Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Physiol Behav ; 275: 114451, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38176291

ABSTRACT

Early exposure of does to sexually active bucks triggers early puberty onset correlating with neuroendocrine changes. However, the sensory pathways that are stimulated by the male are still unknown. Here, we assessed whether responses to olfactory stimuli are modulated by social experience (exposure to males or not) and/or endocrine status (prepubescent or pubescent). We used a calcium imaging approach on goat sensory cells from the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). For both cell types, we observed robust responses to active male hair in females under three physiological conditions: prepubescent females isolated from males (ISOL PrePub), pubescent females exposed to males (INT Pub) and isolated females (ISOL Pub). Response analysis showed overall greater proportion of responses to buck hair in ISOL PrePub. We hypothesized that females would be more responsive to active buck hair during the prepubertal period, with numerous responses perhaps originating from immature neurons. We also observed a greater proportion of mature olfactory neurons in the MOE and VNO of INT Pub females suggesting that male exposure can induce plastic changes on olfactory cell function and organization. To determine whether stimulation by male odor can advance puberty, we exposed prepubescent does to active buck hair (ODOR). In both ODOR and females isolated from males (ISOL) groups, puberty was reached one month after females exposed to intact bucks (INT), suggesting that olfactory stimulation is not sufficient to trigger puberty.


Subject(s)
Ovulation , Sexual Behavior, Animal , Animals , Female , Male , Sexual Behavior, Animal/physiology , Seasons , Ovulation/physiology , Smell , Goats/physiology
2.
Article in English | MEDLINE | ID: mdl-37690081

ABSTRACT

In mammals, especially rodents, social behaviours, such as parenting, territoriality or mate attraction, are largely based on olfactory communication through chemosignals. These behaviours are mediated by species-specific chemosignals, including small organic molecules and proteins that are secreted in the urine or in various fluids from exocrine glands. Chemosignal detection is mainly ensured by olfactory neurons in two specific sensory organs, the vomeronasal organ (VNO) and the main olfactory epithelium (MOE). This study aimed to characterise the olfactory communication in the fossorial ecotype of the water voles, Arvicola terrestris. We first measured the olfactory investigation of urine and lateral scent gland secretions from conspecifics. Our results showed that water voles can discriminate the sex of conspecifics based on the smell of urine, and that urinary male odour is attractive for female voles. Then, we demonstrated the ability of the VNO and MOE to detect volatile organic compounds (VOCs) found in water vole secretions using live-cell calcium imaging in dissociated cells. Finally, we evaluated the attractiveness of two mixtures of VOCs from urine or lateral scent glands in the field during a cyclical outbreak of vole populations.

3.
BMC Biol ; 21(1): 152, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37424020

ABSTRACT

BACKGROUND: Rodents utilize chemical cues to recognize and avoid other conspecifics infected with pathogens. Infection with pathogens and acute inflammation alter the repertoire and signature of olfactory stimuli emitted by a sick individual. These cues are recognized by healthy conspecifics via the vomeronasal or accessory olfactory system, triggering an innate form of avoidance behavior. However, the molecular identity of the sensory neurons and the higher neural circuits involved in the detection of sick conspecifics remain poorly understood. RESULTS: We employed mice that are in an acute state of inflammation induced by systemic administration of lipopolysaccharide (LPS). Through conditional knockout of the G-protein Gαi2 and deletion of other key sensory transduction molecules (Trpc2 and a cluster of 16 vomeronasal type 1 receptors), in combination with behavioral testing, subcellular Ca2+ imaging, and pS6 and c-Fos neuronal activity mapping in freely behaving mice, we show that the Gαi2+ vomeronasal subsystem is required for the detection and avoidance of LPS-treated mice. The active components underlying this avoidance are contained in urine whereas feces extract and two selected bile acids, although detected in a Gαi2-dependent manner, failed to evoke avoidance behavior. Our analyses of dendritic Ca2+ responses in vomeronasal sensory neurons provide insight into the discrimination capabilities of these neurons for urine fractions from LPS-treated mice, and how this discrimination depends on Gαi2. We observed Gαi2-dependent stimulation of multiple brain areas including medial amygdala, ventromedial hypothalamus, and periaqueductal grey. We also identified the lateral habenula, a brain region implicated in negative reward prediction in aversive learning, as a previously unknown target involved in these tasks. CONCLUSIONS: Our physiological and behavioral analyses indicate that the sensing and avoidance of LPS-treated sick conspecifics depend on the Gαi2 vomeronasal subsystem. Our observations point to a central role of brain circuits downstream of the olfactory periphery and in the lateral habenula in the detection and avoidance of sick conspecifics, providing new insights into the neural substrates and circuit logic of the sensing of inflammation in mice.


Subject(s)
Vomeronasal Organ , Mice , Animals , Vomeronasal Organ/physiology , Lipopolysaccharides , Brain , Sensory Receptor Cells , Inflammation
4.
Commun Biol ; 5(1): 383, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35444217

ABSTRACT

Estrogen receptor (ER) α is involved in several estrogen-modulated neural and peripheral functions. To determine its role in the expression of female and male reproductive behavior, a mouse line lacking the ERα in the nervous system was generated. Mutant females did not exhibit sexual behavior despite normal olfactory preference, and had a reduced number of progesterone receptor-immunoreactive neurons in the ventromedial hypothalamus. Mutant males displayed a moderately impaired sexual behavior and unaffected fertility, despite evidences of altered organization of sexually dimorphic populations in the preoptic area. In comparison, males deleted for both neural ERα and androgen receptor (AR) displayed greater sexual deficiencies. Thus, these data highlight a predominant role for neural ERα in females and a complementary role with the AR in males in the regulation of sexual behavior, and provide a solid background for future analyses of neuronal versus glial implication of these signaling pathways in both sexes.


Subject(s)
Estrogen Receptor alpha/metabolism , Sexual Behavior, Animal , Animals , Estrogen Receptor alpha/genetics , Female , Hypothalamus/metabolism , Male , Mice , Neurons/metabolism , Preoptic Area/metabolism
5.
J Exp Biol ; 224(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34494651

ABSTRACT

Mammals living at temperate latitudes typically display annual cyclicity in their reproductive activity: births are synchronized when environmental conditions are most favorable. In a majority of these species, day length is the main proximate factor used to anticipate seasonal changes and to adapt physiology. The brain integrates this photoperiodic signal through key hypothalamic structures, which regulate the reproductive axis. In this context, our study aimed to characterize regulations that occur along the hypothalamo-pituitary-gonadal (HPG) axis in male fossorial water voles (Arvicola terrestris, also known as Arvicola amphibius) throughout the year and to further probe the implication of photoperiod in these seasonal regulations. Our monthly field monitoring showed dramatic seasonal changes in the morphology and activity of reproductive organs, as well as in the androgen-dependent lateral scent glands. Moreover, our data uncovered seasonal variations at the hypothalamic level. During the breeding season, kisspeptin expression in the arcuate nucleus (ARC) decreases, while RFRP3 expression in the dorsomedial hypothalamic nucleus (DMH) increases. Our follow-up laboratory study revealed activation of the reproductive axis and confirmed a decrease in kisspeptin expression in males exposed to a long photoperiod (summer condition) compared with those maintained under a short photoperiod (winter condition) that retain all features reminiscent of sexual inhibition. Altogether, our study characterizes neuroendocrine and anatomical markers of seasonal reproductive rhythmicity in male water voles and further suggests that these seasonal changes are strongly impacted by photoperiod.


Subject(s)
Arvicolinae , Photoperiod , Animals , Hypothalamus , Male , Reproduction , Seasons
6.
Front Cell Neurosci ; 15: 638800, 2021.
Article in English | MEDLINE | ID: mdl-33679330

ABSTRACT

In mice, social behaviors are largely controlled by the olfactory system. Pheromone detection induces naïve virgin females to retrieve isolated pups to the nest and to be sexually receptive to males, but social experience increases the performance of both types of innate behaviors. Whether animals are intrinsically sensitive to the smell of conspecifics, or the detection of olfactory cues modulates experience for the display of social responses is currently unclear. Here, we employed mice with an olfactory-specific deletion of the G protein Gαi2, which partially eliminates sensory function in the vomeronasal organ (VNO), to show that social behavior in female mice results from interactions between intrinsic mechanisms in the vomeronasal system and experience-dependent plasticity. In pup- and sexually-naïve females, Gαi2 deletion elicited a reduction in pup retrieval behavior, but not in sexual receptivity. By contrast, experienced animals showed normal maternal behavior, but the experience-dependent increase in sexual receptivity was incomplete. Further, lower receptivity was accompanied by reduced neuronal activity in the anterior accessory olfactory bulb and the rostral periventricular area of the third ventricle. Therefore, neural mechanisms utilize intrinsic sensitivity in the mouse vomeronasal system and enable plasticity to display consistent social behavior.

7.
Sci Rep ; 10(1): 6242, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32277160

ABSTRACT

Estradiol derived from neural aromatization of testosterone plays a key role in the organization and activation of neural structures underlying male behaviors. This study evaluated the contribution of the estrogen receptor (ER) ß in estradiol-induced modulation of social and mood-related behaviors by using mice lacking the ERß gene in the nervous system. Mutant males exhibited reduced social interaction with same-sex congeners and impaired aggressive behavior. They also displayed increased locomotor activity, and reduced or unaffected anxiety-state level in three paradigms. However, when mice were exposed to unescapable stress in the forced swim and tail suspension tests, they spent more time immobile and a reduced time in swimming and climbing. These behavioral alterations were associated with unaffected circadian and restraint stress-induced corticosterone levels, and unchanged number of tryptophan hydroxylase 2-immunoreactive neurons in the dorsal raphe. By contrast, reduced mRNA levels of oxytocin and arginine-vasopressin were observed in the bed nucleus of stria terminalis, whereas no changes were detected in the hypothalamic paraventricular nucleus. The neural ERß is thus involved to different extent levels in social and mood-related behaviors, with a particular action on oxytocin and arginine-vasopressin signaling pathways of the bed nucleus of stria terminalis, yet the involvement of other brain areas cannot be excluded.


Subject(s)
Affect/physiology , Aggression/physiology , Anxiety/genetics , Estrogen Receptor beta/deficiency , Animals , Anxiety/psychology , Arginine Vasopressin/metabolism , Behavior, Animal/physiology , Disease Models, Animal , Estradiol/metabolism , Estrogen Receptor beta/genetics , Humans , Locomotion/genetics , Male , Mice , Mice, Knockout , Mutation , Neurons/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/physiology , Septal Nuclei/cytology , Septal Nuclei/physiology , Signal Transduction/physiology , Testosterone/metabolism
8.
Proc Natl Acad Sci U S A ; 116(11): 5135-5143, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30804203

ABSTRACT

Aggression is controlled by the olfactory system in many animal species. In male mice, territorial and infant-directed aggression are tightly regulated by the vomeronasal organ (VNO), but how diverse subsets of sensory neurons convey pheromonal information to limbic centers is not yet known. Here, we employ genetic strategies to show that mouse vomeronasal sensory neurons expressing the G protein subunit Gαi2 regulate male-male and infant-directed aggression through distinct circuit mechanisms. Conditional ablation of Gαi2 enhances male-male aggression and increases neural activity in the medial amygdala (MeA), bed nucleus of the stria terminalis, and lateral septum. By contrast, conditional Gαi2 ablation causes reduced infant-directed aggression and decreased activity in MeA neurons during male-infant interactions. Strikingly, these mice also display enhanced parental behavior and elevated neural activity in the medial preoptic area, whereas sexual behavior remains normal. These results identify Gαi2 as the primary G protein α-subunit mediating the detection of volatile chemosignals in the apical layer of the VNO, and they show that Gαi2+ VSNs and the brain circuits activated by these neurons play a central role in orchestrating and balancing territorial and infant-directed aggression of male mice through bidirectional activation and inhibition of different targets in the limbic system.


Subject(s)
Aggression , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Sensory Receptor Cells/metabolism , Territoriality , Vomeronasal Organ/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/physiology , Brain Mapping , Female , Gene Deletion , Male , Mice, Inbred C57BL , Mutation/genetics , Sexual Behavior, Animal
9.
J Endocrinol ; 233(3): 257-267, 2017 06.
Article in English | MEDLINE | ID: mdl-28356400

ABSTRACT

This paper aimed to investigate the mechanisms triggering ERK phosphorylation and its functional role in male sexual behaviour. ERK1/2-phosphorylated form was detected in the medial preoptic area of the hypothalamus (mPOA) during the sexual stimulation of naive and sexually experienced males who were killed 5 min after the first intromission. This mating-induced ERK phosphorylation was increased in sexually experienced males compared to that in naive mice. The functional role of the ERK1/2 pathway activation during sexual behaviour was explored with the administration of a MEK inhibitor, SL-327 (30 mg/kg, i.p.), 45 min before the contact with a receptive female. Inhibition of ERK phosphorylation was found to decrease sexual motivation in both naive and experienced males without altering their copulatory ability. The mechanisms potentially involved in this rapid ERK1/2 pathway activation were specified ex vivo on hypothalamic slices. A thirty-minute incubation with 100 nM of testosterone (T), dihydrotestosterone (DHT) or oestradiol (E2) led to ERK phosphorylation. No changes were observed after incubation with testosterone 3-(O-carboxymethyl)oxime-BSA (T-BSA), an impermeable to the plasma membrane form of testosterone. All these results indicate that ERK phosphorylation within the mPOA could be a key player in the motivational signalling pathway and considered as an index of sexual motivation. They also demonstrate the involvement of oestrogen receptor (ER) and androgen receptor (AR) transduction pathways in steroid-dependent ERK activation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Preoptic Area/metabolism , Sexual Behavior, Animal/physiology , Testosterone/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Male , Mice, Inbred C57BL , Motor Activity/physiology , Phosphorylation , Smell/physiology
10.
J Neurochem ; 138(3): 457-68, 2016 08.
Article in English | MEDLINE | ID: mdl-27216894

ABSTRACT

Odors processed by the main and accessory olfactory bulbs (MOB, AOB) are important for sexual behavior. Interestingly, both structures continue to receive new neurons during adulthood. A role for olfactory neurogenesis in sexual behavior in female mice has recently been shown and gonadal hormones such as estradiol can modulate adult neurogenesis. Therefore, we wanted to determine the role of estradiol in learning the odors of sexual partners and in the adult neurogenesis of female aromatase knockout mice (ArKO), unable to produce estradiol. Female wild-type (WT) and ArKO mice were exposed to male odors during 7 days, and olfactory preferences, cell proliferation, cell survival and functional involvement of newborn neurons were analyzed, using BrdU injections, in combination with a marker of cell activation (Zif268) and neuronal fate (doublecortin, NeuN). Behavioral tasks indicated that both WT and ArKO females were able to discriminate between the odors of two different males, but ArKO mice failed to learn the familiar male odor. Proliferation of newborn cells was reduced in ArKO mice only in the dentate gyrus of the hippocampus. Olfactory exposure decreased cell survival in the AOB in WT females, suggesting a role for estradiol in a structure involved in sexual behavior. Finally, newborn neurons do not seem to be functionally involved in the AOB of ArKO mice compared with WT, when females were exposed to the odor of a familiar male, suggesting that estradiol-induced neurogenesis in the AOB is required for the learning of the male odor in female mice. Aromatase knockout mice (ArKO) presented deficits in olfactory preferences without affecting their olfactory discrimination abilities, and showed no functional involvement of newborn neurons in the accessory olfactory bulb (AOB) in response to the odor of a familiar male. These results suggest that estradiol-induced neurogenesis in the female AOB is required for the learning of the male odor.


Subject(s)
Estradiol/pharmacology , Learning/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Olfactory Bulb/drug effects , Animals , Female , Male , Mice, Knockout , Neurogenesis/physiology , Odorants , Olfactory Bulb/cytology , Ovariectomy/methods , Sex Characteristics , Smell/physiology
11.
Horm Behav ; 80: 1-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26836767

ABSTRACT

Estradiol derived from neural aromatization of gonadal testosterone plays a key role in the perinatal organization of the neural circuitry underlying male sexual behavior. The aim of this study was to investigate the contribution of neural estrogen receptor (ER) ß in estradiol-induced effects without interfering with its peripheral functions. For this purpose, male mice lacking ERß in the nervous system were generated. Analyses of males in two consecutive tests with a time interval of two weeks showed an effect of experience, but not of genotype, on the latencies to the first mount, intromission, pelvic thrusting and ejaculation. Similarly, there was an effect of experience, but not of genotype, on the number of thrusts and mating length. Neural ERß deletion had no effect on the ability of males to adopt a lordosis posture in response to male mounts, after castration and priming with estradiol and progesterone. Indeed, only low percentages of both genotypes exhibited a low lordosis quotient. It also did not affect their olfactory preference. Quantification of tyrosine hydroxylase- and kisspeptin-immunoreactive neurons in the preoptic area showed unaffected sexual dimorphism of both populations in mutants. By contrast, the number of androgen receptor- and ERα-immunoreactive cells was significantly increased in the bed nucleus of stria terminalis of mutant males. These data show that neural ERß does not play a crucial role in the organization and activation of the neural circuitry underlying male sexual behavior. These discrepancies with the phenotype of global ERß knockout models are discussed.


Subject(s)
Estrogen Receptor beta/genetics , Mice , Mutagenesis/genetics , Pregnancy , Sexual Behavior, Animal/physiology , Animals , Chromosome Deletion , Female , Fertility/genetics , Hypothalamus, Anterior/metabolism , Male , Mice, Knockout , Neuroglia/metabolism , Neurons/metabolism , Preoptic Area/physiology , Septal Nuclei/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...