are highly activated and, with the help of an exclusive oxytocin supply from the SOR, potentiate their responses to aggressor cues. After defeat, strong aggressor-induced aVMHvlOXTR cell activation drives the animal to avoid the aggressor and minimizes future defeat. Our study uncovers a neural process that supports rapid social learning caused by defeat and highlights the importance of the brain oxytocin system in social plasticity.


Subject(s)
Aggression , Avoidance Learning , Hypothalamus , Neural Pathways , Neurons , Oxytocin , Social Learning , Animals , Mice , Aggression/physiology , Avoidance Learning/physiology , Cues , Fear/physiology , Hypothalamus/cytology , Hypothalamus/metabolism , Neural Pathways/physiology , Neurons/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Social Behavior , Social Learning/physiology , Supraoptic Nucleus/cytology , Supraoptic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism , Neuronal Plasticity
4.
Nature ; 618(7967): 1006-1016, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37286598

ABSTRACT

In many species, including mice, female animals show markedly different pup-directed behaviours based on their reproductive state1,2. Naive wild female mice often kill pups, while lactating female mice are dedicated to pup caring3,4. The neural mechanisms that mediate infanticide and its switch to maternal behaviours during motherhood remain unclear. Here, on the basis of the hypothesis that maternal and infanticidal behaviours are supported by distinct and competing neural circuits5,6, we use the medial preoptic area (MPOA), a key site for maternal behaviours7-11, as a starting point and identify three MPOA-connected brain regions that drive differential negative pup-directed behaviours. Functional manipulation and in vivo recording reveal that oestrogen receptor α (ESR1)-expressing cells in the principal nucleus of the bed nucleus of stria terminalis (BNSTprESR1) are necessary, sufficient and naturally activated during infanticide in female mice. MPOAESR1 and BNSTprESR1 neurons form reciprocal inhibition to control the balance between positive and negative infant-directed behaviours. During motherhood, MPOAESR1 and BNSTprESR1 cells change their excitability in opposite directions, supporting a marked switch of female behaviours towards the young.


Subject(s)
Infanticide , Maternal Behavior , Preoptic Area , Animals , Female , Mice , Lactation , Maternal Behavior/physiology , Neural Pathways/physiology , Preoptic Area/cytology , Preoptic Area/physiology , Thalamus/cytology , Thalamus/physiology
5.
Nat Rev Neurosci ; 23(5): 257-274, 2022 05.
Article in English | MEDLINE | ID: mdl-35361961

ABSTRACT

Neurotransmitters and neuromodulators have a wide range of key roles throughout the nervous system. However, their dynamics in both health and disease have been challenging to assess, owing to the lack of in vivo tools to track them with high spatiotemporal resolution. Thus, developing a platform that enables minimally invasive, large-scale and long-term monitoring of neurotransmitters and neuromodulators with high sensitivity, high molecular specificity and high spatiotemporal resolution has been essential. Here, we review the methods available for monitoring the dynamics of neurotransmitters and neuromodulators. Following a brief summary of non-genetically encoded methods, we focus on recent developments in genetically encoded fluorescent indicators, highlighting how these novel indicators have facilitated advances in our understanding of the functional roles of neurotransmitters and neuromodulators in the nervous system. These studies present a promising outlook for the future development and use of tools to monitor neurotransmitters and neuromodulators.


Subject(s)
Neurotransmitter Agents , Humans
6.
Nature ; 596(7873): 553-557, 2021 08.
Article in English | MEDLINE | ID: mdl-34381215

ABSTRACT

Maternal care, including by non-biological parents, is important for offspring survival1-8. Oxytocin1,2,9-15, which is released by the hypothalamic paraventricular nucleus (PVN), is a critical maternal hormone. In mice, oxytocin enables neuroplasticity in the auditory cortex for maternal recognition of pup distress15. However, it is unclear how initial parental experience promotes hypothalamic signalling and cortical plasticity for reliable maternal care. Here we continuously monitored the behaviour of female virgin mice co-housed with an experienced mother and litter. This documentary approach was synchronized with neural recordings from the virgin PVN, including oxytocin neurons. These cells were activated as virgins were enlisted in maternal care by experienced mothers, who shepherded virgins into the nest and demonstrated pup retrieval. Virgins visually observed maternal retrieval, which activated PVN oxytocin neurons and promoted alloparenting. Thus rodents can acquire maternal behaviour by social transmission, providing a mechanism for adapting the brains of adult caregivers to infant needs via endogenous oxytocin.


Subject(s)
Learning , Maternal Behavior/psychology , Mothers/psychology , Neurons/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Sexual Abstinence/psychology , Teaching , Animals , Female , Housing, Animal , Litter Size , Mice , Nesting Behavior , Neuronal Plasticity
7.
Nat Methods ; 17(11): 1156-1166, 2020 11.
Article in English | MEDLINE | ID: mdl-33087905

ABSTRACT

Dopamine (DA) plays a critical role in the brain, and the ability to directly measure dopaminergic activity is essential for understanding its physiological functions. We therefore developed red fluorescent G-protein-coupled receptor-activation-based DA (GRABDA) sensors and optimized versions of green fluorescent GRABDA sensors. In response to extracellular DA, both the red and green GRABDA sensors exhibit a large increase in fluorescence, with subcellular resolution, subsecond kinetics and nanomolar-to-submicromolar affinity. Moreover, the GRABDA sensors resolve evoked DA release in mouse brain slices, detect evoked compartmental DA release from a single neuron in live flies and report optogenetically elicited nigrostriatal DA release as well as mesoaccumbens dopaminergic activity during sexual behavior in freely behaving mice. Coexpressing red GRABDA with either green GRABDA or the calcium indicator GCaMP6s allows tracking of dopaminergic signaling and neuronal activity in distinct circuits in vivo.


Subject(s)
Biosensing Techniques/methods , Brain/metabolism , Dopamine/metabolism , Receptors, G-Protein-Coupled/metabolism , Sexual Behavior/physiology , Animals , Drosophila/genetics , Drosophila/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Neurons/metabolism , Rats , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Receptors, G-Protein-Coupled/genetics , Red Fluorescent Protein
8.
Horm Behav ; 151: 105339, 2023 05.
Article in English | MEDLINE | ID: mdl-36878049

ABSTRACT

Reproduction is the biological process by which new individuals are produced by their parents. It is the fundamental feature of all known life and is required for the existence of all species. All mammals reproduce sexually, a process that involves the union of two reproductive cells, one from a male and one from a female. Sexual behaviors are a series of actions leading to reproduction. They are composed of appetitive, action, and refractory phases, each supported by dedicated developmentally-wired neural circuits to ensure high reproduction success. In rodents, successful reproduction can only occur during female ovulation. Thus, female sexual behavior is tightly coupled with ovarian activity, namely the estrous cycle. This is achieved through the close interaction between the female sexual behavior circuit and the hypothalamic-pituitary-gonadal (HPG) axis. In this review, we will summarize our current understanding, learned mainly in rodents, regarding the neural circuits underlying each phase of the female sexual behaviors and their interaction with the HPG axis, highlighting the gaps in our knowledge that require future investigation.


Subject(s)
Ovary , Reproduction , Animals , Female , Male , Ovulation , Estrous Cycle , Mammals
9.
Trends Genet ; 34(10): 755-776, 2018 10.
Article in English | MEDLINE | ID: mdl-30173869

ABSTRACT

Aggression is a fundamental social behavior that is essential for competing for resources and protecting oneself and families in both males and females. As a result of natural selection, aggression is often displayed differentially between the sexes, typically at a higher level in males than females. Here, we highlight the behavioral differences between male and female aggression in rodents. We further outline the aggression circuits in males and females, and compare their differences at each circuit node. Lastly, we summarize our current understanding regarding the generation of sexually dimorphic aggression circuits during development and their maintenance during adulthood. In both cases, gonadal steroid hormones appear to play crucial roles in differentiating the circuits by impacting on the survival, morphology, and intrinsic properties of relevant cells. Many other factors, such as environment and experience, may also contribute to sex differences in aggression and remain to be investigated in future studies.


Subject(s)
Aggression/physiology , Brain/physiology , Selection, Genetic/genetics , Sexual Behavior, Animal/physiology , Animals , Female , Male , Selection, Genetic/physiology
10.
Nature ; 580(7802): 189-190, 2020 04.
Article in English | MEDLINE | ID: mdl-32231321
11.
Nature ; 557(7704): 172-174, 2018 05.
Article in English | MEDLINE | ID: mdl-29730673
12.
Nature ; 470(7333): 221-6, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21307935

ABSTRACT

Electrical stimulation of certain hypothalamic regions in cats and rodents can elicit attack behaviour, but the exact location of relevant cells within these regions, their requirement for naturally occurring aggression and their relationship to mating circuits have not been clear. Genetic methods for neural circuit manipulation in mice provide a potentially powerful approach to this problem, but brain-stimulation-evoked aggression has never been demonstrated in this species. Here we show that optogenetic, but not electrical, stimulation of neurons in the ventromedial hypothalamus, ventrolateral subdivision (VMHvl) causes male mice to attack both females and inanimate objects, as well as males. Pharmacogenetic silencing of VMHvl reversibly inhibits inter-male aggression. Immediate early gene analysis and single unit recordings from VMHvl during social interactions reveal overlapping but distinct neuronal subpopulations involved in fighting and mating. Neurons activated during attack are inhibited during mating, suggesting a potential neural substrate for competition between these opponent social behaviours.


Subject(s)
Aggression/physiology , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Electric Stimulation , Electrophysiology , Female , Gene Expression Regulation , Genes, fos/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neural Inhibition/genetics , Neural Inhibition/physiology , Neural Pathways/physiology , Neurons/physiology , Sexual Behavior, Animal/physiology , Ventromedial Hypothalamic Nucleus/anatomy & histology , Ventromedial Hypothalamic Nucleus/metabolism
13.
J Neurosci ; 34(17): 5971-84, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760856

ABSTRACT

The ventromedial hypothalamus, ventrolateral area (VMHvl) was identified recently as a critical locus for inter-male aggression. Optogenetic stimulation of VMHvl in male mice evokes attack toward conspecifics and inactivation of the region inhibits natural aggression, yet very little is known about its underlying neural activity. To understand its role in promoting aggression, we recorded and analyzed neural activity in the VMHvl in response to a wide range of social and nonsocial stimuli. Although response profiles of VMHvl neurons are complex and heterogeneous, we identified a subpopulation of neurons that respond maximally during investigation and attack of male conspecific mice and during investigation of a source of male mouse urine. These "male responsive" neurons in the VMHvl are tuned to both the inter-male distance and the animal's velocity during attack. Additionally, VMHvl activity predicts several parameters of future aggressive action, including the latency and duration of the next attack. Linear regression analysis further demonstrates that aggression-specific parameters, such as distance, movement velocity, and attack latency, can model ongoing VMHvl activity fluctuation during inter-male encounters. These results represent the first effort to understand the hypothalamic neural activity during social behaviors using quantitative tools and suggest an important role for the VMHvl in encoding movement, sensory, and motivation-related signals.


Subject(s)
Action Potentials/physiology , Aggression/physiology , Behavior, Animal/physiology , Neurons/physiology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Male , Mice
14.
STAR Protoc ; 5(1): 102882, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38340320

ABSTRACT

While brain regions function in coordination to mediate diverse behaviors, techniques allowing simultaneous monitoring of many deep brain regions remain limited. Here, we present a multi-fiber recording protocol that enables simultaneous recording of fluorescence signals from multiple brain regions in freely behaving mice. We describe steps for assembling a multi-fiber array and patch cord, implantation, and recording. We then detail procedures for data extraction and visualization. This protocol enables a comprehensive view of the neural activity at the network level. For complete details on the use and execution of this protocol, please refer to Guo et al.1.


Subject(s)
Brain , Neurons , Mice , Animals , Brain/physiology , Neurons/physiology , Behavior, Animal/physiology , Fluorescence
15.
J Neural Eng ; 21(3)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38861996

ABSTRACT

Objective.Distributed hypothalamic-midbrain neural circuits help orchestrate complex behavioral responses during social interactions. Given rapid advances in optical imaging, it is a fundamental question how population-averaged neural activity measured by multi-fiber photometry (MFP) for calcium fluorescence signals correlates with social behaviors is a fundamental question. This paper aims to investigate the correspondence between MFP data and social behaviors.Approach:We propose a state-space analysis framework to characterize mouse MFP data based on dynamic latent variable models, which include a continuous-state linear dynamical system and a discrete-state hidden semi-Markov model. We validate these models on extensive MFP recordings during aggressive and mating behaviors in male-male and male-female interactions, respectively.Main results:Our results show that these models are capable of capturing both temporal behavioral structure and associated neural states, and produce interpretable latent states. Our approach is also validated in computer simulations in the presence of known ground truth.Significance:Overall, these analysis approaches provide a state-space framework to examine neural dynamics underlying social behaviors and reveals mechanistic insights into the relevant networks.


Subject(s)
Photometry , Social Behavior , Animals , Mice , Photometry/methods , Male , Female , Mice, Inbred C57BL , Nerve Net/physiology , Computer Simulation , Sexual Behavior, Animal/physiology , Aggression/physiology , Models, Neurological
16.
bioRxiv ; 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38234793

ABSTRACT

Distributed hypothalamic-midbrain neural circuits orchestrate complex behavioral responses during social interactions. How population-averaged neural activity measured by multi-fiber photometry (MFP) for calcium fluorescence signals correlates with social behaviors is a fundamental question. We propose a state-space analysis framework to characterize mouse MFP data based on dynamic latent variable models, which include continuous-state linear dynamical system (LDS) and discrete-state hidden semi-Markov model (HSMM). We validate these models on extensive MFP recordings during aggressive and mating behaviors in male-male and male-female interactions, respectively. Our results show that these models are capable of capturing both temporal behavioral structure and associated neural states. Overall, these analysis approaches provide an unbiased strategy to examine neural dynamics underlying social behaviors and reveals mechanistic insights into the relevant networks.

17.
Neuron ; 112(12): 1930-1942.e6, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38547869

ABSTRACT

Norepinephrine (NE) is an essential biogenic monoamine neurotransmitter. The first-generation NE sensor makes in vivo, real-time, cell-type-specific and region-specific NE detection possible, but its low NE sensitivity limits its utility. Here, we developed the second-generation GPCR-activation-based NE sensors (GRABNE2m and GRABNE2h) with a superior response and high sensitivity and selectivity to NE both in vitro and in vivo. Notably, these sensors can detect NE release triggered by either optogenetic or behavioral stimuli in freely moving mice, producing robust signals in the locus coeruleus and hypothalamus. With the development of a novel transgenic mouse line, we recorded both NE release and calcium dynamics with dual-color fiber photometry throughout the sleep-wake cycle; moreover, dual-color mesoscopic imaging revealed cell-type-specific spatiotemporal dynamics of NE and calcium during sensory processing and locomotion. Thus, these new GRABNE sensors are valuable tools for monitoring the precise spatiotemporal release of NE in vivo, providing new insights into the physiological and pathophysiological roles of NE.


Subject(s)
Locus Coeruleus , Mice, Transgenic , Norepinephrine , Optogenetics , Animals , Norepinephrine/metabolism , Mice , Optogenetics/methods , Locus Coeruleus/metabolism , Calcium/metabolism , Wakefulness/physiology , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Hypothalamus/metabolism , Sleep/physiology , Male , Mice, Inbred C57BL , Biosensing Techniques/methods , HEK293 Cells , Photometry/methods
18.
Science ; 382(6669): 399-404, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37883550

ABSTRACT

Sexual, parental, and aggressive behaviors are central to the reproductive success of individuals and species survival and thus are supported by hardwired neural circuits. The reproductive behavior control column (RBCC), which comprises the medial preoptic nucleus (MPN), the ventrolateral part of the ventromedial hypothalamus (VMHvl), and the ventral premammillary nucleus (PMv), is essential for all social behaviors. The RBCC integrates diverse hormonal and metabolic cues and adjusts an animal's physical activity, hence the chance of social encounters. The RBCC further engages the mesolimbic dopamine system to maintain social interest and reinforces cues and actions that are time-locked with social behaviors. We propose that the RBCC and brainstem form a dual-control system for generating moment-to-moment social actions. This Review summarizes recent progress regarding the identities of RBCC cells and their pathways that drive different aspects of social behaviors.


Subject(s)
Hypothalamus , Social Behavior , Animals , Aggression/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Sexual Behavior/physiology , Male , Female , Maternal Behavior/physiology , Paternal Behavior/physiology , Consummatory Behavior
19.
Nat Neurosci ; 26(5): 774-787, 2023 05.
Article in English | MEDLINE | ID: mdl-37037956

ABSTRACT

Aggression is costly and requires tight regulation. Here we identify the projection from estrogen receptor alpha-expressing cells in the caudal part of the medial preoptic area (cMPOAEsr1) to the ventrolateral part of the ventromedial hypothalamus (VMHvl) as an essential pathway for modulating aggression in male mice. cMPOAEsr1 cells increase activity mainly during male-male interaction, which differs from the female-biased response pattern of rostral MPOAEsr1 (rMPOAEsr1) cells. Notably, cMPOAEsr1 cell responses to male opponents correlated with the opponents' fighting capability, which mice could estimate based on physical traits or learn through physical combats. Inactivating the cMPOAEsr1-VMHvl pathway increased aggression, whereas activating the pathway suppressed natural intermale aggression. Thus, cMPOAEsr1 is a key population for encoding opponents' fighting capability-information that could be used to prevent animals from engaging in disadvantageous conflicts with superior opponents by suppressing the activity of VMHvl cells essential for attack behaviors.


Subject(s)
Aggression , Hypothalamus , Mice , Male , Female , Animals , Aggression/physiology , Hypothalamus/physiology , Preoptic Area , Learning
20.
Nat Neurosci ; 26(12): 2131-2146, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37946049

ABSTRACT

Social behaviors are innate and supported by dedicated neural circuits, but the molecular identities of these circuits and how they are established developmentally and shaped by experience remain unclear. Here we show that medial amygdala (MeA) cells originating from two embryonically parcellated developmental lineages have distinct response patterns and functions in social behavior in male mice. MeA cells expressing the transcription factor Foxp2 (MeAFoxp2) are specialized for processing male conspecific cues and are essential for adult inter-male aggression. By contrast, MeA cells derived from the Dbx1 lineage (MeADbx1) respond broadly to social cues, respond strongly during ejaculation and are not essential for male aggression. Furthermore, MeAFoxp2 and MeADbx1 cells show differential anatomical and functional connectivity. Altogether, our results suggest a developmentally hardwired aggression circuit at the MeA level and a lineage-based circuit organization by which a cell's embryonic transcription factor profile determines its social information representation and behavioral relevance during adulthood.


Subject(s)
Corticomedial Nuclear Complex , Neurons , Male , Mice , Animals , Neurons/physiology , Social Behavior , Amygdala/physiology , Transcription Factors/genetics , Homeodomain Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL