Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(45): eadg4800, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37948526

ABSTRACT

A substantial proportion of raphe neurons are glutamatergic. However, little is known about how these glutamatergic neurons modulate the forebrain. We investigated how glutamatergic median raphe nucleus (MRN) input modulates the medial prefrontal cortex (mPFC), a critical component of fear circuitry. We show that vesicular glutamate transporter 3 (VGLUT3)-expressing MRN neurons activate VGLUT3- and somatostatin-expressing neurons in the mPFC. Consistent with this modulation of mPFC GABAergic neurons, activation of MRN (VGLUT3) neurons enhances GABAergic transmission in mPFC pyramidal neurons and attenuates fear memory in female but not male mice. Serotonin plays a key role in MRN (VGLUT3) neuron-mediated GABAergic plasticity in the mPFC. In agreement with these female-specific effects, we observed sex differences in glutamatergic transmission onto MRN (VGLUT3) neurons and in mPFC (VGLUT3) neuron-mediated dual release of glutamate and GABA. Our results demonstrate a cell type-specific modulation of the mPFC by MRN (VGLUT3) neurons and reveal a sex-specific role of this neuromodulation in mPFC synaptic plasticity.


Subject(s)
Raphe Nuclei , Vesicular Glutamate Transport Proteins , Female , Mice , Animals , Male , Vesicular Glutamate Transport Proteins/metabolism , Raphe Nuclei/metabolism , Pyramidal Cells/metabolism , GABAergic Neurons/metabolism , Prefrontal Cortex/metabolism
2.
bioRxiv ; 2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37693545

ABSTRACT

The current understanding of the neuromodulatory role of the median raphe nucleus (MRN) is primarily based on its putative serotonergic output. However, a significant proportion of raphe neurons are glutamatergic. The present study investigated how glutamatergic MRN input modulates the medial prefrontal cortex (mPFC), a critical component of the fear circuitry. Our studies show that VGLUT3-expressing MRN neurons modulate VGLUT3- and somatostatin-expressing neurons in the mPFC. Consistent with this modulation of mPFC GABAergic neurons, activation of MRN (VGLUT3) neurons suppresses mPFC pyramidal neuron activity and attenuates fear memory in female but not male mice. In agreement with these female-specific effects, we observed sex differences in glutamatergic transmission onto MRN (VGLUT3) neurons and mPFC (VGLUT3) neuron-mediated dual release of glutamate and GABA. Thus, our results demonstrate a cell type-specific modulation of the mPFC by MRN (VGLUT3) neurons and reveal a sex-specific role of this neuromodulation in mPFC synaptic plasticity and fear memory.

3.
Cereb Cortex Commun ; 2(1): tgab007, 2021.
Article in English | MEDLINE | ID: mdl-33738453

ABSTRACT

The onset of several neuropsychiatric disorders including anxiety disorders coincides with adolescence. Consistently, threat extinction, which plays a key role in the regulation of anxiety-related behaviors, is diminished during adolescence. Furthermore, this attenuated threat extinction during adolescence is associated with an altered synaptic plasticity in the infralimbic medial prefrontal cortex (IL-mPFC), a brain region critical for threat extinction. However, the mechanism underlying the altered plasticity in the IL-mPFC during adolescence is unclear. Given the purported role of vasoactive intestinal polypeptide expressing interneurons (VIPINs) in disinhibition and hence their potential to affect cortical plasticity, we examined whether VIPINs exhibit an adolescence-specific plasticity in the IL-mPFC. We observed an increase in GABAergic transmission and a decrease in excitability in VIPINs during adolescence. Male mice show a significantly higher VIPIN-pyramidal neuron GABAergic transmission compared with female mice. The observed increase in GABAergic transmission and a decrease in membrane excitability in VIPINs during adolescence could play a role in the altered plasticity in the adolescent IL-mPFC. Furthermore, the suppression of VIPIN-mediated GABAergic transmission in females might be relevant to sex differences in anxiety disorders.

4.
Biol Psychiatry ; 86(9): 682-692, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31235076

ABSTRACT

BACKGROUND: Rodents and humans show an attenuation of fear extinction during adolescence, which coincides with the onset of several psychiatric disorders. Although the ethological relevance and the underlying mechanism are largely unknown, the suppression of fear extinction during adolescence is associated with a diminished plasticity in the glutamatergic neurons of the infralimbic medial prefrontal cortex, a brain region critical for fear extinction. Given the putative effect of synaptic inhibition on glutamatergic neuron activity, we studied whether gamma-aminobutyric acidergic neurons in the infralimbic medial prefrontal cortex are involved in the suppression of fear extinction during adolescence. METHODS: We assessed membrane and synaptic properties in parvalbumin-positive interneurons (PVINs) and somatostatin-positive interneurons (SSTINs) in male preadolescent, adolescent, and adult mice. The effect of fear conditioning and extinction on PVIN-pyramidal neuron and SSTIN-pyramidal neuron synapses in male preadolescent, adolescent, and adult mice was evaluated using an optogenetic approach. RESULTS: The development of the membrane excitability of PVINs is delayed and reaches maturity only by adulthood, while the SSTIN membrane properties are developed early and remain stable during development from preadolescence to adulthood. Although the synaptic inhibition mediated by PVINs undergoes a protracted development, it does not exhibit a fear behavior-specific plasticity. However, the synaptic inhibition mediated by SSTINs undergoes an adolescence-specific enhancement, and this increased inhibition is suppressed by fear learning but is not restored by extinction training. This altered plasticity during adolescence overlapped with a reduction in calcium-permeable glutamate receptors in SSTINs. CONCLUSIONS: The adolescence-specific plasticity in the SSTINs might play a role in fear extinction suppression during adolescence in mice.


Subject(s)
Extinction, Psychological , Interneurons/physiology , Limbic System/growth & development , Neuronal Plasticity , Prefrontal Cortex/growth & development , Animals , Fear , Inhibition, Psychological , Limbic System/cytology , Limbic System/physiology , Male , Mice , Optogenetics , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Synapses/physiology
5.
Neuropsychopharmacology ; 44(10): 1828-1836, 2019 09.
Article in English | MEDLINE | ID: mdl-31005058

ABSTRACT

The medial habenula-interpeduncular nucleus (MHb-IPN) pathway has recently been implicated in the suppression of fear memory. A notable feature of this pathway is the corelease of neurotransmitters and neuropeptides from MHb neurons. Our studies in mice reveal that an activation of substance P-positive dorsomedial habenula (dMHb) neurons results in simultaneous release of glutamate and glycine in the lateral interpeduncular nucleus (LIPN). This glycine receptor activity inhibits an activity-dependent long-lasting potentiation of glutamatergic synapses in LIPN neurons, while substance P enhances this plasticity. An endocannabinoid CB1 receptor-mediated suppression of GABAB receptor activity allows substance P to induce a long-lasting increase in glutamate release in LIPN neurons. Consistent with the substance P-dependent synaptic potentiation in the LIPN, the NK1R in the IPN is involved in fear extinction but not fear conditioning. Thus, our study describes a novel plasticity mechanism in the LIPN and a region-specific role of substance P in fear extinction.


Subject(s)
Glycine/metabolism , Habenula/metabolism , Interpeduncular Nucleus/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Substance P/metabolism , Animals , Electrophysiological Phenomena , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/physiology , Long-Term Potentiation/physiology , Mice , Receptor, Cannabinoid, CB1/metabolism , Receptors, GABA-B/metabolism , Receptors, Neurokinin-1/metabolism , Synaptic Transmission
6.
Synapse ; 73(6): e22090, 2019 06.
Article in English | MEDLINE | ID: mdl-30720888

ABSTRACT

Fear extinction, an inhibitory learning that suppresses a previously learned fear memory, is diminished during adolescence. Earlier studies have shown that this suppressed fear extinction during adolescence involves an altered glutamatergic plasticity in infralimbic medial prefrontal cortical (IL-mPFC) pyramidal neurons. However, it is unclear whether the excitability of IL-mPFC pyramidal neurons plays a role in this development-dependent suppression of fear extinction. Therefore, we examined whether fear conditioning and extinction affect the active and passive membrane properties of IL-mPFC layer 5 pyramidal neurons in preadolescent, adolescent and adult mice. Both preadolescent and adult mice exhibited a bidirectional modulation of the excitability of IL-mPFC layer 5 pyramidal neurons following fear conditioning and extinction, i.e., fear conditioning reduced membrane excitability, whereas fear extinction reversed this effect. However, the fear conditioning-induced suppression of excitability was not reversed in adolescent mice following fear extinction training. Neither fear conditioning nor extinction affected GABAergic transmission in IL-mPFC layer 5 pyramidal neurons, suggesting that GABAergic transmission did not play a role in experience-dependent modulation of neuronal excitability. Our results suggest that the extinction-specific modulation of excitability is impaired during adolescence.


Subject(s)
Extinction, Psychological , Limbic System/growth & development , Neuronal Plasticity , Prefrontal Cortex/growth & development , Animals , Fear , GABAergic Neurons/physiology , Limbic System/cytology , Limbic System/physiology , Male , Mice , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Pyramidal Cells/physiology
7.
Cell Rep ; 20(5): 1111-1122, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28768196

ABSTRACT

The medial habenula-interpeduncular nucleus (MHb-IPN) pathway, which connects the limbic forebrain to the midbrain, has recently been implicated in aversive behaviors. The MHb-IPN circuit is characterized by a unique topographical organization, an excitatory role of GABA, and a prominent co-release of neurotransmitters and neuropeptides. However, little is known about synaptic plasticity in this pathway. An application of a high-frequency stimulation resulted in a long-lasting potentiation of glutamate release in IPN neurons. Our experiments reveal that a Ca2+-permeable AMPA receptor (CPAR)-dependent release of GABA from IPN neurons and a retrograde activation of GABAB receptors on MHb terminals result in a long-lasting enhancement of glutamate release. Strikingly, adolescent IPN neurons lacked CPARs and exhibited an inability to undergo plasticity. In addition, fear conditioning suppressed an activity-dependent potentiation of MHb-IPN synapses, whereas fear extinction reversed this plasticity deficit, suggesting a role of the MHb-IPN synaptic plasticity in the regulation of aversive behaviors.


Subject(s)
Interpeduncular Nucleus/metabolism , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Receptors, GABA-B/metabolism , Synapses/metabolism , Animals , Interpeduncular Nucleus/cytology , Mice , Mice, Transgenic , Receptors, AMPA/genetics , Receptors, GABA-B/genetics , Synapses/genetics
8.
Neuropharmacology ; 112(Pt A): 84-93, 2017 01.
Article in English | MEDLINE | ID: mdl-27378336

ABSTRACT

The Val66Met polymorphism in the brain-derived neurotrophic factor (BDNF) gene disrupts the activity-dependent release of BDNF, which might underlie its involvement in several neuropsychiatric disorders. Consistent with the potential role of regulated release of BDNF in synaptic functions, earlier studies have demonstrated that the BDNF Val66Met polymorphism impairs NMDA receptor-mediated synaptic transmission and plasticity in the hippocampus, the medial prefrontal cortex and the central amygdala. However, it is unknown whether the BDNF Val66Met polymorphism affects synapses in the dorsal striatum, which depends on cortical afferents for BDNF. Electrophysiological experiments revealed an enhanced glutamatergic transmission in the dorsolateral striatum (DLS) of knock-in mice containing the variant polymorphism (BDNFMet/Met) compared to the wild-type (BDNFVal/Val) mice. This increase in glutamatergic transmission is mediated by a potentiation in glutamate release and NMDA receptor transmission in the medium spiny neurons without any alterations in non-NMDA receptor-mediated transmission. We also observed an impairment of synaptic plasticity, both long-term potentiation and depression in the DLS neurons, in BDNFMet/Met mice. Thus, the BDNF Val66Met polymorphism exerts an increase in glutamatergic transmission but impairs synaptic plasticity in the dorsal striatum, which might play a role in its effect on neuropsychiatric symptoms. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Corpus Striatum/physiology , Excitatory Postsynaptic Potentials , Glutamic Acid/physiology , Neuronal Plasticity , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Corpus Striatum/cytology , Dendrites , Electric Stimulation , Gene Knock-In Techniques , Male , Mice , Mice, Transgenic , Neurons/cytology , Polymorphism, Single Nucleotide
9.
Mol Cell Neurosci ; 77: 105-112, 2016 12.
Article in English | MEDLINE | ID: mdl-27793697

ABSTRACT

Of the two major subdivisions of the habenula, the medial and lateral nuclei, the medial habenula is the least understood in terms of synaptic transmission, intrinsic properties and plasticity. The medial habenula (MHb) is composed of glutamatergic neurons which receive the majority of their inputs from the septal region and project predominantly to the interpeduncular nucleus (IPN). To understand the synaptic transmission, we studied both glutamatergic and GABAergic synaptic transmission in the dorsal region of the medial habenula (dMHb). While glutamatergic transmission dominates during early development, an attenuation of glutamatergic transmission and an enhancement of GABAergic transmission occur during development leading into adulthood. Furthermore, as reported previously, GABAA receptor-mediated transmission is excitatory in the adult dMHb, which is consistent with the reduced expression of the K-Cl co-transporter KCC2. Given the potential role of the dMHb in aversive behaviors, we examined whether fear conditioning or exposure to foot shock affects excitability in dMHb neurons. We observed a suppression of the excitability of dMHb neurons in mice that either underwent fear conditioning or were exposed to foot shock. Furthermore, we observed a suppression of GABAergic but not glutamatergic transmission in the dMHb neurons following fear conditioning. These results suggest that aversive experience produces a suppression of the dMHb neuronal activity. Given that the medial habenula is upstream of the median raphe nucleus which is believed to be involved in the negative regulation of aversive memory, the suppression of dMHb neurons following an aversive experience might play a role in strengthening of aversive memories.


Subject(s)
Conditioning, Classical , Habenula/physiology , Neurogenesis , Neuronal Plasticity , Synaptic Transmission , Animals , Fear , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Glutamic Acid/metabolism , Habenula/growth & development , Mice , Mice, Inbred C57BL , gamma-Aminobutyric Acid/metabolism
10.
Sci Rep ; 6: 32553, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27581852

ABSTRACT

The oligomeric amyloid-ß (Aß) peptide is thought to contribute to the subtle amnesic changes in Alzheimer's disease (AD) by causing synaptic dysfunction. Here, we examined the time course of synaptic changes in mouse hippocampal neurons following exposure to Aß42 at picomolar concentrations, mimicking its physiological levels in the brain. We found opposite effects of the peptide with short exposures in the range of minutes enhancing synaptic plasticity, and longer exposures lasting several hours reducing it. The plasticity reduction was concomitant with an increase in the basal frequency of spontaneous neurotransmitter release, a higher basal number of functional presynaptic release sites, and a redistribution of synaptic proteins including the vesicle-associated proteins synapsin I, synaptophysin, and the post-synaptic glutamate receptor I. These synaptic alterations were mediated by cytoskeletal changes involving actin polymerization and p38 mitogen-activated protein kinase. These in vitro findings were confirmed in vivo with short hippocampal infusions of picomolar Aß enhancing contextual memory and prolonged infusions impairing it. Our findings provide a model for initiation of synaptic dysfunction whereby exposure to physiologic levels of Aß for a prolonged period of time causes microstructural changes at the synapse which result in increased transmitter release, failure of synaptic plasticity, and memory loss.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/pharmacology , Memory Disorders/diagnosis , Neuronal Plasticity/drug effects , Neurons/drug effects , Peptide Fragments/pharmacology , Synaptic Transmission/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Animals, Newborn , Disease Models, Animal , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Primary Cell Culture , Protein Multimerization , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Synapses/drug effects , Synapsins/genetics , Synapsins/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Elife ; 52016 06 02.
Article in English | MEDLINE | ID: mdl-27253067

ABSTRACT

Exercise induces beneficial responses in the brain, which is accompanied by an increase in BDNF, a trophic factor associated with cognitive improvement and the alleviation of depression and anxiety. However, the exact mechanisms whereby physical exercise produces an induction in brain Bdnf gene expression are not well understood. While pharmacological doses of HDAC inhibitors exert positive effects on Bdnf gene transcription, the inhibitors represent small molecules that do not occur in vivo. Here, we report that an endogenous molecule released after exercise is capable of inducing key promoters of the Mus musculus Bdnf gene. The metabolite ß-hydroxybutyrate, which increases after prolonged exercise, induces the activities of Bdnf promoters, particularly promoter I, which is activity-dependent. We have discovered that the action of ß-hydroxybutyrate is specifically upon HDAC2 and HDAC3, which act upon selective Bdnf promoters. Moreover, the effects upon hippocampal Bdnf expression were observed after direct ventricular application of ß-hydroxybutyrate. Electrophysiological measurements indicate that ß-hydroxybutyrate causes an increase in neurotransmitter release, which is dependent upon the TrkB receptor. These results reveal an endogenous mechanism to explain how physical exercise leads to the induction of BDNF.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Gene Expression Regulation/drug effects , Acetylation , Animals , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Hippocampus/drug effects , Hippocampus/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylases/chemistry , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neurotransmitter Agents/metabolism , Physical Conditioning, Animal , Receptor, trkB/metabolism
12.
Sci Transl Med ; 8(344): 344ra85, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27334260

ABSTRACT

Accumulating evidence has shown that repeated exposure to general anesthesia during critical stages of brain development results in long-lasting behavioral deficits later in life. To date, there has been no effective treatment to mitigate the neurotoxic effects of anesthesia on brain development. By performing calcium imaging in the mouse motor cortex, we show that ketamine anesthesia causes a marked and prolonged reduction in neuronal activity during the period of post-anesthesia recovery. Administration of the AMPAkine drug CX546 [1-(1,4-benzodioxan-6-ylcarbonyl)piperidine] to potentiate AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor activity during emergence from anesthesia in mice enhances neuronal activity and prevents long-term motor learning deficits induced by repeated neonatal anesthesia. In addition, we show that CX546 administration also ameliorates various synaptic deficits induced by anesthesia, including reductions in synaptic expression of NMDA (N-methyl-d-aspartate) and AMPA receptor subunits, motor training-evoked neuronal activity, and dendritic spine remodeling associated with motor learning. Together, our results indicate that pharmacologically enhancing neuronal activity during the post-anesthesia recovery period could effectively reduce the adverse effects of early-life anesthesia.


Subject(s)
Anesthesia/adverse effects , Receptors, AMPA/metabolism , Animals , Blotting, Western , Dioxoles/pharmacology , Learning/drug effects , Mice , N-Methylaspartate/metabolism , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Piperidines/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism
13.
Nat Commun ; 7: 11475, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27215672

ABSTRACT

Fear can be highly adaptive in promoting survival, yet it can also be detrimental when it persists long after a threat has passed. Flexibility of the fear response may be most advantageous during adolescence when animals are prone to explore novel, potentially threatening environments. Two opposing adolescent fear-related behaviours-diminished extinction of cued fear and suppressed expression of contextual fear-may serve this purpose, but the neural basis underlying these changes is unknown. Using microprisms to image prefrontal cortical spine maturation across development, we identify dynamic BLA-hippocampal-mPFC circuit reorganization associated with these behavioural shifts. Exploiting this sensitive period of neural development, we modified existing behavioural interventions in an age-specific manner to attenuate adolescent fear memories persistently into adulthood. These findings identify novel strategies that leverage dynamic neurodevelopmental changes during adolescence with the potential to extinguish pathological fears implicated in anxiety and stress-related disorders.


Subject(s)
Behavior, Animal/physiology , Fear/psychology , Memory/physiology , Neural Pathways/physiology , Prefrontal Cortex/physiology , Age Factors , Animals , Conditioning, Psychological/physiology , Cues , Extinction, Psychological/physiology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Spinal Cord/physiology
14.
Cell Rep ; 13(9): 1747-56, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655895

ABSTRACT

One of the cardinal features of neural development and adult plasticity is the contribution of activity-dependent signaling pathways. However, the interrelationships between different activity-dependent genes are not well understood. The immediate early gene neuronal-activity-regulated pentraxin (NPTX2 or Narp) encodes a protein that has been associated with excitatory synaptogenesis, AMPA receptor aggregation, and the onset of critical periods. Here, we show that Narp is a direct transcriptional target of brain-derived neurotrophic factor (BDNF), another highly regulated activity-dependent gene involved in synaptic plasticity. Unexpectedly, Narp is bidirectionally regulated by BDNF. Acute BDNF withdrawal results in downregulation of Narp, whereas transcription of Narp is greatly enhanced by BDNF. Furthermore, our results show that BDNF directly regulates Narp to mediate glutamatergic transmission and mossy fiber plasticity. Hence, Narp serves as a significant epistatic target of BDNF to regulate synaptic plasticity during periods of dynamic activity.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , C-Reactive Protein/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , C-Reactive Protein/antagonists & inhibitors , C-Reactive Protein/genetics , Cells, Cultured , Dactinomycin/pharmacology , Down-Regulation/drug effects , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phospholipase C gamma/metabolism , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Synapses/metabolism , Transcriptional Activation/drug effects
15.
Neuropsychopharmacology ; 40(9): 2269-77, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25786582

ABSTRACT

Fear expression is mediated by an activation of the centromedial amygdala (CEm), the major output nucleus of the amygdaloid complex. Consistently, fear extinction is associated with an increased synaptic inhibition as well as a suppression of the excitability of the CEm neurons. However, little is known about the role of CEm glutamatergic synapses in fear regulation and anxiety-like behaviors. The BDNF Val66Met, a single-nucleotide polymorphism in the human BDNF gene, impairs fear extinction and leads to anxiety-like symptoms. To determine whether the BDNF Val66Met polymorphism affects the CEm excitatory synapses, we examined basal glutamatergic synaptic transmission and plasticity in the CEm neurons of BDNF Val66Met knock-in (BDNF(Met/Met)) mice. The BDNF Val66Met single-nucleotide polymorphism exerted an opposite effect on non-NMDA and NMDA receptor transmission with a potentiation of the former and a suppression of the latter. In addition, the decay time of NMDA currents was decreased in BDNF(Met/Met) mice, suggesting a modification of NMDA receptor subunit composition. Unlike the wild-type mice that exhibited a potentiation of non-NMDA receptor transmission following fear conditioning and a depotentiation upon fear extinction, BDNF(Met/Met) mice failed to show this experience-dependent synaptic plasticity in the CEm neurons. Our results suggest that the elevated non-NMDA receptor transmission, the suppression of NMDA receptor transmission, and an impairment of synaptic plasticity in the CEm neurons might contribute to the fear extinction deficit and increased anxiety-like symptoms in BDNF Val66Met carriers.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Central Amygdaloid Nucleus/cytology , Glutamic Acid/metabolism , Neurons/physiology , Polymorphism, Single Nucleotide/genetics , Synapses/genetics , Animals , Conditioning, Classical/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Extinction, Psychological/drug effects , Fear/physiology , In Vitro Techniques , Male , Methionine/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Methylaspartate/pharmacology , Neurons/drug effects , Piperidines/pharmacology , Valine/genetics
16.
Neuropsychopharmacology ; 39(9): 2086-94, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24608267

ABSTRACT

Recent studies suggest that low endogenous estradiol might be a susceptibility factor for anxiety and trauma-related disorders in women. Consistently, fear extinction, a form of inhibitory learning critical for the management of anxiety symptoms, is positively correlated with endogenous estradiol levels. To understand the synaptic basis of the effect of endogenous estradiol on fear extinction, we studied glutamatergic transmission and plasticity in the infralimbic medial prefrontal cortex (IL-mPFC), a brain region crucial for the regulation of fear extinction. Diestrus mice (low estradiol) exhibited a higher basal glutamatergic transmission compared with proestrus mice (high estradiol). Synaptic plasticity was also regulated by endogenous estradiol, which favored synaptic potentiation in a GluN2B-dependent manner. Activation of estrogen receptor ß (ERß) but not ERα rescued synaptic potentiation in diestrus mice by enhancing GluN2B-mediated NMDA receptor transmission. Our results suggest that both endogenous estradiol and ERß activation facilitate the ability of the IL-mPFC synapses to undergo potentiation, a mechanism necessary for the regulation of fear extinction.


Subject(s)
Estradiol/metabolism , Extinction, Psychological/physiology , Fear/physiology , Prefrontal Cortex/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Diestrus/physiology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Estrogen Receptor beta/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Extinction, Psychological/drug effects , Fear/drug effects , Female , Glutamic Acid/metabolism , Male , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Prefrontal Cortex/drug effects , Proestrus/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/drug effects , Synaptic Transmission/drug effects
17.
Mol Cell Neurosci ; 58: 76-84, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24321452

ABSTRACT

The dystrobrevin binding protein (DTNBP) 1 gene has emerged over the last decade as a potential susceptibility locus for schizophrenia. While no causative mutations have been found, reduced expression of the encoded protein, dysbindin, was reported in patients. Dysbindin likely plays a role in the neuronal trafficking of proteins including receptors. One important pathway suspected to be affected in schizophrenia is the fast excitatory glutamatergic transmission mediated by AMPA receptors. Here, we investigated excitatory synaptic transmission and plasticity in hippocampal neurons from dysbindin-deficient sandy mice bred on the DBA/2J strain. In cultured neurons an enhancement of AMPAR responses was observed. The enhancement of AMPAR-mediated transmission was confirmed in hippocampal CA3-CA1 synapses, and was not associated with changes in the expression of GluA1-4 subunits or an increase in GluR2-lacking receptor complexes. Lastly, an enhancement in LTP was also found in these mice. These data provide compelling evidence that dysbindin, a widely suspected susceptibility protein in schizophrenia, is important for AMPAR-mediated synaptic transmission and plasticity in the developing hippocampus.


Subject(s)
Carrier Proteins/metabolism , Long-Term Potentiation , Neurons/metabolism , Receptors, AMPA/metabolism , Synaptic Transmission , Animals , Carrier Proteins/genetics , Cells, Cultured , Dysbindin , Dystrophin-Associated Proteins , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Hippocampus/physiology , Mice , Mice, Inbred DBA , Neurons/physiology , Receptors, AMPA/genetics , Schizophrenia/genetics
18.
Neuropharmacology ; 76 Pt C: 684-95, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23747574

ABSTRACT

Brain derived neurotrophic factor (BDNF), a neurotrophin essential for nervous system development and synaptic plasticity, has been found to have a significant influence on affective behaviors. The notion that an impairment in BDNF signaling might be involved in affective disorders is originated primarily from the opposing effects of antidepressants and stress on BDNF signaling. Antidepressants enhance BDNF signaling and synaptic plasticity. On the other hand, negative environmental factors such as severe stress suppress BDNF signaling, impair synaptic activity and increase susceptibility to affective disorders. Postmortem studies provided strong support for decreased BDNF signaling in depressive disorders. Remarkably, studies in humans with a single nucleotide polymorphism in the BDNF gene, the BDNF Val66Met which affects regulated release of BDNF, showed profound deficits in hippocampal and prefrontal cortical (PFC) plasticity and cognitive behaviors. BDNF regulates synaptic mechanisms responsible for various cognitive processes including attenuation of aversive memories, a key process in the regulation of affective behaviors. The unique role of BDNF in cognitive and affective behaviors suggests that cognitive deficits due to altered BDNF signaling might underlie affective disorders. Understanding how BDNF modulates synapses in neural circuits relevant to affective behaviors, particularly the medial prefrontal cortical (mPFC)-hippocampus-amygdala pathway, and its interaction with development, sex, and environmental risk factors might shed light on potential therapeutic targets for affective disorders. This article is part of the Special Issue entitled 'BDNF Regulation of Synaptic Structure, Function, and Plasticity'.


Subject(s)
Affect/physiology , Brain-Derived Neurotrophic Factor/metabolism , Mood Disorders/metabolism , Neuronal Plasticity/physiology , Synapses/metabolism , Animals , Humans , Mood Disorders/pathology
19.
Cell ; 155(7): 1596-609, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24360280

ABSTRACT

Microglia are the resident macrophages of the CNS, and their functions have been extensively studied in various brain pathologies. The physiological roles of microglia in brain plasticity and function, however, remain unclear. To address this question, we generated CX3CR1(CreER) mice expressing tamoxifen-inducible Cre recombinase that allow for specific manipulation of gene function in microglia. Using CX3CR1(CreER) to drive diphtheria toxin receptor expression in microglia, we found that microglia could be specifically depleted from the brain upon diphtheria toxin administration. Mice depleted of microglia showed deficits in multiple learning tasks and a significant reduction in motor-learning-dependent synapse formation. Furthermore, Cre-dependent removal of brain-derived neurotrophic factor (BDNF) from microglia largely recapitulated the effects of microglia depletion. Microglial BDNF increases neuronal tropomyosin-related kinase receptor B phosphorylation, a key mediator of synaptic plasticity. Together, our findings reveal that microglia serve important physiological functions in learning and memory by promoting learning-related synapse formation through BDNF signaling.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Learning/physiology , Microglia/physiology , Synapses , Animals , CX3C Chemokine Receptor 1 , Gene Expression , Mice , Microglia/cytology , Neuronal Plasticity , Protein Kinases/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Signal Transduction
20.
J Neurosci ; 33(14): 6123-32, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23554493

ABSTRACT

The mechanisms by which natural rewards such as sugar affect synaptic transmission and behavior are largely unexplored. Here, we investigate regulation of nucleus accumbens synapses by sucrose intake. Previous studies have shown that AMPA receptor (AMPAR) trafficking is a major mechanism for regulating synaptic strength, and that in vitro, trafficking of AMPARs containing the GluA1 subunit takes place by a two-step mechanism involving extrasynaptic and then synaptic receptor transport. We report that in rat, repeated daily ingestion of a 25% sucrose solution transiently elevated spontaneous locomotion and potentiated accumbens core synapses through incorporation of Ca(2+)-permeable AMPA receptors (CPARs), which are GluA1-containing, GluA2-lacking AMPARs. Electrophysiological, biochemical, and quantitative electron microscopy studies revealed that sucrose training (7 d) induced a stable (>24 h) intraspinous GluA1 population, and that in these rats a single sucrose stimulus rapidly (5 min) but transiently (<24 h) elevated GluA1 at extrasynaptic sites. CPARs and dopamine D1 receptors were required in vivo for elevated locomotion after sucrose ingestion. Significantly, a 7 d protocol of daily ingestion of a 3% solution of saccharin, a noncaloric sweetener, induced synaptic GluA1 similarly to 25% sucrose ingestion. These findings identify multistep GluA1 trafficking, previously described in vitro, as a mechanism for acute regulation of synaptic transmission in vivo by a natural orosensory reward. Trafficking is stimulated by a chemosensory pathway that is not dependent on the caloric value of sucrose.


Subject(s)
Neurons/metabolism , Receptors, AMPA/metabolism , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Animals , Carrier Proteins , Conditioning, Operant/physiology , Dopamine beta-Hydroxylase/metabolism , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Locomotion/physiology , Male , Microscopy, Electron, Transmission , Neurons/drug effects , Nucleus Accumbens/cytology , Phosphoproteins/metabolism , Post-Synaptic Density/metabolism , Post-Synaptic Density/ultrastructure , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Synaptosomes/metabolism , Synaptosomes/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...