Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Neuron ; 109(11): 1760-1762, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34081917

ABSTRACT

Memory develops during early life, yet the corresponding molecular mechanisms are largely unknown. Leinwand and Scott (2021) reveal a link between juvenile hormone, neural activity, and memory-evoked behavior during a critical period that promotes associative learning in the adult fly.


Subject(s)
Juvenile Hormones , Mushroom Bodies , Animals , Brain , Conditioning, Classical , Memory
2.
eNeuro ; 8(2)2021.
Article in English | MEDLINE | ID: mdl-33622703

ABSTRACT

Elevation of serotonin via postnatal fluoxetine (PNFlx) treatment during critical temporal windows is hypothesized to perturb the development of limbic circuits thus establishing a substratum for persistent disruption of mood-related behavior. We examined the impact of PNFlx treatment on the formation and maintenance of perineuronal nets (PNNs), extracellular matrix (ECM) structures that deposit primarily around inhibitory interneurons, and mark the closure of critical period plasticity. PNFlx treatment evoked a significant decline in PNN number, with a robust reduction in PNNs deposited around parvalbumin (PV) interneurons, within the CA1 and CA3 hippocampal subfields at postnatal day (P)21 in Sprague Dawley rat pups. While the reduction in CA1 subfield PNN number was still observed in adulthood, we observed no change in colocalization of PV-positive interneurons with PNNs in the hippocampi of adult PNFlx animals. PNFlx treatment did not alter hippocampal PV, calretinin (CalR), or Reelin-positive neuron numbers in PNFlx animals at P21 or in adulthood. We did observe a small, but significant increase in somatostatin (SST)-positive interneurons in the DG subfield of PNFlx-treated animals in adulthood. This was accompanied by altered GABA-A receptor subunit composition, increased dendritic complexity of apical dendrites of CA1 pyramidal neurons, and enhanced neuronal activation revealed by increased c-Fos-positive cell numbers within hippocampi of PNFlx-treated animals in adulthood. These results indicate that PNFlx treatment alters the formation of PNNs within the hippocampus, raising the possibility of a disruption of excitation-inhibition (E/I) balance within this key limbic brain region.


Subject(s)
Fluoxetine , Parvalbumins , Animals , Extracellular Matrix/metabolism , Fluoxetine/pharmacology , Hippocampus/metabolism , Interneurons/metabolism , Parvalbumins/metabolism , Rats , Rats, Sprague-Dawley , Reelin Protein
3.
Elife ; 92020 09 21.
Article in English | MEDLINE | ID: mdl-32955432

ABSTRACT

Early adversity is a risk factor for the development of adult psychopathology. Common across multiple rodent models of early adversity is increased signaling via forebrain Gq-coupled neurotransmitter receptors. We addressed whether enhanced Gq-mediated signaling in forebrain excitatory neurons during postnatal life can evoke persistent mood-related behavioral changes. Excitatory hM3Dq DREADD-mediated chemogenetic activation of forebrain excitatory neurons during postnatal life (P2-14), but not in juvenile or adult windows, increased anxiety-, despair-, and schizophrenia-like behavior in adulthood. This was accompanied by an enhanced metabolic rate of cortical and hippocampal glutamatergic and GABAergic neurons. Furthermore, we observed reduced activity and plasticity-associated marker expression, and perturbed excitatory/inhibitory currents in the hippocampus. These results indicate that Gq-signaling-mediated activation of forebrain excitatory neurons during the critical postnatal window is sufficient to program altered mood-related behavior, as well as functional changes in forebrain glutamate and GABA systems, recapitulating aspects of the consequences of early adversity.


Stress and adversity in early childhood can have long-lasting effects, predisposing people to mental illness and mood disorders in adult life. The weeks immediately before and after birth are critical for establishing key networks of neurons in the brain. Therefore, any disruption to these neural circuits during this time can be detrimental to emotional development. However, it is still unclear which cellular mechanisms cause these lasting changes in behavior. Studies in animals suggest that these long-term effects could result from abnormalities in a few signaling pathways in the brain. For example, it has been proposed that overstimulating the cells that activate circuits in the forebrain ­ also known as excitatory neurons ­ may contribute to the behavioral changes that persist into adulthood. To test this theory, Pati et al. used genetic engineering to modulate a signaling pathway in male mice, which is known to stimulate excitatory neurons in the forebrain. The experiments showed that prolonged activation of excitatory neurons in the first two weeks after birth resulted in anxious and despair-like behaviors as the animals aged. The mice also displayed discrepancies in how they responded to certain external sensory information, which is a hallmark of schizophrenia-like behavior. However, engineering the same changes in adolescent and adult mice had no effect on their mood-related behaviors. This animal study reinforces just how critical the first few weeks of life are for optimal brain development. It provides an insight into a possible mechanism of how disruption during this time could alter emotional behavior. The findings are also relevant to psychiatrists interested in the underlying causes of mental illness after early childhood adversity.


Subject(s)
Affect/physiology , Behavior, Animal/physiology , Neurons/physiology , Prosencephalon/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Animals, Newborn/growth & development , Animals, Newborn/physiology , Anxiety/etiology , Female , GABAergic Neurons/physiology , Hippocampus/physiology , Male , Mice
4.
Curr Opin Neurobiol ; 60: 129-135, 2020 02.
Article in English | MEDLINE | ID: mdl-31875530

ABSTRACT

The decision to urinate is a social behavior that is calculated multiple times a day. Many animals perform urine scent-marking which broadcasts their pheromones to regulate the behavior of others and humans are trained at an early age to urinate only at a socially acceptable time and place. The inability to control when and where to void, that is incontinence, causes extreme social discomfort yet targeted therapeutics are lacking because little is known about the underlying circuits and mechanisms. The use of animal models, neurocircuit analysis, and functional manipulation is beginning to reveal basic logic of the circuit that modulates the decision of when and where to void.


Subject(s)
Behavior, Animal , Urination , Animals , Odorants , Pheromones , Social Behavior
5.
Curr Biol ; 28(13): R746-R749, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29990458

ABSTRACT

Becoming a parent changes our choices and actions. Identifying the underlying neural circuits is necessary to understand the transformation of an animal's behavior post-parenthood. Multiple nodes of the 'parenting circuit' have now been identified to reveal the workings of a single brain region key to the orchestration of parent-specific behaviors.


Subject(s)
Maternal Behavior , Social Behavior , Animals , Brain , Female , Humans , Mesencephalon , Parenting
6.
J Biosci ; 43(1): 85-95, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29485117

ABSTRACT

The medial prefrontal cortex (mPFC) is implicated in anxiety-like behaviour. In rodent models, perturbations of mPFC neuronal activity through pharmacological manipulations, optogenetic activation of mPFC neurons or cell-type specific pharmacogenetic inhibition of somatostatin interneurons indicate conflicting effects on anxiety-like behaviour. In the present study we examined the effects of pharmacogenetic activation of Ca 2+/calmodulin-dependent protein kinase alpha (CamKII alpha)-positive excitatory neurons on anxiety-like behaviour. We used clozapine-N-oxide (CNO) to pharmacogenetically activate virally delivered CamKII alpha-hM3Dq-DREADD in mPFC excitatory neurons. The effects of acute CNO or vehicle treatment on anxiety-like behaviour in the open field and elevated plus maze tests were examined in rats virally infected with either CamKII alpha-hM3Dq-DREADD or CamKII alpha-GFP. In addition, the effects of acute CNO treatment on the expression of the neuronal activity marker c-Fos were examined in the mPFC as well as downstream target neuronal circuits using immunohistochemistry. Acute pharmacogenetic activation of mPFC excitatory neurons evoked a significant decrease in anxiety-like behaviour selectively on the elevated plus maze task, but not the open field test. Acute CNO treatment resulted in enhanced c-Fos-immunopositive cell number in the infralimbic, prelimbic and cingulate subdivisions of the mPFC. This was also accompanied by enhanced c-Fos-immunopositive cell number in multiple downstream circuits of the mPFC in CNO-treated hM3Dq animals. Acute pharmacogenetic activation of mPFC excitatory neurons reduces anxietylike behaviour in a task-specific fashion accompanied by enhanced c-Fos expression in the mPFC and multiple target circuits implicated in the regulation of anxiety-like behaviour.


Subject(s)
Antipsychotic Agents/pharmacology , Anxiety/drug therapy , Carrier Proteins/genetics , Clozapine/analogs & derivatives , Pharmacogenetics/methods , Prefrontal Cortex/drug effects , Animals , Anxiety/genetics , Anxiety/metabolism , Anxiety/physiopathology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium-Binding Proteins , Carrier Proteins/metabolism , Clozapine/pharmacology , Dependovirus/genetics , Dependovirus/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Injections, Intraventricular , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Maze Learning , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Stereotaxic Techniques , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...