Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 348
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Nat Neurosci ; 27(4): 702-715, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38347201

ABSTRACT

Social behaviors often consist of a motivational phase followed by action. Here we show that neurons in the ventromedial hypothalamus ventrolateral area (VMHvl) of mice encode the temporal sequence of aggressive motivation to action. The VMHvl receives local inhibitory input (VMHvl shell) and long-range input from the medial preoptic area (MPO) with functional coupling to neurons with specific temporal profiles. Encoding models reveal that during aggression, VMHvl shellvgat+ activity peaks at the start of an attack, whereas activity from the MPO-VMHvlvgat+ input peaks at specific interaction endpoints. Activation of the MPO-VMHvlvgat+ input promotes and prolongs a low motivation state, whereas activation of VMHvl shellvgat+ results in action-related deficits, acutely terminating attack. Moreover, stimulation of MPO-VMHvlvgat+ input is positively valenced and anxiolytic. Together, these data demonstrate how distinct inhibitory inputs to the hypothalamus can independently gate the motivational and action phases of aggression through a single locus of control.


Subject(s)
Aggression , Motivation , Mice , Animals , Aggression/physiology , Social Behavior , Hypothalamus/physiology , Neurons/physiology
2.
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
3.
Horm Behav ; 157: 105452, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37977023

ABSTRACT

Social hierarchies are a prevalent feature of all animal groups, and an individual's rank within the group can significantly affect their overall health, typically at the greatest expense of the lowest-ranked individuals, or omegas. These subjects have been shown to exhibit various stress-related phenotypes, such as increased hypothalamic-pituitary axis activity and increased amygdalar corticotropin-releasing factor levels compared to higher-ranked subjects. However, these findings have been primarily characterized in males and in models requiring exhibition of severe aggression. The goals of the current study, therefore, were to characterize the formation and maintenance of social hierarchies using the tube test and palatable liquid competition in same-sex groups of male and female C57BL/6 J mice. We also aimed to examine the effects of tube test-determined social rank on plasma and hypothalamic oxytocin and vasopressin levels, peptides with established roles in social behaviors and the stress response. Lastly, we assessed the effects of environmental enrichment and length of testing on the measures outlined above. Overall, we demonstrated that males and females develop social hierarchies and that these hierarchies can be determined using the tube test. While we were unable to establish a consistent connection between peptide levels and social rank, we observed transient changes in these peptides reflecting complex interactions between social rank, sex, environment, and length of testing. We also found that many male and female omegas began to exhibit passive coping behavior after repeated tube test losses, demonstrating the potential of this assay to serve as a model of chronic, mild psychosocial stress.


Subject(s)
Hierarchy, Social , Social Behavior , Humans , Animals , Mice , Male , Female , Mice, Inbred C57BL , Aggression/physiology , Hypothalamus
4.
Mol Cell Endocrinol ; 580: 112101, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37923055

ABSTRACT

Terrestrial vertebrates have a population of androgen-dependent vasotocin (VT)-expressing neurons in the extended amygdala that are more abundant in males and mediate male-typical social behaviors, including aggression. Teleosts lack these neurons but instead have novel male-specific VT-expressing neurons in the tuberal hypothalamus. Here we found in medaka that vt expression in these neurons is dependent on post-pubertal gonadal androgens and that androgens can act on these neurons to directly stimulate vt transcription via the androgen receptor subtype Ara. Furthermore, administration of exogenous VT induced aggression in females and alterations in the androgen milieu led to correlated changes in the levels of tuberal hypothalamic vt expression and aggression in both sexes. However, genetic ablation of vt failed to prevent androgen-induced aggression in females. Collectively, our results demonstrate a marked androgen dependence of male-specific vt expression in the teleost tuberal hypothalamus, although its relevance to male-typical aggression needs to be further validated.


Subject(s)
Aggression , Oryzias , Animals , Female , Male , Aggression/physiology , Androgens/pharmacology , Androgens/metabolism , Sexual Behavior, Animal/physiology , Vasotocin/metabolism , Oryzias/metabolism , Hypothalamus/metabolism
5.
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
6.
J Comp Neurol ; 531(15): 1550-1561, 2023 10.
Article in English | MEDLINE | ID: mdl-37496437

ABSTRACT

The transient receptor potential cation channel 2 (TRPC2) conveys pheromonal information from the vomeronasal organ (VNO) to the brain. Both male and female mice lacking this gene show altered sex-typical behavior as adults. We asked whether TRPC2, highly expressed in the VNO, normally participates in the development of VNO-recipient brain regions controlling mounting and aggression, two behaviors affected by TRPC2 loss. We now report significant effects of TRPC2 loss in both the posterodorsal aspect of the medial amygdala (MePD) and ventromedial nucleus of the hypothalamus (VMH) of male and female mice. In the MePD, a sex difference in neuron number was eliminated by the TRPC2 knockout (KO), but the effect was complex, with fewer neurons in the right MePD of females, and fewer neurons in the left MePD of males. In contrast, MePD astrocytes were unaffected by the KO. In the ventrolateral (vl) aspect of the VMH, KO females were like wildtype (WT) females, but TRPC2 loss had a dramatic effect in males, with fewer neurons than WT males and a smaller VMHvl overall. We also discovered a glial sex difference in VMHvl of WTs, with females having more astrocytes than males. Interestingly, TRPC2 loss increased astrocyte number in males in this region. We conclude that TRPC2 normally participates in the sexual differentiation of the mouse MePD and VMHvl. These changes in two key VNO-recipient regions may underlie the effects of the TRPC2 KO on behavior.


Subject(s)
Sex Characteristics , Social Behavior , Animals , Female , Male , Mice , Aggression/physiology , Hypothalamus , Neuroglia
7.
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
8.
Curr Biol ; 33(8): R313-R316, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37098336

ABSTRACT

Mirror neurons have been found mainly in neocortical structures of primates and rodents; however, their functions are still debated. A new study has discovered mirror neurons for aggressive behaviors in the ventromedial hypothalamus of mice, an evolutionarily ancient structure, highlighting a new function key for survival.


Subject(s)
Mirror Neurons , Mice , Animals , Aggression/physiology , Hypothalamus/physiology
9.
CNS Neurosci Ther ; 29(7): 2010-2017, 2023 07.
Article in English | MEDLINE | ID: mdl-36890650

ABSTRACT

INTRODUCTION: Deep brain stimulation (DBS) is a surgical technique used to manage aggression in patients who do not improve despite the use of appropriate drug treatment. OBJECTIVE: The objective of this study is to assess the impact of DBS on aggressive behavior refractory to the pharmacological and behavioral treatment of patients with Intellectual Disabilities (ID). METHODS: A follow-up was conducted on a cohort of 12 patients with severe ID, undergoing DBS in posteromedial hypothalamic nuclei; evaluated with the Overt Aggression Scale (OAS), before the intervention, at 6, 12, and 18 months of medical follow-up. RESULTS: After the surgical procedure, there was a significant reduction in the aggressiveness of patients in the follow-up medical evaluation at 6 months (t = 10.14; p < 0.01), 12 months (t = 14.06; p < 0.01), and 18 months (t = 15.34; p < 0.01), respect to the initial measurement; with a very large effect size (6 months: d = 2.71; 12 months: d = 3.75; 18 months: d = 4.10). From 12 months onward, emotional control stabilized and is sustained at 18 months (t = 1.24; p > 0.05). CONCLUSION: DBS in posteromedial hypothalamic nuclei may be an effective treatment for the management of aggression in patients with ID refractory to pharmacological treatment.


Subject(s)
Deep Brain Stimulation , Intellectual Disability , Humans , Child , Intellectual Disability/therapy , Deep Brain Stimulation/methods , Hypothalamus , Treatment Outcome , Aggression/physiology , Aggression/psychology
10.
Cell ; 186(6): 1195-1211.e19, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36796363

ABSTRACT

Social interactions require awareness and understanding of the behavior of others. Mirror neurons, cells representing an action by self and others, have been proposed to be integral to the cognitive substrates that enable such awareness and understanding. Mirror neurons of the primate neocortex represent skilled motor tasks, but it is unclear if they are critical for the actions they embody, enable social behaviors, or exist in non-cortical regions. We demonstrate that the activity of individual VMHvlPR neurons in the mouse hypothalamus represents aggression performed by self and others. We used a genetically encoded mirror-TRAP strategy to functionally interrogate these aggression-mirroring neurons. We find that their activity is essential for fighting and that forced activation of these cells triggers aggressive displays by mice, even toward their mirror image. Together, we have discovered a mirroring center in an evolutionarily ancient region that provides a subcortical cognitive substrate essential for a social behavior.


Subject(s)
Aggression , Hypothalamus , Mirror Neurons , Animals , Mice , Aggression/physiology , Hypothalamus/cytology , Social Behavior
11.
Cell ; 186(1): 178-193.e15, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36608653

ABSTRACT

The hypothalamus regulates innate social behaviors, including mating and aggression. These behaviors can be evoked by optogenetic stimulation of specific neuronal subpopulations within MPOA and VMHvl, respectively. Here, we perform dynamical systems modeling of population neuronal activity in these nuclei during social behaviors. In VMHvl, unsupervised analysis identified a dominant dimension of neural activity with a large time constant (>50 s), generating an approximate line attractor in neural state space. Progression of the neural trajectory along this attractor was correlated with an escalation of agonistic behavior, suggesting that it may encode a scalable state of aggressiveness. Consistent with this, individual differences in the magnitude of the integration dimension time constant were strongly correlated with differences in aggressiveness. In contrast, approximate line attractors were not observed in MPOA during mating; instead, neurons with fast dynamics were tuned to specific actions. Thus, different hypothalamic nuclei employ distinct neural population codes to represent similar social behaviors.


Subject(s)
Sexual Behavior, Animal , Ventromedial Hypothalamic Nucleus , Animals , Sexual Behavior, Animal/physiology , Ventromedial Hypothalamic Nucleus/physiology , Hypothalamus/physiology , Aggression/physiology , Social Behavior
12.
Science ; 378(6619): 484-485, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36378988

ABSTRACT

An emergent encoding of aggressive motivation in neurons of the hypothalamus.


Subject(s)
Aggression , Hypothalamus , Motivation , Neurons , Hypothalamus/physiology , Neurons/physiology , Aggression/physiology , Birds
13.
Nature ; 608(7924): 741-749, 2022 08.
Article in English | MEDLINE | ID: mdl-35922505

ABSTRACT

Mating and aggression are innate social behaviours that are controlled by subcortical circuits in the extended amygdala and hypothalamus1-4. The bed nucleus of the stria terminalis (BNSTpr) is a node that receives input encoding sex-specific olfactory cues from the medial amygdala5,6, and which in turn projects to hypothalamic nuclei that control mating7-9 (medial preoptic area (MPOA)) and aggression9-14 (ventromedial hypothalamus, ventrolateral subdivision (VMHvl)), respectively15. Previous studies have demonstrated that male aromatase-positive BNSTpr neurons are required for mounting and attack, and may identify conspecific sex according to their overall level of activity16. However, neural representations in BNSTpr, their function and their transformations in the hypothalamus have not been characterized. Here we performed calcium imaging17,18 of male BNSTprEsr1 neurons during social behaviours. We identify distinct populations of female- versus male-tuned neurons in BNSTpr, with the former outnumbering the latter by around two to one, similar to the medial amygdala and MPOA but opposite to VMHvl, in which male-tuned neurons predominate6,9,19. Chemogenetic silencing of BNSTprEsr1 neurons while imaging MPOAEsr1 or VMHvlEsr1 neurons in behaving animals showed, unexpectedly, that the male-dominant sex-tuning bias in VMHvl was inverted to female-dominant whereas a switch from sniff- to mount-selective neurons during mating was attenuated in MPOA. Our data also indicate that BNSTprEsr1 neurons are not essential for conspecific sex identification. Rather, they control the transition from appetitive to consummatory phases of male social behaviours by shaping sex- and behaviour-specific neural representations in the hypothalamus.


Subject(s)
Sexual Behavior, Animal , Social Behavior , Aggression/physiology , Amygdala/cytology , Amygdala/physiology , Animals , Calcium/analysis , Calcium/metabolism , Female , Hypothalamus/cytology , Hypothalamus/physiology , Male , Neurons/physiology , Preoptic Area/cytology , Preoptic Area/physiology , Sex Characteristics , Sexual Behavior, Animal/physiology
14.
Neuron ; 110(18): 3000-3017.e8, 2022 09 21.
Article in English | MEDLINE | ID: mdl-35896109

ABSTRACT

Sexual behavior is fundamental for the survival of mammalian species and thus supported by dedicated neural substrates. The ventrolateral part of ventromedial hypothalamus (VMHvl) is an essential locus for controlling female sexual behaviors, but recent studies revealed the molecular complexity and functional heterogeneity of VMHvl cells. Here, we identify the cholecystokinin A receptor (Cckar)-expressing cells in the lateral VMHvl (VMHvllCckar) as the key controllers of female sexual behaviors. The inactivation of VMHvllCckar cells in female mice diminishes their interest in males and sexual receptivity, whereas activating these cells has the opposite effects. Female sexual behaviors vary drastically over the reproductive cycle. In vivo recordings reveal reproductive-state-dependent changes in VMHvllCckar cell spontaneous activity and responsivity, with the highest activity occurring during estrus. These in vivo response changes coincide with robust alternation in VMHvllCckar cell excitability and synaptic inputs. Altogether, VMHvllCckar cells represent a key neural population dynamically controlling female sexual behaviors over the reproductive cycle.


Subject(s)
Aggression , Hypothalamus , Aggression/physiology , Animals , Female , Hypothalamus/physiology , Male , Mammals , Mice , Receptor, Cholecystokinin A , Sexual Behavior, Animal/physiology
15.
Neuron ; 110(15): 2455-2469.e8, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35654036

ABSTRACT

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.


Subject(s)
Aggression , Vomeronasal Organ , Aggression/physiology , Animals , Hypothalamus , Male , Mice , Neurons/physiology , Pheromones/physiology , Ventromedial Hypothalamic Nucleus , Vomeronasal Organ/physiology
16.
Neuron ; 110(12): 2009-2023.e5, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35443152

ABSTRACT

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.


Subject(s)
Aggression , Oxytocin , Aggression/physiology , Animals , Humans , Hypothalamus/physiology , Male , Mice , Neurons/physiology , Parents
17.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35131854

ABSTRACT

Aggressive behavior is rarely observed in virgin female mice but is specifically triggered in lactation where it facilitates protection of offspring. Recent studies demonstrated that the hypothalamic ventromedial nucleus (VMN) plays an important role in facilitating aggressive behavior in both sexes. Here, we demonstrate a role for the pituitary hormone, prolactin, acting through the prolactin receptor in the VMN to control the intensity of aggressive behavior exclusively during lactation. Prolactin receptor deletion from glutamatergic neurons or specifically from the VMN resulted in hyperaggressive lactating females, with a marked shift from intruder-directed investigative behavior to very high levels of aggressive behavior. Prolactin-sensitive neurons in the VMN project to a wide range of other hypothalamic and extrahypothalamic regions, including the medial preoptic area, paraventricular nucleus, and bed nucleus of the stria terminalis, all regions known to be part of a complex neuronal network controlling maternal behavior. Within this network, prolactin acts in the VMN to specifically restrain male-directed aggressive behavior in lactating females. This action in the VMN may complement the role of prolactin in other brain regions, by shifting the balance of maternal behaviors from defense-related activities to more pup-directed behaviors necessary for nurturing offspring.


Subject(s)
Aggression/physiology , Lactation/metabolism , Prolactin/metabolism , Animals , Female , Hypothalamus/metabolism , Male , Maternal Behavior/physiology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Preoptic Area/metabolism , Receptors, Prolactin/metabolism , Thalamus/metabolism , Ventromedial Hypothalamic Nucleus/metabolism
18.
Neuron ; 110(5): 841-856.e6, 2022 03 02.
Article in English | MEDLINE | ID: mdl-34982958

ABSTRACT

Female mice exhibit opposing social behaviors toward males depending on their reproductive state: virgins display sexual receptivity (lordosis behavior), while lactating mothers attack. How a change in reproductive state produces a qualitative switch in behavioral response to the same conspecific stimulus is unknown. Using single-cell RNA-seq, we identify two distinct subtypes of estrogen receptor-1-positive neurons in the ventrolateral subdivision of the female ventromedial hypothalamus (VMHvl) and demonstrate that they causally control sexual receptivity and aggressiveness in virgins and lactating mothers, respectively. Between- and within-subject bulk-calcium recordings from each subtype reveal that aggression-specific cells acquire an increased responsiveness to social cues during the transition from virginity to maternity, while the responsiveness of the mating-specific population appears unchanged. These results demonstrate that reproductive-state-dependent changes in the relative activity of transcriptomically distinct neural subtypes can underlie categorical switches in behavior associated with physiological state changes.


Subject(s)
Lactation , Sexual Behavior, Animal , Aggression/physiology , Animals , Female , Humans , Hypothalamus/physiology , Male , Mice , Pregnancy , Sexual Behavior, Animal/physiology , Social Behavior
19.
Behav Brain Res ; 412: 113432, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34186145

ABSTRACT

The environmental context during gestation may modulate the postpartum variations in maternal behaviors observed within different animal species. Most of our experimental knowledge on this phenomenon and its physiological effects have been gained by confronting the pregnant mother with stressful situations, with the consensual results indicating a reduced maternal behavior and a hyper reactivity of stress-related neural paths. Here, in contrast, by exposing nulliparous rats strictly during pregnancy to a standard laboratory environment (STD) or a highly stimulating sensory and social environment (EE), we investigated the hypothesis that subjects frequently exposed to social stimuli and novel situations during pregnancy will show postpartum changes in subcortical brain areas' activity related to the processing of social stimuli and novelty, such that there will be modifications in maternal behavior. We found that EE mothers doubled the levels of licking and grooming, and active hovering over pups during the first postpartum week than STD dams, without a difference in the time of contact with the pups. Associated with these behaviors, EE dams showed increased c-Fos immunoreaction in hypothalamic nuclei and distinct responses in amygdalar nuclei, than STD dams. In the maternal defensive test, EE dams tripled the levels of aggressive behaviors of the STD rats. Additionally, in two different tests, EE mothers showed lower levels of postpartum anxiety-like behaviors when confronted with novel situations. Our results demonstrate that the activity of brain areas related to social behavior is adaptable by environmental circumstances experienced during gestation, presumably to prepare the progeny for these particular conditions.


Subject(s)
Maternal Behavior/physiology , Pregnancy/metabolism , Social Environment , Aggression/physiology , Amygdala/metabolism , Animals , Anxiety/physiopathology , Behavior, Animal/physiology , Brain/metabolism , Environment , Exploratory Behavior/physiology , Female , Hypothalamus/metabolism , Lactation/physiology , Male , Maternal Behavior/psychology , Postpartum Period/physiology , Postpartum Period/psychology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Social Behavior , Stress, Psychological/metabolism
20.
J Integr Neurosci ; 20(1): 77-85, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33834693

ABSTRACT

The social behavior mechanisms have not been thoroughly reported in the solitary female striped dwarf hamster (Cricetulus barabensis). In this study, the handling bag test and neutral arena measurements were used to detect the changes of aggression in the face of rivals of different genders of wild striped dwarf hamsters. We found that female hamsters had the highest aggressive performance in proestrus, followed by estrus, and the lowest in metestrus and the dioestrus, and the increased aggression during the proestrus or estrus period was low-intensity aggression such as intimidation, shock, boxing and counterattack, or even ritualized non-harmful behaviors to drive away opponents. When confronted with male individuals, aggression in females decreased significantly during estrus. The concentration of plasma estradiol was the highest in estrus and the lowest in metestrus and dioestrus. In contrast, estrogen receptor 2 relative expression in the hypothalamus is the lowest in proestrus and highest in metestrus and dioestrus. Besides, both estradiol levels in plasma and estrogen receptor 2 mRNA in the hypothalamus were associated with aggression. These results will broaden our understanding of the molecular mechanism of how breeding phenotype is an essential driver in changing the social behavior of female Cricetulus barabensis.


Subject(s)
Aggression/physiology , Behavior, Animal/physiology , Estradiol/blood , Estrogen Receptor beta/metabolism , Estrous Cycle/physiology , Hypothalamus/metabolism , Social Behavior , Animals , Cricetinae , Estrous Cycle/metabolism , Female , Male
SELECTION OF CITATIONS
SEARCH DETAIL