Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
bioRxiv ; 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39229086

RÉSUMÉ

Many sex differences in brain and behavior are established developmentally by the opposing processes of feminization and masculinization, which manifest following differential steroid hormone exposure in early life. The cellular mechanisms underlying masculinization are well-documented, a result of the fact that it is steroid-mediated and can be easily induced in newborn female rodents via exogenous steroid treatment. However, the study of feminization of particular brain regions has largely been relegated to being "not masculinization" given the absence of an identified initiating trigger. As a result, the mechanisms of this key developmental process remain elusive. Here we describe a novel role for microglia, the brain's innate immune cell, in the feminization of the medial amygdala and a complex social behavior, juvenile play. In the developing amygdala, microglia promote proliferation of astrocytes equally in both sexes, with no apparent effect on rates of cell division, but support cell survival selectively in females through the trophic actions of Tumor Necrosis Factor α (TNFα). We demonstrate that disrupting TNFα signaling, either by depleting microglia or inhibiting the associated signaling pathways, prevents the feminization of astrocyte density and increases juvenile play levels to that seen in males. This data, combined with our previous finding that male-like patterns of astrocyte density are sculpted by developmental microglial phagocytosis, reveals that sexual differentiation of the medial amygdala involves opposing tensions between active masculinization and active feminization, both of which require microglia but are achieved via distinct processes.

2.
Adv Neurobiol ; 39: 165-191, 2024.
Article de Anglais | MEDLINE | ID: mdl-39190075

RÉSUMÉ

Engagement of astrocytes within the brain's reward circuitry has been apparent for approximately 30 years, when noncontingent drug administration was observed to lead to cytological markers of reactive astrocytes. Since that time, advanced approaches in rodent behavior and astrocyte monitoring have revealed complex interactions between astrocytes with drug type, animal sex, brain region, and dose and duration of drug administration. A number of studies now collectively reveal that rodent drug self-administration followed by prolonged abstinence results in decreased features of structure and synaptic colocalization of astrocytes. In addition, stimulation of astrocytes in the nucleus accumbens with DREADD receptors or pharmacological compounds opposes drug-seeking behavior. These findings provide a clear path for ongoing investigation into astrocytes as mediators of drug action in the brain and underscore the potential therapeutic utility of astrocytes in the regulation of drug craving and relapse vulnerability.


Sujet(s)
Astrocytes , Neurones , Troubles liés à une substance , Astrocytes/métabolisme , Animaux , Troubles liés à une substance/métabolisme , Humains , Neurones/métabolisme , Noyau accumbens/métabolisme , Comportement de recherche de substances , Encéphale/métabolisme , Récompense , Communication cellulaire/physiologie
3.
NeuroImmune Pharm Ther ; 3(2): 155-168, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39175524

RÉSUMÉ

Objectives: To study the effect of viral inflammation induced by Polyinosinic:polycytidylic acid (PIC) on the cerebellum during a critical period of development in rats. Methods: Neonatal rat pups were treated with PIC on postnatal days (PN) 8 and 10 after which we quantified RNA using Nanostring, qRT-PCR and RNAscope and analyzed immune cells through flow cytometry and immunohistochemistry on PN11. Using the same paradigm, we also analyzed play juvenile behavior, anxiety-like behavior, motor balance using the balance beam and the rotarod assays as well as fine motor behavior using the sunflower seed opening test. Results: We determined that male and female pups treated with PIC reacted with a significant increase in CCL5, a chemotactic cytokine that attracts T-cells, eosinophils and basophils to the site of inflammation, at PN11. PIC treatment also increased the expression of two receptors for CCL5, CCR1 and CCR5 in the cerebellar vermis in both males and females at PN11. In-situ hybridization (RNAscope®) for specific transcripts revealed that microglia express both CCL5 receptors under inflammatory and non-inflammatory conditions in both males and females. PIC treatment also increased the total number of CCL5+ cells in the developing cerebellum which were determined to be both natural killer cells and T-cells. There were modest but significant impacts of PIC treatment on large and fine motor skills and juvenile play behavior. Conclusions: Our findings suggest an important role for CCL5 and other immune cells in mediating inflammation in the developing cerebellum that potentially impact the maturation of cerebellar neurons during a critical period of development.

4.
Proc Natl Acad Sci U S A ; 120(10): e2212646120, 2023 03 07.
Article de Anglais | MEDLINE | ID: mdl-36848562

RÉSUMÉ

The sexually dimorphic nucleus of the preoptic area (SDN-POA) is the oldest and most robust sex difference reported in mammalian brain and is singular for its presence across a wide range of species from rodents to ungulates to man. This small collection of Nissl-dense neurons is reliably larger in volume in males. Despite its notoriety and intense interrogation, both the mechanism establishing the sex difference and the functional role of the SDN have remained elusive. Convergent evidence from rodent studies led to the conclusion that testicular androgens aromatized to estrogens are neuroprotective in males and that higher apoptosis (naturally occurring cell death) in females determines their smaller SDN. In several species, including humans, a smaller SDN correlates with a preference for mating with males. We report here that this volume difference is dependent upon a participatory role of phagocytic microglia which engulf more neurons in the female SDN and assure their destruction. Selectively blocking microglia phagocytosis temporarily spared neurons from apoptotic death and increased SDN volume in females without hormone treatment. Increasing the number of neurons in the SDN in neonatal females resulted in loss of preference for male odors in adulthood, an effect paralleled by dampened excitation of SDN neurons as evidenced by reduced immediate early gene (IEG) expression when exposed to male urine. Thus, the mechanism establishing a sex difference in SDN volume includes an essential role for microglia, and SDN function as a regulator of sexual partner preference is confirmed.


Sujet(s)
Microglie , Aire préoptique , Humains , Rats , Femelle , Mâle , Animaux , Comportement sexuel , Reproduction , Phagocytose , Mammifères
5.
Cell Rep ; 41(12): 111860, 2022 12 20.
Article de Anglais | MEDLINE | ID: mdl-36543121

RÉSUMÉ

Spatially distant areas of the cerebral cortex coordinate their activity into networks that are integral to cognitive processing. A common structural motif of cortical networks is co-activation of frontal and posterior cortical regions. The neural circuit mechanisms underlying such widespread inter-areal cortical coordination are unclear. Using a discovery based functional magnetic resonance imaging (fMRI) approach in mouse, we observe frontal and posterior cortical regions that demonstrate significant functional connectivity with the subcortical nucleus, the claustrum. Examining whether the claustrum synaptically supports such frontoposterior cortical network architecture, we observe cortico-claustro-cortical circuits reflecting the fMRI data: significant trans-claustral synaptic connectivity from frontal cortices to posteriorly lying sensory and sensory association cortices contralaterally. These data reveal discrete cortical pathways through the claustrum that are positioned to support cortical network motifs central to cognitive control functions and add to the canon of major extended cortico-subcortico-cortical systems in the mammalian brain.


Sujet(s)
Claustrum , Souris , Animaux , Noyaux gris centraux/physiologie , Cortex cérébral , Lobe frontal , Lobe pariétal/physiologie , Imagerie par résonance magnétique , Voies nerveuses/physiologie , Mammifères
6.
Pharmacol Res Perspect ; 10(5): e00999, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-36029006

RÉSUMÉ

Modulation of the endocannabinoid system (ECS) is a novel putative target for therapeutic intervention in depressive disorders. Altering concentrations of one of the principal endocannabinoids, N-arachidonoylethanolamine, also known as anandamide (AEA) can affect depressive-like behaviors through several mechanisms including anti-inflammatory, hormonal, and neural circuit alterations. Recently, isoflavonoids, a class of plant-derived compounds, have been of therapeutic interest given their ability to modulate the metabolism of the endogenous ligands of the ECS. To determine the therapeutic potential of isoflavonoids, we screened several candidate compounds (Genistein, Biochanin-A, and 7-hydroxyflavone) in silico to determine their binding properties with fatty acid amide hydrolase (FAAH), the primary degrative enzyme for AEA. We further validated the ability of these compounds to inhibit FAAH and determined their effects on depressive-like and locomotor behaviors in the forced swim test (FST) and open field test in male and female mice. We found that while genistein was the most potent FAAH inhibitor, 7-hydroxyflavone was most effective at reducing immobility time in the forced swim test. Finally, we measured blood corticosterone and prefrontal cortex AEA concentrations following the forced swim test and found that all tested compounds decreased corticosterone and increased AEA, demonstrating that isoflavonoids are promising therapeutic targets as FAAH inhibitors.


Sujet(s)
Endocannabinoïdes , Génistéine , Amidohydrolases , Animaux , Antidépresseurs , Acides arachidoniques , Corticostérone , Souris , Amides gras polyinsaturés N-alkylés
8.
Front Behav Neurosci ; 16: 1076765, 2022.
Article de Anglais | MEDLINE | ID: mdl-36755666

RÉSUMÉ

Social play is a dynamic and rewarding behavior abundantly expressed by most mammals during the juvenile period. While its exact function is debated, various rodent studies on the effects of juvenile social isolation suggest that participating in play is essential to appropriate behavior and reproductive success in adulthood. However, the vast majority of these studies were conducted in one sex only, a critical concern given the fact that there are known sex differences in play's expression: across nearly all species that play, males play more frequently and intensely than females, and there are qualitative sex differences in play patterns. Further limiting our understanding of the importance of play is the use of total isolation to prevent interactions with other juveniles. Here, we employed a novel cage design to specifically prevent play in rats while allowing for other forms of social interaction. We find that play deprivation during the juvenile period results in enduring sex-specific effects on later-life behavior, primarily in males. Males prevented from playing as juveniles exhibited decreased sexual behavior, hypersociability, and increased aggressiveness in adulthood, with no effects on these measures in females. Importantly, play deprivation had no effect on anxiety-like behavior, object memory, sex preference, or social recognition in either sex, showing the specificity of the identified impairments, though there were overall sex differences in many of these measures. Additionally, acute play deprivation impaired performance on a test of prosocial behavior in both sexes, indicating a difference in the motivation and/or ability to acquire this empathy-driven task. Together, these findings provide novel insight into the importance and function of juvenile social play and how this differs in males and females.

9.
J Neurosci Res ; 100(3): 713-730, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34882838

RÉSUMÉ

Cannabis use during pregnancy has increased over the past few decades, with recent data indicating that, in youth and young adults especially, up to 22% of people report using cannabis during pregnancy. Animal models provide the ability to study prenatal cannabis exposure (PCE) with control over timing and dosage; however, these studies utilize both injection and inhalation approaches. While it is known that Δ9-tetrahydrocannabinol (THC; primary psychoactive component of cannabis) can cross the placenta, examination of the transmission and concentration of THC and its metabolites from maternal blood into the placenta and fetal brain remains relatively unknown, and the influence of route of administration has never been examined. Pregnant female rats were exposed to either vaporized THC-dominant cannabis extract for pulmonary consumption or subcutaneous injection of THC repeatedly during the gestational period. Maternal blood, placenta, and fetal brains were collected following the final administration of THC for analysis of THC and its metabolites, as well as endocannabinoid concentrations, through mass spectrometry. Both routes of administration resulted in the transmission of THC and its metabolites in placenta and fetal brain. Repeated exposure to inhaled THC vapor resulted in fetal brain THC concentrations that were about 30% of those seen in maternal blood, whereas repeated injections resulted in roughly equivalent concentrations of THC in maternal blood and fetal brain. Neither inhalation nor injection of THC during pregnancy altered fetal brain endocannabinoid concentrations. Our data provide the first characterization of maternal-fetal transmission of THC and its metabolites following both vaporized delivery and injection routes of administration. These data are important to establish the maternal-fetal transmission in preclinical injection and inhalation models of PCE and may provide insight into predicting fetal exposure in human studies.


Sujet(s)
Dronabinol , Placenta , Adolescent , Animaux , Agonistes des récepteurs de cannabinoïdes , Femelle , Humains , Grossesse , Rats
10.
Brain Behav Immun Health ; 18: 100373, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34761244

RÉSUMÉ

Microglia, the innate immune cells of the brain, are indispensable for proper brain development. As professional phagocytes, microglia engulf other cells within distinct developmental niches to sculpt the architecture of the brain. Here, I highlight the age-, brain region-, and substrate-dependent diversity of developmental phagocytosis, and pose the idea that phagocytosis may, in turn, drive changes in microglia phenotype. Ultimately, phagocytosis might be just as important for shaping microglia function as it is for shaping the brain.

11.
eNeuro ; 8(5)2021.
Article de Anglais | MEDLINE | ID: mdl-34417284

RÉSUMÉ

Neuroscience has been transformed by the ability to genetically modify inbred mice, including the ability to express fluorescent markers specific to cell types or activation states. This approach has been put to particularly good effect in the study of the innate immune cells of the brain, microglia. These specialized macrophages are exceedingly small and complex, but also highly motile and mobile. To date, there have been no tools similar to those in mice available for studying these fundamental cells in the rat brain, and we seek to fill that gap with the generation of the genetically modified Sprague Dawley rat line: SD-Tg(Iba1-EGFP)Mmmc Using CRISPR-Cas/9 technology, we knocked in EGFP to the promoter of the gene Iba1 With four male and three female founders confirmed by quantitative PCR analysis to have appropriate and specific insertion, we established a breeding colony with at least three generations of backcrosses to obtain stable and reliable Iba1-EGFP expression. The specificity of EGFP expression to microglia was established by flow cytometry for CD45low/CD11b+ cells and by immunohistochemistry. Microglial EGFP expression was detected in neonates and persisted into adulthood. Blood macrophages and monocytes were found to express low levels of EGFP, as expected. Last, we show that EGFP expression is suitable for live imaging of microglia processes in acute brain slices and via intravital two-photon microscopy.


Sujet(s)
Microglie , Rodentia , Animaux , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Rats , Rat Sprague-Dawley , Rats transgéniques
12.
J Neurosci ; 40(23): 4609-4619, 2020 06 03.
Article de Anglais | MEDLINE | ID: mdl-32350039

RÉSUMÉ

Idebenone is a synthetic quinone that on reduction in cells can bypass mitochondrial Complex I defects by donating electrons to Complex III. The drug is used clinically to treat the Complex I disease Leber's hereditary optic neuropathy (LHON), but has been less successful in clinical trials for other neurodegenerative diseases. NAD(P)H:quinone oxidoreductase 1 (NQO1) appears to be the main intracellular enzyme catalyzing idebenone reduction. However, NQO1 is not universally expressed by cells of the brain. Using primary rat cortical cells pooled from both sexes, we tested the hypotheses that the level of endogenous NQO1 activity limits the ability of neurons, but not astrocytes, to use idebenone as an electron donor to support mitochondrial respiration. We then tested the prediction that NQO1 induction by pharmacological activation of the transcription factor nuclear erythroid 2-related factor 2 (Nrf2) enables idebenone to bypass Complex I in cells with poor NQO1 expression. We found that idebenone stimulated respiration by astrocytes but reduced the respiratory capacity of neurons. Importantly, idebenone supported mitochondrial oxygen consumption in the presence of a Complex I inhibitor in astrocytes but not neurons, and this ability was reversed by inhibiting NQO1. Conversely, recombinant NQO1 delivery to neurons prevented respiratory impairment and conferred Complex I bypass activity. Nrf2 activators failed to increase NQO1 in neurons, but carnosic acid induced NQO1 in COS-7 cells that expressed little endogenous enzyme. Carnosic acid-idebenone combination treatment promoted NQO1-dependent Complex I bypass activity in these cells. Thus, combination drug strategies targeting NQO1 may promote the repurposing of idebenone for additional disorders.SIGNIFICANCE STATEMENT Idebenone is used clinically to treat loss of visual acuity in Leber's hereditary optic neuropathy. Clinical trials for several additional diseases have failed. This study demonstrates a fundamental difference in the way idebenone affects mitochondrial respiration in cortical neurons compared with cortical astrocytes. Cortical neurons are unable to use idebenone as a direct mitochondrial electron donor due to NQO1 deficiency. Our results suggest that idebenone behaves as an NQO1-dependent prodrug, raising the possibility that lack of neuronal NQO1 activity has contributed to the limited efficacy of idebenone in neurodegenerative disease treatment. Combination therapy with drugs able to safely induce NQO1 in neurons, as well as other brain cell types, may be able to unlock the neuroprotective therapeutic potential of idebenone or related quinones.


Sujet(s)
Antioxydants/pharmacologie , Astrocytes/enzymologie , Respiration cellulaire/physiologie , Mitochondries/enzymologie , NADPH dehydrogenase (quinone)/métabolisme , Ubiquinones/analogues et dérivés , Animaux , Animaux nouveau-nés , Astrocytes/effets des médicaments et des substances chimiques , Cellules COS , Respiration cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Chlorocebus aethiops , Relation dose-effet des médicaments , Activation enzymatique/effets des médicaments et des substances chimiques , Activation enzymatique/physiologie , Femelle , Mâle , Mitochondries/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Ubiquinones/pharmacologie
13.
Glia ; 68(6): 1100-1113, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-31691400

RÉSUMÉ

Microglia, the innate immune cells of the brain, have recently been removed from the position of mere sentinels and promoted to the role of active sculptors of developing circuits and cells. Alongside their functions in normal brain development, microglia coordinate sexual differentiation of the brain, a set of processes which vary by region and endpoint like that of microglia function itself. In this review, we highlight the ways microglia are both targets and drivers of brain sexual differentiation. We examine the factors that may drive sex differences in microglia, with a special focus on how changing microenvironments in the developing brain dictate microglia phenotypes and discuss how their diverse functions sculpt lasting sex-specific changes in the brain. Finally, we consider how sex-specific early life environments contribute to epigenetic programming and lasting sex differences in microglia identity.


Sujet(s)
Encéphale/cytologie , Microglie/cytologie , Caractères sexuels , Différenciation sexuelle/physiologie , Animaux , Différenciation cellulaire/physiologie , Humains , Neurones/cytologie
14.
Bio Protoc ; 10(1): e3481, 2020 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-33654714

RÉSUMÉ

Play is a complex social behavior that is highly conserved across mammals. In most species, males engage in more frequent and vigorous play as juveniles than females, which reflects subtle yet impactful sex differences in brain circuitry and development. In this protocol, we describe a behavioral testing paradigm to assess social play in male and female juvenile rats. We highlight the behavior scoring criteria for distinguishing rough-and-tumble play from other play-related social behaviors. By analyzing both sexes, play behavior can be leveraged as a powerful tool to understand the sex-specific development and expression of social behavior.

15.
Int J Play ; 9(1): 58-75, 2020.
Article de Anglais | MEDLINE | ID: mdl-33717644

RÉSUMÉ

Social play consists of reciprocal physical interactions between conspecifics with many features conserved across species, including the propensity for males to engage in play more frequently and with higher physical intensity. Animal models, such as the laboratory rat, reveal that the underlying neural circuitry of play is subject to sexual differentiation during a critical period early in life. In this review, we discuss the developmental processes that produce distinct neural nodes which modulate both shared and sex-specific aspects of play with a focus on the medial amygdala, lateral septum, and prefrontal cortex. While the cellular mechanisms determining sex differences in play are beginning to be uncovered, the ultimate advantages of play continue to be debated.

16.
Neuron ; 102(2): 435-449.e6, 2019 04 17.
Article de Anglais | MEDLINE | ID: mdl-30827729

RÉSUMÉ

Brain sex differences are established developmentally and generate enduring changes in circuitry and behavior. Steroid-mediated masculinization of the rat amygdala during perinatal development produces higher levels of juvenile rough-and-tumble play by males. This sex difference in social play is highly conserved across mammals, yet the mechanisms by which it is established are unknown. Here, we report that androgen-induced increases in endocannabinoid tone promote microglia phagocytosis during a critical period of amygdala development. Phagocytic microglia engulf more viable newborn cells in males; in females, less phagocytosis allows more astrocytes to survive to the juvenile age. Blocking complement-dependent phagocytosis in males increases astrocyte survival and prevents masculinization of play. Moreover, increased astrocyte density in the juvenile amygdala reduces neuronal excitation during play. These findings highlight novel mechanisms of brain development whereby endocannabinoids induce microglia phagocytosis to regulate newborn astrocyte number and shape the sexual differentiation of social circuitry and behavior. VIDEO ABSTRACT.


Sujet(s)
Amygdale (système limbique)/métabolisme , Astrocytes/métabolisme , Endocannabinoïdes/métabolisme , Microglie/physiologie , Phagocytose/physiologie , Jeu et accessoires de jeu , Caractères sexuels , Comportement social , Amygdale (système limbique)/cytologie , Amygdale (système limbique)/effets des médicaments et des substances chimiques , Amygdale (système limbique)/croissance et développement , Antagonistes des androgènes/pharmacologie , Androgènes/métabolisme , Androgènes/pharmacologie , Animaux , Animaux nouveau-nés , Acides arachidoniques/métabolisme , Comportement animal , Survie cellulaire , Protéines du système du complément/métabolisme , Protéines du système du complément/physiologie , Endocannabinoïdes/physiologie , Femelle , Flutamide/pharmacologie , Glycérides/métabolisme , Mâle , Microglie/effets des médicaments et des substances chimiques , Phagocytose/effets des médicaments et des substances chimiques , Amides gras polyinsaturés N-alkylés/métabolisme , Rats , Testostérone/métabolisme , Testostérone/pharmacologie
17.
Dev Neurobiol ; 78(6): 580-592, 2018 Jun.
Article de Anglais | MEDLINE | ID: mdl-29243403

RÉSUMÉ

The proverbial role of microglia during brain development is shifting from passive members of the brain's immune system to active participants that are able to dictate enduring outcomes. Despite these advances, little attention has been paid to one of the most critical components of early brain development-sexual differentiation. Mounting evidence suggests that the normal developmental functions microglia perform-cell number regulation and synaptic connectivity-may be involved in the sex-specific patterning of the brain during these early sensitive periods, and may have lasting sex-dependent and sex-independent effects on behavior. In this review, we outline the known functions of microglia during developmental sensitive periods, and highlight the role they play in the establishment of sex differences in brain and behavior. We also propose a framework for how researchers can incorporate microglia in their study of sex differences and vice versa. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 580-592, 2018.


Sujet(s)
Encéphale/croissance et développement , Encéphale/physiologie , Microglie/physiologie , Caractères sexuels , Animaux , Humains
18.
eNeuro ; 4(1)2017.
Article de Anglais | MEDLINE | ID: mdl-28144625

RÉSUMÉ

Juvenile social play behavior is a shared trait across a wide variety of mammalian species. When play is characterized by the frequency or duration of physical contact, males usually display more play relative to females. The endocannabinoid system contributes to the development of the sex difference in social play behavior in rats. Treating newborn pups with a nonspecific endocannabinoid agonist, WIN55,212-2, masculinizes subsequent juvenile rough-and-tumble play behavior by females. Here we use specific drugs to target signaling through either the CB1 or CB2 endocannabinoid receptor (CB1R or CB2R) to determine which modulates the development of sex differences in play. Our data reveal that signaling through both CB1R and CB2R must be altered neonatally to modify development of neural circuitry regulating sex differences in play. Neonatal co-agonism of CB1R and CB2R masculinized play by females, whereas co-antagonism of these receptors feminized rates of male play. Because of a known role for the medial amygdala in the sexual differentiation of play, we reconstructed Golgi-impregnated neurons in the juvenile medial amygdala and used factor analysis to identify morphological parameters that were sexually differentiated and responsive to dual agonism of CB1R and CB2R during the early postnatal period. Our results suggest that sex differences in the medial amygdala are modulated by the endocannabinoid system during early development. Sex differences in play behavior are loosely correlated with differences in neuronal morphology.


Sujet(s)
Amygdale (système limbique)/croissance et développement , Amygdale (système limbique)/métabolisme , Récepteur cannabinoïde de type CB1/métabolisme , Récepteur cannabinoïde de type CB2/métabolisme , Caractères sexuels , Comportement social , Amygdale (système limbique)/cytologie , Amygdale (système limbique)/effets des médicaments et des substances chimiques , Animaux , Animaux nouveau-nés , Modulateurs des récepteurs de cannabinoïdes/pharmacologie , Femelle , Mâle , Voies nerveuses/cytologie , Voies nerveuses/effets des médicaments et des substances chimiques , Voies nerveuses/croissance et développement , Voies nerveuses/métabolisme , Neurones/cytologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Rat Sprague-Dawley , Récepteur cannabinoïde de type CB1/agonistes , Récepteur cannabinoïde de type CB1/antagonistes et inhibiteurs , Récepteur cannabinoïde de type CB2/agonistes , Récepteur cannabinoïde de type CB2/antagonistes et inhibiteurs
19.
eNeuro ; 3(6)2016.
Article de Anglais | MEDLINE | ID: mdl-27957532

RÉSUMÉ

Microglia are the primary immune cells of the brain and function in multiple ways to facilitate proper brain development. However, our current understanding of how these cells influence the later expression of normal behaviors is lacking. Using the laboratory rat, we administered liposomal clodronate centrally to selectively deplete microglia in the developing postnatal brain. We then assessed a range of developmental, juvenile, and adult behaviors. Liposomal clodronate treatment on postnatal days 0, 2, and 4 depleted microglia with recovery by about 10 days of age and induced a hyperlocomotive phenotype, observable in the second postnatal week. Temporary microglia depletion also increased juvenile locomotion in the open field test and decreased anxiety-like behaviors in the open field and elevated plus maze. These same rats displayed reductions in predator odor-induced avoidance behavior, but increased their risk assessment behaviors compared with vehicle-treated controls. In adulthood, postnatal microglia depletion resulted in significant deficits in male-specific sex behaviors. Using factor analysis, we identified two underlying traits-behavioral disinhibition and locomotion-as being significantly altered by postnatal microglia depletion. These findings further implicate microglia as being critically important to the development of juvenile and adult behavior.


Sujet(s)
Comportement animal/physiologie , Encéphale/croissance et développement , Encéphale/physiologie , Microglie/physiologie , Caractères sexuels , Animaux , Animaux nouveau-nés , Anxiété/physiopathologie , Apprentissage par évitement/physiologie , Encéphale/anatomopathologie , Acide clodronique , Femelle , Inhibition psychologique , Liposomes , Mâle , Microglie/anatomopathologie , Modèles animaux , Activité motrice/physiologie , Comportement prédateur , Rat Sprague-Dawley , Prise de risque
20.
eNeuro ; 2(4)2015.
Article de Anglais | MEDLINE | ID: mdl-26464992

RÉSUMÉ

Microglia are being increasingly recognized as playing important roles in neurodevelopment. The cerebellum matures postnatally, undergoing major growth, but the role of microglia in the developing cerebellum is not well understood. Using the laboratory rat we quantified and morphologically categorized microglia throughout the vermis and across development using a design-based unbiased stereology method. We found that microglial morphology changed from amoeboid to ramified during the first 3 postnatal weeks in a region specific manner. These morphological changes were accompanied by the sudden appearance of phagocytic cups during the third postnatal week from P17 to P19, with an approximately fourfold increase compared with the first week, followed by a prompt decline at the end of the third week. The microglial phagocytic cups were significantly higher in the granular layer (∼69%) than in the molecular layer (ML; ∼31%) during a 3 d window, and present on ∼67% of microglia with thick processes and ∼33% of microglia with thin processes. Similar proportions of phagocytic cups associated to microglia with either thick or thin processes were found in the ML. We observed cell nuclei fragmentation and cleaved caspase-3 expression within some microglial phagocytic cups, presumably from dying granule neurons. At P17 males showed an approximately twofold increase in microglia with thin processes compared with females. Our findings indicate a continuous process of microglial maturation and a nonuniform distribution of microglia in the cerebellar cortex that implicates microglia as an important cellular component of the developing cerebellum.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE