Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
eNeuro ; 10(8)2023 08.
Article in English | MEDLINE | ID: mdl-37553240

ABSTRACT

Expanding knowledge about the cellular composition of subcortical brain regions demonstrates large heterogeneity and differences from the cortical architecture. Previously we described three subtypes of somatostatin-expressing (Sst) neurons in the mouse ventral tegmental area (VTA) and showed their local inhibitory action on the neighboring dopaminergic neurons (Nagaeva et al., 2020). Here, we report that Sst+ neurons especially from the anterolateral part of the mouse VTA also project far outside the VTA and innervate forebrain regions that are mainly involved in the regulation of emotional behavior, including the ventral pallidum, lateral hypothalamus, the medial part of the central amygdala, anterolateral division of the bed nucleus of stria terminalis, and paraventricular thalamic nucleus. Deletion of these VTASst neurons in mice affected several behaviors, such as home cage activity, sensitization of locomotor activity to morphine, fear conditioning responses, and reactions to the inescapable stress of forced swimming, often in a sex-dependent manner. Together, these data demonstrate that VTASst neurons have selective projection targets distinct from the main targets of VTA dopamine neurons. VTASst neurons are involved in the regulation of behaviors primarily associated with the stress response, making them a relevant addition to the efferent VTA pathways and stress-related neuronal network.


Subject(s)
Dopaminergic Neurons , Ventral Tegmental Area , Mice , Animals , Ventral Tegmental Area/metabolism , Efferent Pathways/metabolism , Dopaminergic Neurons/metabolism , Hypothalamic Area, Lateral , Somatostatin/metabolism
2.
Commun Biol ; 6(1): 789, 2023 07 29.
Article in English | MEDLINE | ID: mdl-37516746

ABSTRACT

Cholesterol is an essential membrane structural component and steroid hormone precursor, and is involved in numerous signaling processes. Astrocytes regulate brain cholesterol homeostasis and they supply cholesterol to the needs of neurons. ATP-binding cassette transporter A1 (ABCA1) is the main cholesterol efflux transporter in astrocytes. Here we show dysregulated cholesterol homeostasis in astrocytes generated from human induced pluripotent stem cells (iPSCs) derived from males with fragile X syndrome (FXS), which is the most common cause of inherited intellectual disability. ABCA1 levels are reduced in FXS human and mouse astrocytes when compared with controls. Accumulation of cholesterol associates with increased desmosterol and polyunsaturated phospholipids in the lipidome of FXS mouse astrocytes. Abnormal astrocytic responses to cytokine exposure together with altered anti-inflammatory and cytokine profiles of human FXS astrocyte secretome suggest contribution of inflammatory factors to altered cholesterol homeostasis. Our results demonstrate changes of astrocytic lipid metabolism, which can critically regulate membrane properties and affect cholesterol transport in FXS astrocytes, providing target for therapy in FXS.


Subject(s)
Fragile X Syndrome , Induced Pluripotent Stem Cells , Male , Animals , Mice , Humans , Fragile X Syndrome/genetics , Astrocytes , Lipid Metabolism , Cytokines , Homeostasis
3.
Neuropsychopharmacology ; 48(7): 1021-1030, 2023 06.
Article in English | MEDLINE | ID: mdl-36944718

ABSTRACT

Critical period-like plasticity (iPlasticity) can be reinstated in the adult brain by several interventions, including drugs and optogenetic modifications. We have demonstrated that a combination of iPlasticity with optimal training improves behaviors related to neuropsychiatric disorders. In this context, the activation of TrkB, a receptor for BDNF, in Parvalbumin-positive (PV+) interneurons has a pivotal role in cortical network changes. However, it is unknown if the activation of TrkB in PV+ interneurons is important for other plasticity-related behaviors, especially for learning and memory. Here, using mice with heterozygous conditional TrkB deletion in PV+ interneurons (PV-TrkB hCKO) in IntelliCage and fear erasure paradigms, we show that chronic treatment with fluoxetine, a widely prescribed antidepressant drug that is known to promote the activation of TrkB, enhances behavioral flexibility in spatial and fear memory, largely depending on the expression of the TrkB receptor in PV+ interneurons. In addition, hippocampal long-term potentiation was enhanced by chronic treatment with fluoxetine in wild-type mice, but not in PV-TrkB hCKO mice. Transcriptomic analysis of PV+ interneurons after fluoxetine treatment indicated intrinsic changes in synaptic formation and downregulation of enzymes involved in perineuronal net formation. Consistently, immunohistochemistry has shown that the fluoxetine treatment alters PV expression and reduces PNNs in PV+ interneurons, and here we show that TrkB expression in PV+ interneurons is required for these effects. Together, our results provide molecular and network mechanisms for the induction of critical period-like plasticity in adulthood.


Subject(s)
Parvalbumins , Reversal Learning , Mice , Animals , Parvalbumins/metabolism , Fluoxetine/pharmacology , Receptor, trkB/metabolism , Interneurons/physiology , Fear , Antidepressive Agents/pharmacology , Antidepressive Agents/metabolism
4.
Front Mol Neurosci ; 15: 1032224, 2022.
Article in English | MEDLINE | ID: mdl-36407765

ABSTRACT

Brain-derived neurotrophic factor (BDNF) signaling through its receptor TrkB has for a long time been recognized as a critical mediator of the antidepressant drug action, but BDNF signaling has been considered to be activated indirectly through the action of typical and rapid-acting antidepressants through monoamine transporters and glutamate NMDA receptors, respectively. However, recent findings demonstrate that both typical and the fast-acting antidepressants directly bind to TrkB and thereby allosterically potentiate BDNF signaling, suggesting that TrkB is the direct target for antidepressant drugs. Increased TrkB signaling particularly in the parvalbumin-expressing interneurons orchestrates iPlasticity, a state of juvenile-like enhanced plasticity in the adult brain. iPlasticity sensitizes neuronal networks to environmental influences, enabling rewiring of networks miswired by adverse experiences. These findings have dramatically changed the position of TrkB in the antidepressant effects and they propose a new end-to-end model of the antidepressant drug action. This model emphasizes the enabling role of antidepressant treatment and the active participation of the patient in the process of recovery from mood disorders.

5.
Int J Mol Sci ; 23(18)2022 Sep 06.
Article in English | MEDLINE | ID: mdl-36142154

ABSTRACT

The activation of tropomyosin receptor kinase B (TrkB), the receptor of brain-derived neurotrophic factor (BDNF), plays a key role in induced juvenile-like plasticity (iPlasticity), which allows restructuring of neural networks in adulthood. Optically activatable TrkB (optoTrkB) can temporarily and spatially evoke iPlasticity, and recently, optoTrkB (E281A) was developed as a variant that is highly sensitive to light stimulation while having lower basal activity compared to the original optoTrkB. In this study, we validate optoTrkB (E281A) activated in alpha calcium/calmodulin-dependent protein kinase type II positive (CKII+) pyramidal neurons or parvalbumin-positive (PV+) interneurons in the mouse visual cortex by immunohistochemistry. OptoTrkB (E281A) was activated in PV+ interneurons and CKII+ pyramidal neurons with blue light (488 nm) through the intact skull and fur, and through a transparent skull, respectively. LED light stimulation significantly increased the intensity of phosphorylated ERK and CREB even through intact skull and fur. These findings indicate that the highly sensitive optoTrkB (E281A) can be used in iPlasticity studies of both inhibitory and excitatory neurons, with flexible stimulation protocols in behavioural studies.


Subject(s)
Brain-Derived Neurotrophic Factor , Visual Cortex , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calcium , Mice , Neurons/metabolism , Parvalbumins/metabolism , Receptor, trkB/metabolism , Tropomyosin/metabolism , Visual Cortex/metabolism
6.
Mol Psychiatry ; 26(12): 7247-7256, 2021 12.
Article in English | MEDLINE | ID: mdl-34321594

ABSTRACT

Elevated states of brain plasticity typical for critical periods of early postnatal life can be reinstated in the adult brain through interventions, such as antidepressant treatment and environmental enrichment, and induced plasticity may be critical for the antidepressant action. Parvalbumin-positive (PV) interneurons regulate the closure of developmental critical periods and can alternate between high and low plasticity states in response to experience in adulthood. We now show that PV plasticity states and cortical networks are regulated through the activation of TrkB neurotrophin receptors. Visual cortical plasticity induced by fluoxetine, a widely prescribed selective serotonin reuptake inhibitor (SSRI) antidepressant, was lost in mice with reduced expression of TrkB in PV interneurons. Conversely, optogenetic gain-of-function studies revealed that activation of an optically activatable TrkB (optoTrkB) specifically in PV interneurons switches adult cortical networks into a state of elevated plasticity within minutes by decreasing the intrinsic excitability of PV interneurons, recapitulating the effects of fluoxetine. TrkB activation shifted cortical networks towards a low PV configuration, promoting oscillatory synchrony, increased excitatory-inhibitory balance, and ocular dominance plasticity. OptoTrkB activation promotes the phosphorylation of Kv3.1 channels and reduces the expression of Kv3.2 mRNA providing a mechanism for the lower excitability. In addition, decreased expression and puncta of Synaptotagmin2 (Syt2), a presynaptic marker of PV interneurons involved in Ca2+-dependent neurotransmitter release, suggests lower inputs onto pyramidal neurons suppressing feed-forward inhibition. Together, the results provide mechanistic insights into how TrkB activation in PV interneurons orchestrates the activity of cortical networks and mediating antidepressant responses in the adult brain.


Subject(s)
Interneurons , Neuronal Plasticity , Visual Cortex , Animals , Interneurons/metabolism , Mice , Neuronal Plasticity/physiology , Parvalbumins/metabolism , Synaptic Transmission , Synaptotagmin II/metabolism , Visual Cortex/metabolism
7.
J Neurosci ; 41(5): 972-980, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33293360

ABSTRACT

Perineuronal nets (PNNs) are an extracellular matrix structure rich in chondroitin sulfate proteoglycans (CSPGs), which preferentially encase parvalbumin-containing (PV+) interneurons. PNNs restrict cortical network plasticity but the molecular mechanisms involved are unclear. We found that reactivation of ocular dominance plasticity in the adult visual cortex induced by chondroitinase ABC (chABC)-mediated PNN removal requires intact signaling by the neurotrophin receptor TRKB in PV+ neurons. Additionally, we demonstrate that chABC increases TRKB phosphorylation (pTRKB), while PNN component aggrecan attenuates brain-derived neurotrophic factor (BDNF)-induced pTRKB in cortical neurons in culture. We further found that protein tyrosine phosphatase σ (PTPσ, PTPRS), receptor for CSPGs, interacts with TRKB and restricts TRKB phosphorylation. PTPσ deletion increases phosphorylation of TRKB in vitro and in vivo in male and female mice, and juvenile-like plasticity is retained in the visual cortex of adult PTPσ-deficient mice (PTPσ+/-). The antidepressant drug fluoxetine, which is known to promote TRKB phosphorylation and reopen critical period-like plasticity in the adult brain, disrupts the interaction between TRKB and PTPσ by binding to the transmembrane domain of TRKB. We propose that both chABC and fluoxetine reopen critical period-like plasticity in the adult visual cortex by promoting TRKB signaling in PV+ neurons through inhibition of TRKB dephosphorylation by the PTPσ-CSPG complex.SIGNIFICANCE STATEMENT Critical period-like plasticity can be reactivated in the adult visual cortex through disruption of perineuronal nets (PNNs) by chondroitinase treatment, or by chronic antidepressant treatment. We now show that the effects of both chondroitinase and fluoxetine are mediated by the neurotrophin receptor TRKB in parvalbumin-containing (PV+) interneurons. We found that chondroitinase-induced visual cortical plasticity is dependent on TRKB in PV+ neurons. Protein tyrosine phosphatase σ (PTPσ, PTPRS), a receptor for PNNs, interacts with TRKB and inhibits its phosphorylation, and chondroitinase treatment or deletion of PTPσ increases TRKB phosphorylation. Antidepressant fluoxetine disrupts the interaction between TRKB and PTPσ, thereby increasing TRKB phosphorylation. Thus, juvenile-like plasticity induced by both chondroitinase and antidepressant treatment is mediated by TRKB activation in PV+ interneurons.


Subject(s)
Antidepressive Agents/pharmacology , Chondroitinases and Chondroitin Lyases/pharmacology , Membrane Glycoproteins/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Parvalbumins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Neurons/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology
8.
Sci Rep ; 10(1): 14984, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32917932

ABSTRACT

P75 neurotrophic receptor (p75NTR) is an important receptor for the role of neurotrophins in modulating brain plasticity and apoptosis. The current understanding of the role of p75NTR in cellular adaptation following pathological insults remains blurred, which makes p75NTR's related signaling networks an interesting and challenging initial point of investigation. We identified p75NTR and related genes through extensive data mining of a PubMed literature search including published works related to p75NTR from the past 20 years. Bioinformatic network and pathway analyses of identified genes (n = 235) were performed using ReactomeFIViz in Cytoscape based on the highly reliable Reactome functional interaction network algorithm. This approach merges interactions extracted from human curated pathways with predicted interactions from machine learning. Genome-wide pathway analysis showed total of 16 enriched hierarchical clusters. A total of 278 enriched single pathways were also identified (p < 0.05, false discovery rate corrected). Gene network analyses showed multiple known and new targets in the p75NTR gene network. This study provides a comprehensive analysis and investigation into the current knowledge of p75NTR signaling networks and pathways. These results also identify several genes and their respective protein products as involved in the p75NTR network, which have not previously been clearly studied in this pathway. These results can be used to generate novel hypotheses to gain a greater understanding of p75NTR in acute brain injuries, neurodegenerative diseases and general response to cellular damage.


Subject(s)
Algorithms , Brain Injuries , Data Mining , Gene Regulatory Networks , Metabolic Networks and Pathways , Nerve Tissue Proteins , Neurodegenerative Diseases , Receptors, Nerve Growth Factor , Signal Transduction , Brain Injuries/genetics , Brain Injuries/metabolism , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , PubMed , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
9.
eNeuro ; 7(4)2020.
Article in English | MEDLINE | ID: mdl-32788298

ABSTRACT

NETO2 is an auxiliary subunit for kainate-type glutamate receptors that mediate normal cued fear expression and extinction. Since the amygdala is critical for these functions, we asked whether Neto2-/- mice have compromised amygdala function. We measured the abundance of molecular markers of neuronal maturation and plasticity, parvalbumin-positive (PV+), perineuronal net-positive (PNN+), and double positive (PV+PNN+) cells in the Neto2-/- amygdala. We found that Neto2-/- adult, but not postnatal day (P)23, mice had 7.5% reduction in the fraction of PV+PNN+ cells within the total PNN+ population, and 23.1% reduction in PV staining intensity compared with Neto2+/+ mice, suggesting that PV interneurons in the adult Neto2-/- amygdala remain in an immature state. An immature PV inhibitory network would be predicted to lead to stronger amygdalar excitation. In the amygdala of adult Neto2-/- mice, we identified increased glutamatergic and reduced GABAergic transmission using whole-cell patch-clamp recordings. This was accompanied by increased spine density of thin dendrites in the basal amygdala (BA) compared with Neto2+/+ mice, indicating stronger glutamatergic synapses. Moreover, after fear acquisition Neto2-/- mice had a higher number of c-Fos-positive cells than Neto2+/+ mice in the lateral amygdala (LA), BA, and central amygdala (CE). Altogether, our findings indicate that Neto2 is involved in the maturation of the amygdala PV interneuron network. Our data suggest that this defect, together with other processes influencing amygdala principal neurons, contribute to increased amygdalar excitability, higher fear expression, and delayed extinction in cued fear conditioning, phenotypes that are common in fear-related disorders, including the posttraumatic stress disorder (PTSD).


Subject(s)
Fear , Receptors, Kainic Acid , Amygdala/metabolism , Animals , Interneurons/metabolism , Membrane Proteins , Mice , Parvalbumins/metabolism , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism
10.
Front Behav Neurosci ; 14: 51, 2020.
Article in English | MEDLINE | ID: mdl-32317945

ABSTRACT

The medial prefrontal cortex (mPFC) has been classically defined as the brain region responsible for higher cognitive functions, including the decision-making process. Ample information has been gathered during the last 40 years in an attempt to understand how it works. We now know extensively about the connectivity of this region and its relationship with neuromodulatory ascending projection areas, such as the dorsal raphe nucleus (DRN) or the ventral tegmental area (VTA). Both areas are well-known regulators of the reward-based decision-making process and hence likely to be involved in processes like evidence integration, impulsivity or addiction biology, but also in helping us to predict the valence of our future actions: i.e., what is "good" and what is "bad." Here we propose a hypothesis of a critical period, during which the inputs of the mPFC compete for target innervation, establishing specific prefrontal network configurations in the adult brain. We discuss how these different prefrontal configurations are linked to brain diseases such as addiction or neuropsychiatric disorders, and especially how drug abuse and other events during early life stages might lead to the formation of more vulnerable prefrontal network configurations. Finally, we show different promising pharmacological approaches that, when combined with the appropriate stimuli, will be able to re-establish these functional prefrontocortical configurations during adulthood.

11.
Mol Brain ; 12(1): 107, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31822292

ABSTRACT

Bipolar disorder is a major mental illness characterized by severe swings in mood and activity levels which occur with variable amplitude and frequency. Attempts have been made to identify mood states and biological features associated with mood changes to compensate for current clinical diagnosis, which is mainly based on patients' subjective reports. Here, we used infradian (a cycle > 24 h) cyclic locomotor activity in a mouse model useful for the study of bipolar disorder as a proxy for mood changes. We show that metabolome patterns in peripheral blood could retrospectively predict the locomotor activity levels. We longitudinally monitored locomotor activity in the home cage, and subsequently collected peripheral blood and performed metabolomic analyses. We then constructed cross-validated linear regression models based on blood metabolome patterns to predict locomotor activity levels of individual mice. Our analysis revealed a significant correlation between actual and predicted activity levels, indicative of successful predictions. Pathway analysis of metabolites used for successful predictions showed enrichment in mitochondria metabolism-related terms, such as "Warburg effect" and "citric acid cycle." In addition, we found that peripheral blood metabolome patterns predicted expression levels of genes implicated in bipolar disorder in the hippocampus, a brain region responsible for mood regulation, suggesting that the brain-periphery axis is related to mood-change-associated behaviors. Our results may serve as a basis for predicting individual mood states through blood metabolomics in bipolar disorder and other mood disorders and may provide potential insight into systemic metabolic activity in relation to mood changes.


Subject(s)
Affect , Bipolar Disorder/blood , Bipolar Disorder/metabolism , Metabolome , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Disease Models, Animal , Gene Expression Regulation , Hippocampus/metabolism , Infradian Rhythm/genetics , Male , Mice , Mitochondria/metabolism , Motor Activity/genetics
12.
Mol Brain ; 11(1): 38, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29976232

ABSTRACT

AIM: Maturation abnormalities of the brain cells have been suggested in several neuropsychiatric disorders, including schizophrenia, bipolar disorder, autism spectrum disorders, and epilepsy. In this study, we examined the expression patterns of neuronal maturation markers in the brain of a mouse model of dementia with Lewy body-linked mutant ß-synuclein (ßS), especially in the hippocampus, to explore whether such brain abnormalities occur in neurodegenerative disorders as well. METHODS: Quantitative PCR (qPCR) and immunohistochemical analyses were performed using the hippocampus of 14-month-old P123H ßS transgenic (Tg) mice to evaluate the expression of molecular markers for maturation of dentate granule cells. RESULTS: Based on qPCR results, expression of Tdo2 and Dsp (markers of mature granule cells) was decreased and that of Drd1a (a marker of immature granule cells) was increased in the hippocampus of P123H ßS Tg mice compared to that in wild-type controls. Immunohistochemical analysis revealed decreased expression of mature granule cell markers Calb1 and Gria1, along with increased expression of the microglial marker Iba1, in the hippocampal dentate gyrus region of P123H ßS Tg mice. P123H ßS Tg mice exhibited immature-like neuronal molecular expression patterns and microgliosis in the hippocampus. Pseudo-immaturity of dentate granule cells, associated with neuroinflammation, may be a shared endophenotype in the brains of at least a subgroup of patients with neuropsychiatric disorders and neurodegenerative diseases.


Subject(s)
Dementia/genetics , Hippocampus/metabolism , Hippocampus/pathology , Lewy Bodies/genetics , Lewy Bodies/pathology , Mutation/genetics , beta-Synuclein/genetics , Animals , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Disease Models, Animal , Mice, Transgenic , Phenotype , beta-Synuclein/metabolism
13.
Psychiatry Clin Neurosci ; 72(9): 633-653, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29802758

ABSTRACT

The network hypothesis of depression proposes that mood disorders reflect problems in information processing within particular neural networks. Antidepressants (AD), including selective serotonin reuptake inhibitors (SSRI), function by gradually improving information processing within these networks. AD have been shown to induce a state of juvenile-like plasticity comparable to that observed during developmental critical periods: Such critical-period-like plasticity allows brain networks to better adapt to extrinsic and intrinsic signals. We have coined this drug-induced state of juvenile-like plasticity 'iPlasticity.' A combination of iPlasticity induced by chronic SSRI treatment together with training, rehabilitation, or psychotherapy improves symptoms of neuropsychiatric disorders and issues underlying the developmentally or genetically malfunctioning networks. We have proposed that iPlasticity might be a critical component of AD action. We have demonstrated that iPlasticity occurs in the visual cortex, fear erasure network, extinction of aggression caused by social isolation, and spatial reversal memory in rodent models. Chronic SSRI treatment is known to promote neurogenesis and to cause dematuration of granule cells in the dentate gyrus and of interneurons, especially parvalbumin interneurons enwrapped by perineuronal nets in the prefrontal cortex, visual cortex, and amygdala. Brain-derived neurotrophic factor (BDNF), via its receptor tropomyosin kinase receptor B, is involved in the processes of synaptic plasticity, including neurogenesis, neuronal differentiation, weight of synapses, and gene regulation of synaptic formation. BDNF can be activated by both chronic SSRI treatment and neuronal activity. Accordingly, the BDNF/tropomyosin kinase receptor B pathway is critical for iPlasticity, but further analyses will be needed to provide mechanical insight into the processes of iPlasticity.


Subject(s)
Brain/drug effects , Brain/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Brain-Derived Neurotrophic Factor/physiology , Humans , Neurogenesis/drug effects , Neurogenesis/physiology , Receptor, trkB/physiology
14.
Neuropsychopharmacology ; 43(2): 235-245, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28685757

ABSTRACT

Escalated or abnormal aggression induced by early adverse experiences is a growing issue of social concern and urges the development of effective treatment strategies. Here we report that synergistic interactions between psychosocial and biological factors specifically ameliorate escalated aggression induced by early adverse experiences. Rats reared in isolation from weaning until early adulthood showed abnormal forms of aggression and social deficits that were temporarily ameliorated by re-socialization, but aggression again escalated in a novel environment. We demonstrate that when re-socialization was combined with the antidepressant fluoxetine, which has been shown to reactivate juvenile-like state of plasticity, escalated aggression was greatly attenuated, while neither treatment alone was effective. Early isolation induced a permanent, re-socialization-resistant reduction in Bdnf expression in the amygdala and the infralimbic cortex. Only the combined treatment of fluoxetine and re-socialization was able to recover Bdnf expression via epigenetic regulation. Moreover, the behavior improvement after the combined treatment was dependent on TrkB activity. Combined treatment specifically strengthened the input from the ventral hippocampus to the mPFC, suggesting that this pathway is an important mediator of the beneficial behavioral effects of the combined psychosocial and pharmacological treatment of abnormal aggression. Our findings suggest that synergy between pharmacological induction of plasticity and psychosocial rehabilitation could enhance the efficacy of therapies for pathological aggression.


Subject(s)
Aggression/physiology , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/metabolism , Fluoxetine/pharmacology , Neuronal Plasticity/physiology , Prefrontal Cortex/metabolism , Receptor, trkB/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Signal Transduction/physiology , Social Isolation , Social Learning/physiology , Socialization , Aggression/drug effects , Amygdala/drug effects , Amygdala/metabolism , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/physiology , Fluoxetine/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Receptor, trkB/drug effects , Selective Serotonin Reuptake Inhibitors/administration & dosage , Signal Transduction/drug effects , Social Learning/drug effects
15.
Cell Rep ; 14(12): 2784-96, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-27028761

ABSTRACT

Bipolar disorder, also known as manic-depressive illness, causes swings in mood and activity levels at irregular intervals. Such changes are difficult to predict, and their molecular basis remains unknown. Here, we use infradian (longer than a day) cyclic activity levels in αCaMKII (Camk2a) mutant mice as a proxy for such mood-associated changes. We report that gene-expression patterns in the hippocampal dentate gyrus could retrospectively predict whether the mice were in a state of high or low locomotor activity (LA). Expression of a subset of circadian genes, as well as levels of cAMP and pCREB, possible upstream regulators of circadian genes, were correlated with LA states, suggesting that the intrinsic molecular circuitry changes concomitant with infradian oscillatory LA. Taken together, these findings shed light onto the molecular basis of how irregular biological rhythms and behavior are controlled by the brain.


Subject(s)
Circadian Rhythm/genetics , Locomotion/physiology , Mood Disorders/pathology , Animals , Anxiety , Calcium-Calmodulin-Dependent Protein Kinase Type 2/deficiency , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Depression , Disease Models, Animal , Heterozygote , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Mice , Mice, Knockout , Mood Disorders/metabolism , Transcriptome
16.
Front Cell Neurosci ; 10: 22, 2016.
Article in English | MEDLINE | ID: mdl-26903807

ABSTRACT

Neuronal plasticity peaks during critical periods of postnatal development and is reduced towards adulthood. Recent data suggests that windows of juvenile-like plasticity can be triggered in the adult brain by antidepressant drugs such as Fluoxetine. Although the exact mechanisms of how Fluoxetine promotes such plasticity remains unknown, several studies indicate that inhibitory circuits play an important role. The polysialylated form of the neural cell adhesion molecules (PSA-NCAM) has been suggested to mediate the effects of Fluoxetine and it is expressed in the adult brain by mature interneurons. Moreover, the enzymatic removal of PSA by neuroaminidase-N not only affects the structure of interneurons but also has been shown to play a role in the onset of critical periods during development. We have here used ocular dominance plasticity in the mouse visual cortex as a model to investigate whether removal of PSA might influence the Fluoxetine-induced plasticity. We demonstrate that PSA removal in the adult visual cortex alters neither the baseline ocular dominance, nor the fluoxetine-induced shift in the ocular dominance. We also show that both chronic Fluoxetine treatment and PSA removal independently increase the basal FosB expression in parvalbumin (PV) interneurons in the primary visual cortex. Therefore, our data suggest that although PSA-NCAM regulates inhibitory circuitry, it is not required for the reactivation of juvenile-like plasticity triggered by Fluoxetine.

17.
Cereb Cortex ; 26(3): 1287-94, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26637448

ABSTRACT

Inputs to sensory cortices are known to compete for target innervation through an activity-dependent mechanism during critical periods. To investigate whether this principle also applies to association cortices such as the medial prefrontal cortex (mPFC), we produced a bilateral lesion during early development to the ventral hippocampus (vHC), an input to the mPFC, and analyzed the intensity of the projection from another input, the basolateral amgydala (BLA). We found that axons from the BLA had a higher density of "en passant" boutons in the mPFC of lesioned animals. Furthermore, the density of neurons labeled with retrograde tracers was increased, and neurons projecting from the BLA to the mPFC showed increased expression of FosB. Since neonatal ventral hippocampal lesion has been used as an animal model of schizophrenia, we investigated its effects on behavior and found a negative correlation between the density of retrogradely labeled neurons in the BLA and the reduction of the startle response in the prepulse inhibition test. Our results not only indicate that the inputs from the BLA and the vHC compete for target innervation in the mPFC during postnatal development but also that subsequent abnormal rewiring might underlie the pathophysiology of neuropsychiatric disorders such as schizophrenia.


Subject(s)
Basolateral Nuclear Complex/cytology , Hippocampus/cytology , Neurons/cytology , Prefrontal Cortex/cytology , Animals , Basolateral Nuclear Complex/physiology , Disease Models, Animal , Exploratory Behavior/physiology , Hippocampus/physiology , Hippocampus/physiopathology , Ibotenic Acid , Immunohistochemistry , Microscopy, Confocal , Neural Pathways/cytology , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques , Neurons/physiology , Prefrontal Cortex/physiology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Long-Evans , Sensory Gating/physiology
18.
J Neurosci Res ; 94(1): 74-89, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26389685

ABSTRACT

Cell adhesion molecules play important roles in the development of the nervous system. Among the contactin-associated protein (Caspr; also known as Cntnap) family, which belongs to the neurexin superfamily of proteins, Caspr and Caspr2 are indispensable for the formation and maintenance of myelinated nerves. In contrast, a physiological role for Caspr3 remains to be elucidated. This study examines the expression and localization of Caspr3 in the mouse brain using newly generated Caspr3 antibodies. Caspr3 was expressed abundantly between the first and the second postnatal weeks. During this period, Caspr3 was localized especially to the basal ganglia, including the striatum, external segment of the globus pallidus, and substantia nigra, and no gross abnormalities were apparent in the basal ganglia of Caspr3 knockout mice. In the striatum, Caspr3 was expressed by a subpopulation of medium spiny neurons that constitute the direct and indirect pathways. Caspr3 immunostaining was observed as punctate around the cell bodies as well as in the soma. These Caspr3 signals did not, however, overlap with those of synaptic markers. Our findings suggest that Caspr3 may play an important role in basal ganglia development during early postnatal stages.


Subject(s)
Basal Ganglia/growth & development , Basal Ganglia/metabolism , Gene Expression Regulation, Developmental/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Corpus Striatum/cytology , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tubulin/metabolism , Tyrosine 3-Monooxygenase/metabolism
19.
Int J Dev Neurosci ; 44: 55-64, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25997908

ABSTRACT

The in utero exposure to common chemical stressors, environmental pollutant methylmercury and antidepressant fluoxetine, results in behavioral impairments persistent into adulthood. Modulation of critical periods in brain development may alter proper network formation and lastingly impair brain function. To investigate whether early-life stressors can modulate critical periods, we analyzed the development of parvalbumin (PV) and perineuronal nets (PNNs) in the dentate gyrus and CA1 area of the hippocampus and the basolateral amygdala in mice perinatally exposed to either fluoxetine or methylmercury. The number of PV and PNN neurons, and PV intensity, were analyzed by fluorescent immunohistochemistry at the postnatal ages P17 (ongoing critical period) and P24 (closing critical period). The exposure to fluoxetine did not affect the number of PV cells and PV intensity but decreased PNN formation around the cells at P17 and P24 in all tissues. In contrast, perinatal methylmercury inhibited the development of PV interneurons and PV expression at P17 only, but at P24 these parameters were restored. Methylmercury strongly increased PNN formation from P17 to P24 in the amygdala only. We suggest that perinatal fluoxetine and methylmercury might delay the closure and the onset, respectively, of the critical periods in the amygdala and hippocampus.


Subject(s)
Brain , Fluoxetine/toxicity , Methylmercury Compounds/toxicity , Nerve Net/pathology , Parvalbumins/metabolism , Prenatal Exposure Delayed Effects/pathology , Selective Serotonin Reuptake Inhibitors/toxicity , Age Factors , Animals , Animals, Newborn , Brain/growth & development , Brain/metabolism , Brain/pathology , Cell Count , Extracellular Matrix Proteins/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Nerve Net/metabolism , Pregnancy
20.
Cell Mol Neurobiol ; 35(2): 189-96, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25204460

ABSTRACT

The L-type calcium channel blocker nimodipine improves clinical outcome produced by delayed cortical ischemia or vasospasm associated with subarachnoid hemorrhage. While vasoactive mechanisms are strongly implicated in these therapeutic actions of nimodipine, we sought to test whether nimodipine might also regulate neurotrophic and neuroplastic signaling events associated with TrkB neurotrophin receptor activation. Adult male mice were acutely treated with vehicle or nimodipine (10 mg/kg, s.c., 1.5 h) after which the phosphorylation states of TrkB, cyclic-AMP response element binding protein (CREB), protein kinase B (Akt), extracellular regulated kinase (ERK), mammalian target of rapamycin (mTor) and p70S6 kinase (p70S6k) from prefrontal cortex and hippocampus were assessed. Nimodipine increased the phosphorylation of the TrkB catalytic domain and the phosphoslipase-Cγ1 (PLCγ1) domain, whereas phosphorylation of the TrkB Shc binding site remained unaltered. Nimodipine-induced TrkB phosphorylation was associated with increased phosphorylation levels of Akt and CREB in the prefrontal cortex and the hippocampus whereas phosphorylation of ERK, mTor and p70S6k remained unaltered. Nimodipine-induced TrkB signaling was not associated with changes in BDNF mRNA or protein levels. These nimodipine-induced changes on TrkB signaling mimic those produced by antidepressant drugs and thus propose common mechanisms and long-term functional consequences for the effects of these medications. This work provides a strong basis for investigating the role of TrkB-associated signaling underlying the neuroprotective and neuroplastic effects of nimodipine in translationally relevant animal models of brain trauma or compromised synaptic plasticity.


Subject(s)
Hippocampus/metabolism , Neuronal Plasticity/drug effects , Neuroprotective Agents/metabolism , Nimodipine/pharmacology , Prefrontal Cortex/metabolism , Receptor, trkB/metabolism , Signal Transduction/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Male , Mice, Inbred C57BL , Nimodipine/administration & dosage , Phosphorylation/drug effects , Prefrontal Cortex/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...