Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 626(7997): 136-144, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38267578

RESUMO

Humans and animals exhibit various forms of prosocial helping behaviour towards others in need1-3. Although previous research has investigated how individuals may perceive others' states4,5, the neural mechanisms of how they respond to others' needs and goals with helping behaviour remain largely unknown. Here we show that mice engage in a form of helping behaviour towards other individuals experiencing physical pain and injury-they exhibit allolicking (social licking) behaviour specifically towards the injury site, which aids the recipients in coping with pain. Using microendoscopic imaging, we found that single-neuron and ensemble activity in the anterior cingulate cortex (ACC) encodes others' state of pain and that this representation is different from that of general stress in others. Furthermore, functional manipulations demonstrate a causal role of the ACC in bidirectionally controlling targeted allolicking. Notably, this behaviour is represented in a population code in the ACC that differs from that of general allogrooming, a distinct type of prosocial behaviour elicited by others' emotional stress. These findings advance our understanding of the neural coding and regulation of helping behaviour.


Assuntos
Comportamento Animal , Empatia , Giro do Cíngulo , Comportamento de Ajuda , Dor , Comportamento Social , Animais , Camundongos , Empatia/fisiologia , Giro do Cíngulo/citologia , Giro do Cíngulo/fisiologia , Comportamento Animal/fisiologia , Ferimentos e Lesões , Capacidades de Enfrentamento , Estresse Psicológico , Asseio Animal
3.
Horm Behav ; 153: 105391, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37301130

RESUMO

Comforting is a crucial form of prosocial behavior that is important for maintaining social unity and improving the physical and emotional well-being of social species. It is often expressed through affiliative social touch toward someone in distress, providing relief for their distressed state. In the face of increasing global distress, these actions are paramount to the continued improvement of individual welfare and the collective good. Understanding the neural mechanisms responsible for promoting actions focused on benefitting others is particularly important and timely. Here, we review prosocial comforting behavior, emphasizing synthesizing recent studies carried out using rodent models. We discuss its underlying behavioral expression and motivations, and then explore both the neurobiology of prosocial comforting in a helper animal and the neurobiology of stress relief following social touch in a recipient as part of a feedback loop interaction.


Assuntos
Comportamento Social , Tato , Animais , Motivação , Emoções , Altruísmo
4.
Front Pharmacol ; 13: 970324, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36120311

RESUMO

MAOIs, a well-established class of antidepressant that operate through the inhibition of monoamine oxidase to increase available serotonin, have recently been identified as a surprisingly effective candidate for the circumvention of tumor-induced immune suppression due to their abilities to enhance antitumor T cell activity through autocrine serotonin signaling and depolarize alternatively activated tumor-associated macrophages through a reduction in reactive oxygen species production. However, this impressive class of antidepressants-turned-cancer-drugs can induce aggressive behavioral side effects when administered in immunotherapeutic doses. In this study, we investigated the possibility of avoiding these neurological side effects while simultaneously improving antitumor activity by establishing crosslinked multilamellar liposomal vesicles (cMLVs) containing the MAOI phenelzine (PLZ). Our results showed that cMLV-PLZ treatment increases antitumor efficacy in a B16-OVA mouse melanoma model compared to treatment with free phenelzine. We also found that nanoformulation resulted in the complete elimination of MAOI-related aggression. These findings suggest a promising direction for the future of MAOIs repurposed for cancer immunotherapies.

6.
Trends Neurosci ; 45(10): 749-762, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35853793

RESUMO

The ability to behave in ways that benefit other individuals' well-being is among the most celebrated human characteristics crucial for social cohesiveness. Across mammalian species, animals display various forms of prosocial behaviors - comforting, helping, and resource sharing - to support others' emotions, goals, and/or material needs. In this review, we provide a cross-species view of the behavioral manifestations, proximate and ultimate drives, and neural mechanisms of prosocial behaviors. We summarize key findings from recent studies in humans and rodents that have shed light on the neural mechanisms underlying different processes essential for prosocial interactions, from perception and empathic sharing of others' states to prosocial decisions and actions.


Assuntos
Altruísmo , Comportamento Social , Animais , Emoções , Empatia , Humanos , Mamíferos
7.
Anal Chim Acta ; 1190: 339244, 2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-34857137

RESUMO

Salmonella contamination is a major concern in food and public health safety, and carrying out episodic monitoring of Salmonella contamination in food and water bodies is essential for safeguarding public health and the economy. Therefore, there is an urgent need to develop an easy-to-operate Salmonella-targeting point-of-care detection platform. To this end, we designed two activity-based latent ratiometric electrochemical molecular substrates, denoted as Sal-CAF and Sal-NBAF, specifically for achieving easy, rapid, and selective profiling of Salmonella esterase (a Salmonella biomarker) under physiological conditions. The octyl esters of the substrates were cleaved by the esterase and triggered the trimethyl lock to eject the electron-rich aminoferrocene derivatives (CAF and NBAF), and the corresponding electrochemical signals were tracked at the negative region (-0.08 V vs Ag/AgCl) of the voltammetric spectrum. The Sal-CAF substrate was used to determine the concentration of Salmonella in a wide dynamic range (1.03 × 105-1.1 × 1010 CFU mL-1) with a low detection limit of 39.27 × 103 CFU mL-1. The developed probes were tested against various bacteria but were only activated by live Salmonella. Furthermore, the Sal-CAF probe was used directly in quantifying spiked live Salmonella spiked in milk samples and also used to effectively monitor and quantify Salmonella production in real-time. These achievements indicated the Sal-CAF probe to be a promising platform for point-of-care Salmonella analysis.


Assuntos
Técnicas Biossensoriais , Salmonella
8.
Nature ; 599(7884): 262-267, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34646019

RESUMO

The ability to help and care for others fosters social cohesiveness and is vital to the physical and emotional well-being of social species, including humans1-3. Affiliative social touch, such as allogrooming (grooming behaviour directed towards another individual), is a major type of prosocial behaviour that provides comfort to others1-6. Affiliative touch serves to establish and strengthen social bonds between animals and can help to console distressed conspecifics. However, the neural circuits that promote prosocial affiliative touch have remained unclear. Here we show that mice exhibit affiliative allogrooming behaviour towards distressed partners, providing a consoling effect. The increase in allogrooming occurs in response to different types of stressors and can be elicited by olfactory cues from distressed individuals. Using microendoscopic calcium imaging, we find that neural activity in the medial amygdala (MeA) responds differentially to naive and distressed conspecifics and encodes allogrooming behaviour. Through intersectional functional manipulations, we establish a direct causal role of the MeA in controlling affiliative allogrooming and identify a select, tachykinin-expressing subpopulation of MeA GABAergic (γ-aminobutyric-acid-expressing) neurons that promote this behaviour through their projections to the medial preoptic area. Together, our study demonstrates that mice display prosocial comforting behaviour and reveals a neural circuit mechanism that underlies the encoding and control of affiliative touch during prosocial interactions.


Assuntos
Emoções , Comportamento Social , Estresse Psicológico , Tato/fisiologia , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Animais , Comportamento Cooperativo , Feminino , Masculino , Camundongos , Vias Neurais , Neurônios/fisiologia , Área Pré-Óptica/citologia , Área Pré-Óptica/fisiologia , Estresse Psicológico/prevenção & controle , Estresse Psicológico/psicologia
9.
J Neurosci ; 41(42): 8790-8800, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34470806

RESUMO

Social behaviors, including reproductive behaviors, often display sexual dimorphism. Lordosis, the measure of female sexual receptivity, is one of the most apparent sexually dimorphic reproductive behaviors. Lordosis is regulated by estrogen and progesterone (P4) acting within a hypothalamic-limbic circuit, consisting of the arcuate, medial preoptic, and ventromedial nuclei of the hypothalamus. Social cues are integrated into the circuit through the amygdala. The posterodorsal part of the medial amygdala (MeApd) is involved in sexually dimorphic social and reproductive behaviors, and sends projections to hypothalamic neuroendocrine regions. GABA from the MeApd appears to facilitate social behaviors, while glutamate may play the opposite role. To test these hypotheses, adult female vesicular GABA transporter (VGAT)-Cre and vesicular glutamate transporter 2 (VGluT2)-Cre mice were transfected with halorhodopsin (eNpHR)-expressing or channelrhodopsin-expressing adeno-associated viruses (AAVs), respectively, in the MeApd. The lordosis quotient (LQ) was measured following either photoinhibition of VGAT or photoexcitation of VGluT2 neurons, and brains were assessed for c-Fos immunohistochemistry (IHC). Photoinhibition of VGAT neurons in the MeApd decreased LQ, and decreased c-Fos expression within VGAT neurons, within the MeApd as a whole, and within the ventrolateral part of the ventromedial nucleus (VMHvl). Photoexcitation of VGluT2 neurons did not affect LQ, but did increase time spent self-grooming, and increased c-Fos expression within VGluT2 neurons in the MeApd. Neither condition altered c-Fos expression in the medial preoptic nucleus (MPN) or the arcuate nucleus (ARH). These data support a role for MeApd GABA in the facilitation of lordosis. Glutamate from the MeApd does not appear to be directly involved in the lordosis circuit, but appears to direct behavior away from social interactions.SIGNIFICANCE STATEMENT Lordosis, the measure of female sexual receptivity, is a sexually dimorphic behavior regulated within a hypothalamic-limbic circuit. Social cues are integrated through the amygdala, and the posterodorsal part of the medial amygdala (MeApd) is involved in sexually dimorphic social and reproductive behaviors. Photoinhibition of GABAergic neurons in the MeApd inhibited lordosis, while photoactivation of glutamate neurons had no effect on lordosis, but increased self-grooming. These data support a role for MeApd GABA in the facilitation of social behaviors and MeApd glutamate projections in anti-social interactions.


Assuntos
Complexo Nuclear Corticomedial/metabolismo , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Comportamento Social , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Animais , Feminino , Ácido Glutâmico/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Caracteres Sexuais , Comportamento Sexual Animal/fisiologia , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
10.
PLoS One ; 16(8): e0256148, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34407144

RESUMO

In females, estrogens have two main modes of action relating to gonadotropin secretion: positive feedback and negative feedback. Estrogen positive and negative feedback are controlled by different regions of the hypothalamus: the preoptic area/anterior portion (mainly the anteroventral periventricular nucleus, AVPV) of the hypothalamus is associated with estrogen positive feedback while the mediobasal hypothalamus (mainly the arcuate nucleus of the hypothalamus, ARH), is associated with estrogen negative feedback. In this study, we examined the temporal pattern of gene transcription in these two regions following estrogen treatment. Adult, ovariectomized, Long Evans rats received doses of estradiol benzoate (EB) or oil every 4 days for 3 cycles. On the last EB priming cycle, hypothalamic tissues were dissected into the AVPV+ and ARH+ at 0 hrs (baseline/oil control), 6 hrs, or 24 hrs after EB treatment. RNA was extracted and sequenced using bulk RNA sequencing. Differential gene analysis, gene ontology, and weighted correlation network analysis (WGCNA) was performed. Overall, we found that the AVPV+ and ARH+ respond differently to estradiol stimulation. In both regions, estradiol treatment resulted in more gene up-regulation than down-regulation. S100g was very strongly up-regulated by estradiol in both regions at 6 and 24 hrs after EB treatment. In the AVPV+ the highest number of differentially expressed genes occurred 24 hrs after EB. In the ARH+, the highest number of genes differentially expressed by EB occurred between 6 and 24 hrs after EB, while in the AVPV+, the fewest genes changed their expression between these time points, demonstrating a temporal difference in the way that EB regulates transcription these two areas. Several genes strongly implicated in gonadotropin release were differentially affected by estradiol including Esr1, encoding estrogen receptor-α and Kiss1, encoding kisspeptin. As an internal validation, Kiss1 was up-regulated in the AVPV+ and down-regulated in the ARH+. Gene network analysis revealed the vastly different clustering of genes modulated by estradiol in the AVPV+ compared with the ARH+. These results indicate that gene expression in these two hypothalamic regions have specific responses to estradiol in timing and direction.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo Anterior/metabolismo , Hipotálamo/metabolismo , Análise de Sequência de RNA/métodos , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo Anterior/efeitos dos fármacos , Kisspeptinas/metabolismo , Modelos Animais , Ovariectomia/métodos , Ratos , Ratos Long-Evans
11.
Genes (Basel) ; 12(7)2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34356117

RESUMO

Psychiatric disorders are complex brain disorders with a high degree of genetic heterogeneity, affecting millions of people worldwide. Despite advances in psychiatric genetics, the underlying pathogenic mechanisms of psychiatric disorders are still largely elusive, which impedes the development of novel rational therapies. There has been accumulating evidence suggesting that the genetics of complex disorders can be viewed through an omnigenic lens, which involves contextualizing genes in highly interconnected networks. Thus, applying network-based multi-omics integration methods could cast new light on the pathophysiology of psychiatric disorders. In this review, we first provide an overview of the recent advances in psychiatric genetics and highlight gaps in translating molecular associations into mechanistic insights. We then present an overview of network methodologies and review previous applications of network methods in the study of psychiatric disorders. Lastly, we describe the potential of such methodologies within a multi-tissue, multi-omics approach, and summarize the future directions in adopting diverse network approaches.


Assuntos
Redes Reguladoras de Genes/genética , Transtornos Mentais/genética , Transtornos Mentais/patologia , Genômica/métodos , Humanos , Metabolômica/métodos , Proteômica/métodos
12.
Curr Opin Neurobiol ; 68: 124-136, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33940499

RESUMO

The medial amygdala (MeA) is critical for the expression of a broad range of social behaviors, and is also connected to many other brain regions that mediate those same behaviors. Here, we summarize recent advances toward elucidating mechanisms that enable the MeA to regulate a diversity of social behaviors, and also consider what role the MeA plays within the broader network of regions that orchestrate social sensorimotor transformations. We outline the molecular, anatomical, and electrophysiological features of the MeA that segregate distinct social behaviors, propose experimental strategies to disambiguate sensory representations from behavioral function in the context of a social interaction, and consider to what extent MeA function may overlap with other regions mediating similar behaviors.


Assuntos
Tonsila do Cerebelo , Comportamento Social , Fenômenos Eletrofisiológicos
13.
Nat Neurosci ; 24(6): 831-842, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33820999

RESUMO

Social interactions and relationships are often rewarding, but the neural mechanisms through which social interaction drives positive experience remain poorly understood. In this study, we developed an automated operant conditioning system to measure social reward in mice and found that adult mice of both sexes display robust reinforcement of social interaction. Through cell-type-specific manipulations, we identified a crucial role for GABAergic neurons in the medial amygdala (MeA) in promoting the positive reinforcement of social interaction. Moreover, MeA GABAergic neurons mediate social reinforcement behavior through their projections to the medial preoptic area (MPOA) and promote dopamine release in the nucleus accumbens. Finally, activation of this MeA-to-MPOA circuit can robustly overcome avoidance behavior. Together, these findings establish the MeA as a key node for regulating social reward in both sexes, providing new insights into the regulation of social reward beyond the classic mesolimbic reward system.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Operante/fisiologia , Hipotálamo/fisiologia , Rede Nervosa/fisiologia , Recompensa , Comportamento Social , Tonsila do Cerebelo/química , Animais , Feminino , Hipotálamo/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/química , Optogenética/métodos , Reforço Psicológico
14.
J Neurosci ; 41(15): 3446-3461, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33637560

RESUMO

Trauma can cause dysfunctional fear regulation leading some people to develop disorders, such as post-traumatic stress disorder (PTSD). The amygdala regulates fear, whereas PACAP (pituitary adenylate activating peptide) and PAC1 receptors are linked to PTSD symptom severity at genetic/epigenetic levels, with a strong link in females with PTSD. We discovered a PACAPergic projection from the basomedial amygdala (BMA) to the medial intercalated cells (mICCs) in adult mice. In vivo optogenetic stimulation of this pathway increased CFOS expression in mICCs, decreased fear recall, and increased fear extinction. Selective deletion of PAC1 receptors from the mICCs in females reduced fear acquisition, but enhanced fear generalization and reduced fear extinction in males. Optogenetic stimulation of the BMA-mICC PACAPergic pathway produced EPSCs in mICC neurons, which were enhanced by the PAC1 receptor antagonist, PACAP 6-38. Our findings show that mICCs modulate contextual fear in a dynamic and sex-dependent manner via a microcircuit containing the BMA and mICCs, and in a manner that was dependent on behavioral state.SIGNIFICANCE STATEMENT Traumatic stress can affect different aspects of fear behaviors, including fear learning, generalization of learned fear to novel contexts, how the fear of the original context is recalled, and how fear is reduced over time. While the amygdala has been studied for its role in regulation of different aspects of fear, the molecular circuitry of this structure is quite complex. In addition, aspects of fear can be modulated differently in males and females. Our findings show that a specific circuitry containing the neuropeptide PACAP and its receptor, PAC1, regulates various aspects of fear, including acquisition, generalization, recall, and extinction in a sexually dimorphic manner, characterizing a novel pathway that modulates traumatic fear.


Assuntos
Tonsila do Cerebelo/fisiologia , Medo , Neurônios/fisiologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Tonsila do Cerebelo/citologia , Animais , Potenciais Pós-Sinápticos Excitadores , Extinção Psicológica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Optogenética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Fatores Sexuais
15.
Neuron ; 107(5): 941-953.e7, 2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32663438

RESUMO

A central question related to virtually all social decisions is how animals integrate sex-specific cues from conspecifics. Using microendoscopic calcium imaging in mice, we find that sex information is represented in the dorsal medial prefrontal cortex (dmPFC) across excitatory and inhibitory neurons. These cells form a distributed code that differentiates the sex of conspecifics and is strengthened with social experience. While males and females both represent sex in the dmPFC, male mice show stronger encoding of female cues, and the relative strength of these sex representations predicts sex preference behavior. Using activity-dependent optogenetic manipulations of natively active ensembles, we further show that these specific representations modulate preference behavior toward males and females. Together, these results define a functional role for native representations of sex in shaping social behavior and reveal a neural mechanism underlying male- versus female-directed sociality.


Assuntos
Comportamento Animal/fisiologia , Sinais (Psicologia) , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Caracteres Sexuais , Comportamento Social , Animais , Feminino , Masculino , Camundongos
16.
Trends Neurosci ; 43(9): 651-666, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32709376

RESUMO

Social interaction can be seen as a dynamic feedback loop that couples action, reaction, and internal cognitive processes across individual agents. A fuller understanding of the social brain requires a description of how the neural dynamics across coupled brains are linked and how they coevolve over time. We elaborate a multi-brain framework that considers social interaction as an integrated network of neural systems that dynamically shape behavior, shared cognitive states, and social relationships. We describe key findings from multi-brain experiments in humans and animal models that shed new light on the function of social circuits in health and disease. Finally, we discuss recent progress in elucidating the cellular-level mechanisms underlying inter-brain neural dynamics and outline key areas for future research.


Assuntos
Encéfalo , Interação Social , Animais , Humanos , Relações Interpessoais
17.
eNeuro ; 7(1)2020.
Artigo em Inglês | MEDLINE | ID: mdl-31941660

RESUMO

Estrogen and progesterone (P4) act in neural circuits to elicit lordosis, the stereotypical female sexual receptivity behavior. Estradiol acts through membrane receptors to rapidly activate a limbic-hypothalamic circuit consisting of the arcuate (ARH), medial preoptic (MPN), and ventromedial (VMH) nuclei of the hypothalamus. This initial activation results in a transient but necessary inhibition of lordosis, which appears to be a result of the release of ß-endorphin (ß-End) from proopiomelanocortin (POMC) terminals onto cells containing the µ-opioid receptor (MOR) in the MPN. To functionally examine the role of the MOR in the hypothalamic lordosis circuit, we transfected a channelrhodopsin (ChR2) adeno-associated virus into POMC cell bodies in the ARH and photostimulated POMC/ß-End axon terminals in the MPN in sexually receptive female Pomc-cre mice. Following estrogen and P4 priming, sexual receptivity was assessed by measuring the lordosis quotient (LQ). Following an initial trial for sexual receptivity, mice were photostimulated during behavioral testing, and brains were processed for MOR immunohistochemistry (IHC). Photostimulation decreased the LQ only in ChR2-expressing Pomc-cre mice. Furthermore, photostimulation of ChR2 in POMC/ß-End axon terminals in the MPN resulted in the internalization of MOR, indicating activation of the receptor. Our results suggest that the activation of the MOR in the MPN is sufficient to attenuate lordosis behavior in a hormone-primed, sexually receptive female mouse. These data support a central role of MOR in female sexual behavior, and provide further insight into the hypothalamus control of sexual receptivity.


Assuntos
Área Pré-Óptica , beta-Endorfina , Animais , Núcleo Arqueado do Hipotálamo , Estradiol , Feminino , Camundongos , Optogenética , Postura , Progesterona , Ratos , Ratos Long-Evans , Comportamento Sexual Animal
18.
Cell ; 178(2): 429-446.e16, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31230711

RESUMO

Social interactions involve complex decision-making tasks that are shaped by dynamic, mutual feedback between participants. An open question is whether and how emergent properties may arise across brains of socially interacting individuals to influence social decisions. By simultaneously performing microendoscopic calcium imaging in pairs of socially interacting mice, we find that animals exhibit interbrain correlations of neural activity in the prefrontal cortex that are dependent on ongoing social interaction. Activity synchrony arises from two neuronal populations that separately encode one's own behaviors and those of the social partner. Strikingly, interbrain correlations predict future social interactions as well as dominance relationships in a competitive context. Together, our study provides conclusive evidence for interbrain synchrony in rodents, uncovers how synchronization arises from activity at the single-cell level, and presents a role for interbrain neural activity coupling as a property of multi-animal systems in coordinating and sustaining social interactions between individuals.


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Animais , Sinalização do Cálcio , Comportamento Competitivo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/metabolismo , Análise de Componente Principal , Predomínio Social
19.
Cell ; 176(5): 1206-1221.e18, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30773317

RESUMO

Social behaviors, including behaviors directed toward young offspring, exhibit striking sex differences. Understanding how these sexually dimorphic behaviors are regulated at the level of circuits and transcriptomes will provide insights into neural mechanisms of sex-specific behaviors. Here, we uncover a sexually dimorphic role of the medial amygdala (MeA) in governing parental and infanticidal behaviors. Contrary to traditional views, activation of GABAergic neurons in the MeA promotes parental behavior in females, while activation of this population in males differentially promotes parental versus infanticidal behavior in an activity-level-dependent manner. Through single-cell transcriptomic analysis, we found that molecular sex differences in the MeA are specifically represented in GABAergic neurons. Collectively, these results establish crucial roles for the MeA as a key node in the neural circuitry underlying pup-directed behaviors and provide important insight into the connection between sex differences across transcriptomes, cells, and circuits in regulating sexually dimorphic behavior.


Assuntos
Complexo Nuclear Corticomedial/fisiologia , Caracteres Sexuais , Comportamento Sexual Animal/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Comportamento Animal/fisiologia , Complexo Nuclear Corticomedial/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Poder Familiar , Fatores Sexuais , Comportamento Social
20.
Neuron ; 98(1): 16-30, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29621486

RESUMO

We live in a world that is largely socially constructed, and we are constantly involved in and fundamentally influenced by a broad array of complex social interactions. Social behaviors among conspecifics, either conflictive or cooperative, are exhibited by all sexually reproducing animal species and are essential for the health, survival, and reproduction of animals. Conversely, impairment in social function is a prominent feature of several neuropsychiatric disorders, such as autism spectrum disorders and schizophrenia. Despite the importance of social behaviors, many fundamental questions remain unanswered. How is social sensory information processed and integrated in the nervous system? How are different social behavioral decisions selected and modulated in brain circuits? Here we discuss conceptual issues and recent advances in our understanding of brain regions and neural circuit mechanisms underlying the regulation of social behaviors.


Assuntos
Encéfalo/fisiologia , Rede Nervosa/fisiologia , Transtornos do Comportamento Social/fisiopatologia , Comportamento Social , Animais , Humanos , Transtornos do Comportamento Social/diagnóstico , Transtornos do Comportamento Social/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...