Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
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
2.
Neurobiol Stress ; 19: 100460, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35734023

ABSTRACT

This manuscript is dedicated to the memory of Bruce S. McEwen, to commemorate the impact he had on how we understand stress and neuronal plasticity, and the profound influence he exerted on our scientific careers. The focus of this review is the impact of stressors on inhibitory circuits, particularly those of the limbic system, but we also consider other regions affected by these adverse experiences. We revise the effects of acute and chronic stress during different stages of development and lifespan, taking into account the influence of the sex of the animals. We review first the influence of stress on the physiology of inhibitory neurons and on the expression of molecules related directly to GABAergic neurotransmission, and then focus on specific interneuron subpopulations, particularly on parvalbumin and somatostatin expressing cells. Then we analyze the effects of stress on molecules and structures related to the plasticity of inhibitory neurons: the polysialylated form of the neural cell adhesion molecule and perineuronal nets. Finally, we review the potential of antidepressants or environmental manipulations to revert the effects of stress on inhibitory circuits.

3.
Article in English | MEDLINE | ID: mdl-35066055

ABSTRACT

The effects of intense stressors can last a long time and may lead to the development of psychiatric disorders, including posttraumatic stress disorder. The basolateral amygdala (BLA) plays a critical role in these diseases and is extremely sensitive to stress. Here, we explored in male and female mice the long-term (35 days) impact of a 24-h restraint stress on BLA circuitry. We used Thy1-YFP mice to discriminate 2 subpopulations of excitatory neurons, which participate in "Fear-On" (Thy1-) and "Fear-Off" (Thy1+) circuits. The stress decreased the density of parvalbumin (PV) + inhibitory neurons in both sexes but did not alter their dendritic complexity. We also analyzed the perisomatic input of basket interneurons on Thy1+ and Thy1- neurons, finding sex dependent effects. In males, we did not find alterations in the density of PV+ puncta or in that of cannabinoid receptor 1 (CB1R) + puncta from cholecystokinin+ basket cells. By contrast, in females we found increased the density of PV+ puncta on Thy1+ neurons and reduced on the Thy1- neurons. This adverse experience also reduced in the long term the density of CB1R+ puncta both on Thy1+ and Thy1- cells in females. The expression of the activity marker FosB was not altered in PV+ interneurons and in Thy1+ neurons of stressed animals. The density of perineuronal nets, a specialized region of the extracellular matrix, which covers particularly PV+ interneurons and regulates their connectivity, was increased by stress in male mice. Our findings indicate that a single stressful event can produce long-term alterations in the inhibitory circuits of the BLA, especially on PV+ neurons and their plasticity, and that there is a differential impact depending on the sex and the fear-related circuits involved.


Subject(s)
Basolateral Nuclear Complex/metabolism , Fear/physiology , Immunohistochemistry , Interneurons/metabolism , Restraint, Physical/physiology , Animals , Female , Male , Mice , Mice, Transgenic , Parvalbumins/metabolism , Sex Factors
4.
Neuroendocrinology ; 112(1): 51-67, 2022.
Article in English | MEDLINE | ID: mdl-33550289

ABSTRACT

The estrous cycle is caused by the changing concentration of ovarian hormones, particularly 17ß-estradiol, a hormone whose effect on excitatory circuits has been extensively reported. However, fewer studies have tried to elucidate how this cycle, or this hormone, affects the plasticity of inhibitory networks and the structure of interneurons. Among these cells, somatostatin-expressing O-LM neurons of the hippocampus are especially interesting. They have a role in the modulation of theta oscillations, and they receive direct input from the entorhinal cortex, which place them in the center of hippocampal function. In this study, we report that the expression of polysialylated form of the neural cell adhesion molecule (PSA-NCAM) in the hippocampus, a molecule involved in the plasticity of somatostatin-expressing interneurons in the adult brain, fluctuated through the different stages of the estrous cycle. Likewise, these stages and the expression of PSA-NCAM affected the density of dendritic spines of O-LM cells. We also describe that 17ß-estradiol replacement of adult ovariectomized female mice caused an increase in the perisomatic inhibitory puncta in O-LM interneurons as well as an increase in their axonal bouton density. Interestingly, this treatment also induced a decrease in their dendritic spine density, specifically in O-LM interneurons lacking PSA-NCAM expression. Finally, using an ex vivo real-time assay with entorhinal-hippocampal organotypic cultures, we show that this hormone decreased the dynamics in spinogenesis, altogether highlighting the modulatory effect that 17ß-estradiol has on inhibitory circuits.


Subject(s)
Entorhinal Cortex/physiology , Estradiol/metabolism , Hippocampus/physiology , Interneurons/physiology , Nerve Net/physiology , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Animals , Cells, Cultured , Dendritic Spines/physiology , Entorhinal Cortex/cytology , Entorhinal Cortex/metabolism , Female , Hippocampus/cytology , Hippocampus/metabolism , Interneurons/metabolism , Mice , Mice, Transgenic , Nerve Net/metabolism , Ovariectomy , Somatostatin/metabolism
5.
Transl Psychiatry ; 10(1): 177, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32488050

ABSTRACT

The prefrontal cortex (PFC) continues its development during adolescence and alterations in its structure and function, particularly of inhibitory networks, have been detected in schizophrenic patients. Since cannabis use during adolescence is a risk factor for this disease, our main objective was to investigate whether THC administration during this period might exacerbate alterations in prefrontocortical inhibitory networks in mice subjected to a perinatal injection of MK801 and postweaning social isolation. This double-hit model (DHM) combines a neurodevelopmental manipulation and the exposure to an aversive experience during early life; previous work has shown that DHM mice have important alterations in the structure and connectivity of PFC interneurons. In the present study we found that DHM had reductions in prepulse inhibition of the startle reflex (PPI), GAD67 expression and cingulate 1 cortex volume. Interestingly, THC by itself induced increases in PPI and decreases in the dendritic complexity of somatostatin expressing interneurons. Both THC and DHM reduced the density of parvalbumin expressing cells surrounded by perineuronal nets and, when combined, they disrupted the ratio between the density of puncta expressing excitatory and inhibitory markers. Our results support previous work showing alterations in parameters involving interneurons in similar animal models and schizophrenic patients. THC treatment does not modify further these parameters, but changes some others related also to interneurons and their plasticity, in some cases in the opposite direction to those induced by the DHM, suggesting a protective effect.


Subject(s)
Dronabinol , Receptors, N-Methyl-D-Aspartate , Adolescent , Adult , Animals , Dronabinol/pharmacology , Humans , Interneurons/metabolism , Mice , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Social Isolation
6.
J Neurosci ; 40(26): 5008-5018, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32457072

ABSTRACT

Parvalbumin-expressing (PV+) interneurons play a key role in the maturation and synchronization of cortical circuitry and alterations in these inhibitory neurons, especially in the medial prefrontal cortex (mPFC), have been found in different psychiatric disorders. The formation of perineuronal nets (PNNs) around many of these interneurons at the end of the critical periods reduces their plasticity and sets their connectivity. Consequently, the presence of PNNs must have an important impact on the synaptic input and the physiology of PV+ cells. In the present study, we have found that in adult male mice, prefrontocortical PV+ cells surrounded by PNNs show higher density of perisomatic excitatory and inhibitory puncta, longer axonal initial segments (AISs), and higher PV expression when compared with PV+ cells lacking PNNs. In order to better understand the impact of PNNs on the connectivity and physiology of PV+ interneurons in the mPFC, we have digested enzymatically these structures and have found a decrease in the density of inhibitory puncta on their perisomatic region but not on the PV+ perisomatic puncta on pyramidal neurons. Moreover, extracellular recordings show that the digestion of PNNs induces a decrease in γ activity, an oscillation dependent on PV+ cells, in the mPFC of anesthetized mice. Our results suggest that the presence of PNNs enwrapping PV+ cells regulates their inhibitory input and has a potent influence on their activity. These results may be relevant for psychiatric research, given the alterations in PNNs, PV+ interneurons and their physiology described in different mental disorders.SIGNIFICANCE STATEMENT Parvalbumin-expressing (PV+) interneurons are surrounded by specializations of the extracellular matrix, the perineuronal nets (PNNs). PNNs regulate the development and plasticity of PV+ cells and, consequently, their presence must influence their synaptic input and physiology. We have found, in the adult prefrontal cortex (PFC), substantial differences in the structure and connectivity of PV+ interneurons depending on the presence of PNNs. The depletion of PNNs from the PFC has also a potent effect on the connectivity of PV+ cells and on neural oscillations that depend on these cells. These findings are relevant to understand the role of PNNs in the adult brain and in certain psychiatric disorders in which alterations in PNNs and PV+ interneurons have been described.


Subject(s)
Extracellular Matrix , Gamma Rhythm/physiology , Interneurons/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Parvalbumins
7.
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.

8.
J Comp Neurol ; 528(8): 1349-1366, 2020 06.
Article in English | MEDLINE | ID: mdl-31792992

ABSTRACT

Several experimental manipulations, including visual deprivation, are able to induce critical period-like plasticity in the visual cortex of adult animals. In this regard, many studies have analyzed the effects of dark exposure in adult animals, but still little is known about the role of interneurons and plasticity-related molecules on such mechanisms. In this study, we analyzed the effects of 10 days of dark exposure on the connectivity and structure of interneurons, both in the primary visual cortex and in the rest of cerebral regions implicated in the transmission of visual stimulus. We found that this environmental manipulation induces changes in the expression of synaptic molecules throughout the visual pathway and in the structure of interneurons in the primary visual cortex. Moreover, we found altered expression in the polysialylated form of the neural cell adhesion molecule and in perineuronal nets surrounding parvalbumin expressing interneurons, suggesting that these plasticity-related molecules may be involved in the changes produced by dark exposure. Together, our findings indicate that dark exposure produces an important alteration of inhibitory circuits and molecules related to their plasticity, not only in the visual cortex but throughout the visual pathway.


Subject(s)
Darkness/adverse effects , Interneurons/metabolism , Nerve Net/metabolism , Neuronal Plasticity/physiology , Sensory Deprivation/physiology , Visual Cortex/metabolism , Age Factors , Animals , Interneurons/chemistry , Male , Mice , Mice, Transgenic , Nerve Net/chemistry , Nerve Net/growth & development , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Visual Cortex/chemistry , Visual Cortex/growth & development
9.
Int J Bipolar Disord ; 7(1): 24, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31728775

ABSTRACT

BACKGROUND: Alterations in the structure and physiology of interneurons in the prefrontal cortex (PFC) are important factors in the etiopathology of different psychiatric disorders. Among the interneuronal subpopulations, parvalbumin (PV) expressing cells appear to be specially affected. Interestingly, during development and adulthood the connectivity of these interneurons is regulated by the presence of perineuronal nets (PNNs), specialized regions of the extracellular matrix, which are frequently surrounding PV expressing neurons. Previous reports have found anomalies in the density of PNNs in the PFC of schizophrenic patients. However, although some studies have described alterations in PNNs in some extracortical regions of bipolar disorder patients, there are no studies focusing on the prefrontocortical PNNs of bipolar or major depression patients. For this reason, we have analyzed the density of PNNs in post-mortem sections of the dorsolateral PFC (DLPFC) from the Stanley Neuropathology Consortium, which includes controls, schizophrenia, bipolar and major depression patients. RESULTS: We have not observed differences in the distribution of PV+ cells or PNNs, or in the percentage of PV+ interneurons surrounded by PNNs. The density of PV+ interneurons was similar in all the experimental groups, but there was a significantly lower density of PNNs in the DLPFC of bipolar disorder patients and a tendency towards a decrease in schizophrenic patients. No differences were found when evaluating the density of PV+ cells surrounded by PNNs. Interestingly, when assessing the influence of demographic data, we found an inverse correlation between the density of PNNs and the presence of psychosis. CONCLUSIONS: The present results point to prefrontocortical PNNs and their role in the regulation of neuronal plasticity as putative players in the etiopathology of bipolar disorder and schizophrenia. Our findings also suggest a link between these specialized regions of the extracellular matrix and the presence of psychosis.

10.
Front Cell Neurosci ; 13: 197, 2019.
Article in English | MEDLINE | ID: mdl-31133813

ABSTRACT

Chronic stress has an important impact on the adult brain. However, most of the knowledge on its effects is focused on principal neurons and less on inhibitory neurons. Consequently, recent reports have begun to describe stress-induced alterations in the structure, connectivity and neurochemistry of interneurons. Some of these changes appear to be mediated by certain molecules particularly associated to interneurons, such as the polysialylated form of the neural cell adhesion molecule (PSA-NCAM) and components of the perineuronal nets (PNN), specialized regions of the extracellular matrix. These plasticity-related molecules modulate interneuronal structure and connectivity, particularly of parvalbumin expressing basket interneurons, both during development and adult life. These inhibitory neurons are specially affected after chronic stress and in some stress-related disorders, in which the expression of PSA-NCAM and certain components of PNN are also altered. For these reasons we have decided to study PSA-NCAM, PNN and parvalbumin expressing interneurons after 10 days of chronic restraint stress, a time point in which its behavioral consequences are starting to appear. We have focused initially on the medial prefrontal cortex (mPFC), basolateral amygdala (BLA) and hippocampus, regions affected by stress and stress-related psychiatric diseases, but we have also explored the habenula and the thalamic reticular nucleus (TRN) due to the important presence of PNN and their relationship with certain disorders. PSA-NCAM expression was increased by stress in the stratum lacunosum-moleculare of CA1. Increases in parvalbumin immunoreactive cells were detected in the mPFC and the BLA, but were not accompanied by increases in the number of parvalbumin expressing perisomatic puncta on the somata of principal neurons. The number of PNN was also increased in the mPFC and the habenula, although habenular PNN were not associated to parvalbumin cells. Increased expression of parvalbumin and components of PNN were also detected in the TRN after chronic restraint stress, revealing for the first time substantial effects on this region. Our study shows that, even a short chronic stress protocol, can induce consistent changes in interneuronal plasticity-related molecules in cortical and extracortical regions, which may represent initial responses of inhibitory circuits to counteract the effects of this aversive experience.

11.
Neuroscience ; 386: 205-213, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30018016

ABSTRACT

Although the precise mechanism of action of antidepressant drugs remains elusive, the neuroplastic hypothesis has gained acceptance during the last two decades. Several studies have shown that treatment with antidepressants such as Fluoxetine is associated with enhanced plasticity in control animals, especially in regions such as the visual cortex, the hippocampus and the medial prefrontal cortex. More recently, the basolateral amygdala has been shown to be affected by Fluoxetine leading to a reopening of critical period-like plasticity in the fear and aggression circuits. One of the key elements triggering this type of brain plasticity are inhibitory networks, especially parvalbumin interneurons. However, recent work on fast-acting antidepressants has shown also an important role for somatostatin interneurons. Here we show that Fluoxetine reorganizes inhibitory circuits through increased expression of the plasticity-related molecule PSA-NCAM which regulates interneuronal structure and connectivity. In addition, we demonstrate that treatment with this antidepressant alters the structure of somatostatin interneurons both at the level of dendritic spines and of axonal en passant boutons. Our findings suggest that new strategies targeting somatostatin interneuron activity might help us to better understand depression and the action of antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Basolateral Nuclear Complex/metabolism , Fluoxetine/pharmacology , Interneurons/metabolism , Somatostatin/metabolism , Animals , Basolateral Nuclear Complex/drug effects , Interneurons/drug effects , Male , Mice , Mice, Transgenic , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology
12.
Heliyon ; 4(6): e00669, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30003163

ABSTRACT

The quantification of the expression of different molecules is a key question in both basic and applied sciences. While protein quantification through molecular techniques leads to the loss of spatial information and resolution, immunohistochemistry is usually associated with time-consuming image analysis and human bias. In addition, the scarce automatic software analysis is often proprietary and expensive and relies on a fixed threshold binarization. Here we describe and share a set of macros ready for automated fluorescence analysis of large batches of fixed tissue samples using FIJI/ImageJ. The quantification of the molecules of interest are based on an automatic threshold analysis of immunofluorescence images to automatically identify the top brightest structures of each image. These macros measure several parameters commonly quantified in basic neuroscience research, such as neuropil density and fluorescence intensity of synaptic puncta, perisomatic innervation and col-localization of different molecules and analysis of the neurochemical phenotype of neuronal subpopulations. In addition, these same macro functions can be easily modified to improve similar analysis of fluorescent probes in human biopsies for diagnostic purposes based on the expression patterns of several molecules.

13.
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
14.
Sci Rep ; 7(1): 7811, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798343

ABSTRACT

A brief burst-suppressing isoflurane anesthesia has been shown to rapidly alleviate symptoms of depression in a subset of patients, but the neurobiological basis of these observations remains obscure. We show that a single isoflurane anesthesia produces antidepressant-like behavioural effects in the learned helplessness paradigm and regulates molecular events implicated in the mechanism of action of rapid-acting antidepressant ketamine: activation of brain-derived neurotrophic factor (BDNF) receptor TrkB, facilitation of mammalian target of rapamycin (mTOR) signaling pathway and inhibition of glycogen synthase kinase 3ß (GSK3ß). Moreover, isoflurane affected neuronal plasticity by facilitating long-term potentiation in the hippocampus. We also found that isoflurane increased activity of the parvalbumin interneurons, and facilitated GABAergic transmission in wild type mice but not in transgenic mice with reduced TrkB expression in parvalbumin interneurons. Our findings strengthen the role of TrkB signaling in the antidepressant responses and encourage further evaluation of isoflurane as a rapid-acting antidepressant devoid of the psychotomimetic effects and abuse potential of ketamine.


Subject(s)
Antidepressive Agents/administration & dosage , Hippocampus/physiology , Isoflurane/administration & dosage , Receptor, trkB/metabolism , Animals , Antidepressive Agents/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Helplessness, Learned , Hippocampus/drug effects , Hippocampus/metabolism , Isoflurane/pharmacology , Ketamine/pharmacology , Long-Term Potentiation , Male , Mice , Parvalbumins/metabolism , Rats , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
15.
Front Cell Neurosci ; 11: 166, 2017.
Article in English | MEDLINE | ID: mdl-28659763

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) are present in both pyramidal neurons and interneurons of the hippocampus. These receptors play an important role in the adult structural plasticity of excitatory neurons, but their impact on the remodeling of interneurons is unknown. Among hippocampal interneurons, somatostatin-expressing cells located in the stratum oriens are of special interest because of their functional importance and structural characteristics: they display dendritic spines, which change density in response to different stimuli. In order to understand the role of NMDARs on the structural plasticity of these interneurons, we have injected acutely MK-801, an NMDAR antagonist, to adult mice which constitutively express enhanced green fluorescent protein (EGFP) in these cells. We have behaviorally tested the animals, confirming effects of the drug on locomotion and anxiety-related behaviors. NMDARs were expressed in the somata and dendritic spines of somatostatin-expressing interneurons. Twenty-four hours after the injection, the density of spines did not vary, but we found a significant increase in the density of their en passant boutons (EPB). We have also used entorhino-hippocampal organotypic cultures to study these interneurons in real-time. There was a rapid decrease in the apparition rate of spines after MK-801 administration, which persisted for 24 h and returned to basal levels afterwards. A similar reversible decrease was detected in spine density. Our results show that both spines and axons of interneurons can undergo remodeling and highlight NMDARs as regulators of this plasticity. These results are specially relevant given the importance of all these players on hippocampal physiology and the etiopathology of certain psychiatric disorders.

16.
Front Cell Neurosci ; 10: 65, 2016.
Article in English | MEDLINE | ID: mdl-27013976

ABSTRACT

Reelin, a glycoprotein expressed by Cajal-Retzius neurons throughout the marginal layer of developing neocortex, has been extensively shown to play an important role during brain development, guiding neuronal migration and detachment from radial glia. During the adult life, however, many studies have associated Reelin expression to enhanced neuronal plasticity. Although its mechanism of action in the adult brain remains mostly unknown, Reelin is expressed mainly by a subset of mature interneurons. Here, we confirm the described phenotype of this subpopulation in the adult neocortex. We show that these mature interneurons, although being in close proximity, lack polysialylated neural cell adhesion molecule (PSA-NCAM) expression, a molecule expressed by a subpopulation of mature interneurons, related to brain development and involved in neuronal plasticity of the adult brain as well. However, in the layer II of Piriform cortex there is a high density of cells expressing Reelin whose neurochemical phenotype and connectivity has not been described before. Interestingly, in close proximity to these Reelin expressing cells there is a numerous subpopulation of immature neurons expressing PSA-NCAM and doublecortin (DCX) in this layer of the Piriform cortex. Here, we show that Reelin cells express the neuronal marker Neuronal Nuclei (NeuN), but however the majority of neurons lack markers of mature excitatory or inhibitory neurons. A detail analysis of its morphology indicates these that some of these cells might correspond to semilunar neurons. Interestingly, we found that the majority of these cells express T-box brain 1 (TBR-1) a transcription factor found not only in post-mitotic neurons that differentiate to glutamatergic excitatory neurons but also in Cajal-Retzius cells. We suggest that the function of these Reelin expressing cells might be similar to that of the Cajal-Retzius cells during development, having a role in the maintenance of the immature phenotype of the PSA-NCAM/DCX neurons through its receptors apolipoprotein E receptor 2 (ApoER2) and very low density lipoprotein receptor (VLDLR) in the Piriform cortex layer II during adulthood.

17.
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.

18.
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
19.
Int J Neuropsychopharmacol ; 17(10): 1635-46, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24786752

ABSTRACT

Novel hypotheses suggest that antidepressants, such as the selective serotonin reuptake inhibitor fluoxetine, induce neuronal structural plasticity, resembling that of the juvenile brain, although the underlying mechanisms of this reopening of the critical periods still remain unclear. However, recent studies suggest that inhibitory networks play an important role in this structural plasticity induced by fluoxetine. For this reason we have analysed the effects of a chronic fluoxetine treatment in the hippocampus and medial prefrontal cortex (mPFC) of transgenic mice displaying eGFP labelled interneurons. We have found an increase in the expression of molecules related to critical period plasticity, such as the polysialylated form of the neural cell adhesion molecule (PSA-NCAM), GAD67/65 and synaptophysin, as well as a reduction in the number of parvalbumin expressing interneurons surrounded by perineuronal nets. We have also described a trend towards decrease in the perisomatic inhibitory puncta on pyramidal neurons in the mPFC and an increase in the density of inhibitory puncta on eGFP interneurons. Finally, we have found that chronic fluoxetine treatment affects the structure of interneurons in the mPFC, increasing their dendritic spine density. The present study provides evidence indicating that fluoxetine promotes structural changes in the inhibitory neurons of the adult cerebral cortex, probably through alterations in plasticity-related molecules of neurons or the extracellular matrix surrounding them, which are present in interneurons and are known to be crucial for the development of the critical periods of plasticity in the juvenile brain.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Cerebral Cortex/cytology , Fluoxetine/pharmacology , Interneurons/drug effects , Nerve Net/drug effects , Neuronal Plasticity/drug effects , Animals , Cell Count , Dendritic Spines/drug effects , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Interneurons/cytology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Sialic Acids/metabolism , Time Factors , Vesicular Glutamate Transport Protein 1/metabolism
20.
Neuropsychopharmacology ; 39(5): 1148-58, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24213355

ABSTRACT

Neuroligins (NLGNs) are cell adhesion molecules that are important for proper synaptic formation and functioning, and are critical regulators of the balance between neural excitation/inhibition (E/I). Mutations in NLGNs have been linked to psychiatric disorders in humans involving social dysfunction and are related to similar abnormalities in animal models. Chronic stress increases the likelihood for affective disorders and has been shown to induce changes in neural structure and function in different brain regions, with the hippocampus being highly vulnerable to stress. Previous studies have shown evidence of chronic stress-induced changes in the neural E/I balance in the hippocampus. Therefore, we hypothesized that chronic restraint stress would lead to reduced hippocampal NLGN-2 levels, in association with alterations in social behavior. We found that rats submitted to chronic restraint stress in adulthood display reduced sociability and increased aggression. This occurs along with a reduction of NLGN-2, but not NLGN-1 expression (as shown with western blot, immunohistochemistry, and electron microscopy analyses), throughout the hippocampus and detectable in different layers of the CA1, CA3, and DG subfields. Furthermore, using synthetic peptides that comprise sequences in either NLGN-1 (neurolide-1) or NLGN-2 (neurolide-2) involved in the interaction with their presynaptic partner neurexin (NRXN)-1, intra-hippocampal administration of neurolide-2 led also to reduced sociability and increased aggression. These results highlight hippocampal NLGN-2 as a key molecular substrate regulating social behaviors and underscore NLGNs as promising targets for the development of novel drugs for the treatment of dysfunctional social behaviors.


Subject(s)
Aggression/physiology , Cell Adhesion Molecules, Neuronal/metabolism , Hippocampus/physiopathology , Nerve Tissue Proteins/metabolism , Social Behavior , Stress, Psychological/physiopathology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Chronic Disease , Corticosterone/blood , Hippocampus/pathology , Male , Nerve Tissue Proteins/genetics , Neurites/physiology , Neurons/physiology , Organ Size , Peptides/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Cell Surface/metabolism , Restraint, Physical , Stress, Psychological/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...