Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Proc Natl Acad Sci U S A ; 121(31): e2407472121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39047038

ABSTRACT

The integrated stress response (ISR), a pivotal protein homeostasis network, plays a critical role in the formation of long-term memory (LTM). The precise mechanism by which the ISR controls LTM is not well understood. Here, we report insights into how the ISR modulates the mnemonic process by using targeted deletion of the activating transcription factor 4 (ATF4), a key downstream effector of the ISR, in various neuronal and non-neuronal cell types. We found that the removal of ATF4 from forebrain excitatory neurons (but not from inhibitory neurons, cholinergic neurons, or astrocytes) enhances LTM formation. Furthermore, the deletion of ATF4 in excitatory neurons lowers the threshold for the induction of long-term potentiation, a cellular model for LTM. Transcriptomic and proteomic analyses revealed that ATF4 deletion in excitatory neurons leads to upregulation of components of oxidative phosphorylation pathways, which are critical for ATP production. Thus, we conclude that ATF4 functions as a memory repressor selectively within excitatory neurons.


Subject(s)
Activating Transcription Factor 4 , Memory, Long-Term , Neurons , Animals , Mice , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Astrocytes/metabolism , Long-Term Potentiation , Memory, Long-Term/physiology , Mice, Knockout , Neurons/metabolism , Prosencephalon/metabolism , Male
2.
Alzheimers Dement ; 20(8): 5398-5410, 2024 08.
Article in English | MEDLINE | ID: mdl-38934107

ABSTRACT

INTRODUCTION: Impaired brain protein synthesis, synaptic plasticity, and memory are major hallmarks of Alzheimer's disease (AD). The ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) has been shown to modulate protein synthesis, but its effects on memory in AD models remain elusive. METHODS: We investigated the effects of HNK on hippocampal protein synthesis, long-term potentiation (LTP), and memory in AD mouse models. RESULTS: HNK activated extracellular signal-regulated kinase 1/2 (ERK1/2), mechanistic target of rapamycin (mTOR), and p70S6 kinase 1 (S6K1)/ribosomal protein S6 signaling pathways. Treatment with HNK rescued hippocampal LTP and memory deficits in amyloid-ß oligomers (AßO)-infused mice in an ERK1/2-dependent manner. Treatment with HNK further corrected aberrant transcription, LTP and memory in aged APP/PS1 mice. DISCUSSION: Our findings demonstrate that HNK induces signaling and transcriptional responses that correct synaptic and memory deficits in AD mice. These results raise the prospect that HNK could serve as a therapeutic approach in AD. HIGHLIGHTS: The ketamine metabolite HNK activates hippocampal ERK/mTOR/S6 signaling pathways. HNK corrects hippocampal synaptic and memory defects in two mouse models of AD. Rescue of synaptic and memory impairments by HNK depends on ERK signaling. HNK corrects aberrant transcriptional signatures in APP/PS1 mice.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Hippocampus , Ketamine , Mice, Transgenic , Neuronal Plasticity , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Ketamine/analogs & derivatives , Ketamine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Neuronal Plasticity/drug effects , Mice , Long-Term Potentiation/drug effects , Amyloid beta-Peptides/metabolism , Protein Biosynthesis/drug effects , TOR Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Memory/drug effects , Male , Memory Disorders/drug therapy , Mice, Inbred C57BL , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Presenilin-1/genetics , Humans
3.
Mol Brain ; 17(1): 35, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858726

ABSTRACT

The brain responds to experience through modulation of synaptic transmission, that is synaptic plasticity. An increase in the strength of synaptic transmission is manifested as long-term potentiation (LTP), while a decrease in the strength of synaptic transmission is expressed as long-term depression (LTD). Most of the studies of synaptic plasticity have been carried out by induction via electrophysiological stimulation. It is largely unknown in which behavioural tasks such synaptic plasticity occurs. Moreover, some stimuli can induce both LTP and LTD, thus making it difficult to separately study the different forms of synaptic plasticity. Two studies have shown that an aversive memory task - inhibitory avoidance learning and contextual fear conditioning - physiologically and selectively induce LTP and an LTP-like molecular change, respectively, in the hippocampus in vivo. Here, we show that a non-aversive behavioural task - exploration of new space - physiologically and selectively elicits a biochemical change in the hippocampus that is a hallmark of LTP. Specifically, we found that exploration of new space induces an increase in the phosphorylation of GluA1(Ser831), without affecting the phosphorylation of GluA1(Ser845), which are biomarkers of early-LTP and not NMDAR-mediated LTD. We also show that exploration of new space engenders the phosphorylation of the translational regulator S6K and the expression of Arc, which are features of electrophysiologically-induced late-LTP in the hippocampus. Therefore, our results show that exploration of new space is a novel non-aversive behavioural paradigm that elicits molecular changes in vivo that are analogous to those occurring during early- and late-LTP, but not during NMDAR-mediated LTD.


Subject(s)
Cytoskeletal Proteins , Hippocampus , Long-Term Potentiation , Nerve Tissue Proteins , Receptors, AMPA , Animals , Long-Term Potentiation/physiology , Phosphorylation , Hippocampus/metabolism , Hippocampus/physiology , Receptors, AMPA/metabolism , Male , Nerve Tissue Proteins/metabolism , Cytoskeletal Proteins/metabolism , Exploratory Behavior/physiology , Serine/metabolism
4.
Proc Natl Acad Sci U S A ; 120(49): e2308671120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38015848

ABSTRACT

Activation of neuronal protein synthesis upon learning is critical for the formation of long-term memory. Here, we report that learning in the contextual fear conditioning paradigm engenders a decrease in eIF2α (eukaryotic translation initiation factor 2) phosphorylation in astrocytes in the hippocampal CA1 region, which promotes protein synthesis. Genetic reduction of eIF2α phosphorylation in hippocampal astrocytes enhanced contextual and spatial memory and lowered the threshold for the induction of long-lasting plasticity by modulating synaptic transmission. Thus, learning-induced dephosphorylation of eIF2α in astrocytes bolsters hippocampal synaptic plasticity and consolidation of long-term memories.


Subject(s)
Astrocytes , Long-Term Potentiation , Long-Term Potentiation/physiology , Neuronal Plasticity/genetics , Hippocampus/physiology , Protein Biosynthesis , CA1 Region, Hippocampal , Memory, Long-Term/physiology
5.
Mol Brain ; 16(1): 55, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37400913

ABSTRACT

Plasticity of principal cells and inhibitory interneurons underlies hippocampal memory. Bidirectional modulation of somatostatin cell mTORC1 activity, a crucial translational control mechanism in synaptic plasticity, causes parallel changes in hippocampal CA1 somatostatin interneuron (SOM-IN) long-term potentiation and hippocampus-dependent memory, indicating a key role in learning. However, SOM-IN activity changes and behavioral correlates during learning, and the role of mTORC1 in these processes, remain ill-defined. To address these questions, we used two-photon Ca2+ imaging from SOM-INs during a virtual reality goal-directed spatial memory task in head-fixed control mice (SOM-IRES-Cre mice) or in mice with conditional knockout of Rptor (SOM-Rptor-KO mice) to block mTORC1 activity in SOM-INs. We found that control mice learn the task, but SOM-Raptor-KO mice exhibit a deficit. Also, SOM-IN Ca2+ activity became increasingly related to reward during learning in control mice but not in SOM-Rptor-KO mice. Four types of SOM-IN activity patterns related to reward location were observed, "reward off sustained", "reward off transient", "reward on sustained" and "reward on transient", and these responses showed reorganization after reward relocation in control but not SOM-Rptor-KO mice. Thus, SOM-INs develop mTORC1-dependent reward- related activity during learning. This coding may bi-directionally interact with pyramidal cells and other structures to represent and consolidate reward location.


Subject(s)
Hippocampus , Interneurons , Mice , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Interneurons/metabolism , Hippocampus/metabolism , Somatostatin/metabolism , Reward
6.
Neuron ; 111(19): 3028-3040.e6, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37473758

ABSTRACT

Dysregulation of protein synthesis is one of the key mechanisms underlying autism spectrum disorder (ASD). However, the role of a major pathway controlling protein synthesis, the integrated stress response (ISR), in ASD remains poorly understood. Here, we demonstrate that the main arm of the ISR, eIF2α phosphorylation (p-eIF2α), is suppressed in excitatory, but not inhibitory, neurons in a mouse model of fragile X syndrome (FXS; Fmr1-/y). We further show that the decrease in p-eIF2α is mediated via activation of mTORC1. Genetic reduction of p-eIF2α only in excitatory neurons is sufficient to increase general protein synthesis and cause autism-like behavior. In Fmr1-/y mice, restoration of p-eIF2α solely in excitatory neurons reverses elevated protein synthesis and rescues autism-related phenotypes. Thus, we reveal a previously unknown causal relationship between excitatory neuron-specific translational control via the ISR pathway, general protein synthesis, and core phenotypes reminiscent of autism in a mouse model of FXS.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Fragile X Syndrome , Animals , Mice , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Mental Retardation Protein/genetics , Neurons/metabolism , Phenotype , Mice, Knockout , Disease Models, Animal
7.
Front Cell Dev Biol ; 11: 1205112, 2023.
Article in English | MEDLINE | ID: mdl-37293130

ABSTRACT

Tuberous sclerosis complex (TSC) is a rare monogenic disorder co-diagnosed with high rates of autism and is caused by loss of function mutations in the TSC1 or TSC2 genes. A key pathway hyperactivated in TSC is the mammalian/mechanistic target of rapamycin complex 1 (mTORC1), which regulates cap-dependent mRNA translation. We previously demonstrated that exaggerated cap-dependent translation leads to autism-related phenotypes and increased mRNA translation and protein expression of Neuroligin 1 (Nlgn1) in mice. Inhibition of Nlgn1 expression reversed social behavior deficits in mice with increased cap-dependent translation. Herein, we report elevated translation of Nlgn1 mRNA and an increase in its protein expression. Genetic or pharmacological inhibition of Nlgn1 expression in Tsc2 +/- mice rescued impaired hippocampal mGluR-LTD, contextual discrimination and social behavior deficits in Tsc2 +/- mice, without correcting mTORC1 hyperactivation. Thus, we demonstrate that reduction of Nlgn1 expression in Tsc2 +/- mice is a new therapeutic strategy for TSC and potentially other neurodevelopmental disorders.

8.
eNeuro ; 10(5)2023 05.
Article in English | MEDLINE | ID: mdl-37072176

ABSTRACT

SYNGAP1 haploinsufficiency in humans causes intellectual disability (ID). SYNGAP1 is highly expressed in cortical excitatory neurons and, reducing its expression in mice accelerates the maturation of excitatory synapses during sensitive developmental periods, restricts the critical period window for plasticity, and impairs cognition. However, its specific role in interneurons remains largely undetermined. In this study, we investigated the effects of conditional Syngap1 disruption in medial ganglionic eminence (MGE)-derived interneurons on hippocampal interneuron firing properties and excitatory synaptic inputs, as well as on pyramidal cell synaptic inhibition and synaptic integration. We show that conditional Syngap1 disruption in MGE-derived interneurons results in cell-specific impairment of firing properties of hippocampal Nkx2.1 fast-spiking interneurons, with enhancement of their AMPA receptor (AMPAR)-mediated excitatory synaptic inputs but compromised short-term plasticity. In contrast, regular-spiking Nkx2.1 interneurons are largely unaffected. These changes are associated with impaired pyramidal cell synaptic inhibition and enhanced summation of excitatory responses. Unexpectedly, we found that the Syngap1flox allele used in this study contains inverted loxP sites and that its targeted recombination in MGE-derived interneurons induces some cell loss during embryonic development and the reversible inversion of the sequence flanked by the loxP sites in postmitotic cells. Together, these results suggest that Syngap1 plays a role in cell-specific regulation of hippocampal interneuron function and inhibition of pyramidal cells in mice. However, because of our finding that the Syngap1flox allele used in this study contains inverted loxP sites, it will be important to further investigate interneuron function using a different Syngap1 conditional allele.


Subject(s)
Interneurons , Pyramidal Cells , Humans , Mice , Animals , Mice, Transgenic , Interneurons/physiology , Pyramidal Cells/physiology , Hippocampus/metabolism , Recombination, Genetic , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
9.
Mol Brain ; 16(1): 9, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36650535

ABSTRACT

The consolidation of learned information into long-lasting memories requires the strengthening of synaptic connections through de novo protein synthesis. Translation initiation factors play a cardinal role in gating the production of new proteins thereby regulating memory formation. Both positive and negative regulators of translation play a critical role in learning and memory consolidation. The eukaryotic initiation factor 4E (eIF4E) homologous protein (4EHP, encoded by the gene Eif4e2) is a pivotal negative regulator of translation but its role in learning and memory is unknown. To address this gap in knowledge, we generated excitatory (glutamatergic: CaMKIIα-positive) and inhibitory (GABAergic: GAD65-positive) conditional knockout mice for 4EHP, which were analyzed in various behavioral memory tasks. Knockout of 4EHP in Camk2a-expressing neurons (4EHP-cKOexc) did not impact long-term memory in either contextual fear conditioning or Morris water maze tasks. Similarly, long-term contextual fear memory was not altered in Gad2-directed 4EHP knockout mice (4EHP-cKOinh). However, when subjected to a short-term T-maze working memory task, both mouse models exhibited impaired cognition. We therefore tested the hypothesis that de novo protein synthesis plays a direct role in working memory. We discovered that phosphorylation of ribosomal protein S6, a measure of mTORC1 activity, is dramatically reduced in the CA1 hippocampus of 4EHP-cKOexc mice. Consistently, genetic reduction of mTORC1 activity in either excitatory or inhibitory neurons was sufficient to impair working memory. Taken together, these findings indicate that translational control by 4EHP and mTORC1 in both excitatory and inhibitory neurons are necessary for working memory.


Subject(s)
Eukaryotic Initiation Factor-4E , Learning , Memory, Short-Term , Animals , Mice , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout , RNA Cap-Binding Proteins/metabolism , Eukaryotic Initiation Factor-4E/metabolism
10.
J Clin Invest ; 133(2)2023 01 17.
Article in English | MEDLINE | ID: mdl-36394958

ABSTRACT

Repeated or prolonged, but not short-term, general anesthesia during the early postnatal period causes long-lasting impairments in memory formation in various species. The mechanisms underlying long-lasting impairment in cognitive function are poorly understood. Here, we show that repeated general anesthesia in postnatal mice induces preferential apoptosis and subsequent loss of parvalbumin-positive inhibitory interneurons in the hippocampus. Each parvalbumin interneuron controls the activity of multiple pyramidal excitatory neurons, thereby regulating neuronal circuits and memory consolidation. Preventing the loss of parvalbumin neurons by deleting a proapoptotic protein, mitochondrial anchored protein ligase (MAPL), selectively in parvalbumin neurons rescued anesthesia-induced deficits in pyramidal cell inhibition and hippocampus-dependent long-term memory. Conversely, partial depletion of parvalbumin neurons in neonates was sufficient to engender long-lasting memory impairment. Thus, loss of parvalbumin interneurons in postnatal mice following repeated general anesthesia critically contributes to memory deficits in adulthood.


Subject(s)
Anesthesia , Parvalbumins , Mice , Animals , Parvalbumins/genetics , Parvalbumins/metabolism , Interneurons/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Hippocampus/metabolism , Memory Disorders/chemically induced , Memory Disorders/genetics , Memory Disorders/metabolism
11.
Mol Brain ; 15(1): 101, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36544185

ABSTRACT

Hippocampus-dependent learning and memory originate from long-term synaptic changes in hippocampal networks. The activity of CA1 somatostatin interneurons (SOM-INs) during aversive stimulation is necessary for contextual fear memory formation. In addition, mTORC1-dependent long-term potentiation (LTP) of SOM-IN excitatory input synapses from local pyramidal cells (PC-SOM synapses) contributes to the consolidation of fear motivated spatial and contextual memories. Although, it remains unknown if SOM-IN activity and LTP are necessary and sufficient for novelty motivated spatial episodic memory such as the object location memory, and if so when it is required. Here we use optogenetics to examine whether dorsal CA1 SOM-IN activity and LTP are sufficient to regulate object location memory. First, we found that silencing SOM-INs during object location learning impaired memory. Second, optogenetic induction of PC-SOM synapse LTP (TBSopto) given 30 min before object location training, resulted in facilitation of memory. However, in mice with mTORC1 pathway genetically inactivated in SOM-INs, which blocks PC-SOM synapse LTP, TBSopto failed to facilitate object location memory. Our results indicate that SOM-IN activity is necessary during object location learning and that optogenetic induction of PC-SOM synapse LTP is sufficient to facilitate consolidation of object location memory. Thus, hippocampal somatostatin interneuron activity is required for object location learning, a hippocampus-dependent form of novelty motivated spatial learning that is facilitated by plasticity at PC-SOM synapses.


Subject(s)
Long-Term Potentiation , Mechanistic Target of Rapamycin Complex 1 , Spatial Learning , Animals , Mice , Hippocampus/metabolism , Interneurons/metabolism , Long-Term Potentiation/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Somatostatin/metabolism , Spatial Learning/physiology , Synapses/metabolism
12.
Mol Brain ; 15(1): 81, 2022 09 19.
Article in English | MEDLINE | ID: mdl-36123709

ABSTRACT

Somatostatin-expressing interneurons (SOM-INs) are a major subpopulation of GABAergic cells in CA1 hippocampus that receive excitation from pyramidal cells (PCs) and provide feedback control of synaptic inputs onto PC dendrites. Excitatory synapses from PCs onto SOM-INs (PC-SOM synapses) exhibit long-term potentiation (LTP) mediated by type 1a metabotropic glutamate receptors (mGluR1a). LTP at PC-SOM synapses translates in lasting regulation of metaplasticity of entorhinal and CA3 synaptic inputs on PCs and contributes to hippocampus-dependent learning. A persistent form of PC-SOM synapse LTP lasting hours is prevented by blockers of transcription and translation, and a more transient form of PC-SOM synapse LTP lasting tens of minutes requires mTORC1-signaling, suggesting an involvement of protein synthesis. However, the role of protein synthesis in these forms of plasticity has not been directly demonstrated. Here we use the SUrface SEnsing of Translation (SUnSET) assay of protein synthesis to directly show that the induction protocols for both forms of LTP at PC-SOM synapses stimulate protein synthesis in SOM-INs. Moreover, protein synthesis stimulated by persistent LTP induction was prevented in mice with a SOM-IN conditional knock-out of Raptor, an essential component of mTORC1, indicating a critical role of mTORC1 in the control of translation in PC-SOM synapse plasticity. Moreover, protein synthesis induced by both forms of LTP may share common mechanisms as transient LTP induction occluded further stimulation of protein synthesis by persistent LTP induction. Our findings highlight a crucial role of protein synthesis and its control by mTORC1 in SOM-INs that is important for hippocampus-dependent memory function.


Subject(s)
Optogenetics , Receptors, Metabotropic Glutamate , Animals , Hippocampus/metabolism , Interneurons/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/metabolism , Somatostatin/metabolism
14.
Mol Brain ; 15(1): 56, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35715811

ABSTRACT

Hippocampal CA1 parvalbumin-expressing interneurons (PV INs) play a central role in controlling principal cell activity and orchestrating network oscillations. PV INs receive excitatory inputs from CA3 Schaffer collaterals and local CA1 pyramidal cells, and they provide perisomatic inhibition. Schaffer collateral excitatory synapses onto PV INs express Hebbian and anti-Hebbian types of long-term potentiation (LTP), as well as elicit LTP of intrinsic excitability (LTPIE). LTPIE requires the activation of type 5 metabotropic glutamate receptors (mGluR5) and is mediated by downregulation of potassium channels Kv1.1. It is sensitive to rapamycin and thus may involve activation of the mammalian target of rapamycin complex 1 (mTORC1). LTPIE facilitates PV INs recruitment in CA1 and maintains an excitatory-inhibitory balance. Impaired CA1 PV INs activity or LTP affects network oscillations and memory. However, whether LTPIE in PV INs plays a role in hippocampus-dependent memory remains unknown. Here, we used conditional deletion of the obligatory component of mTORC1, the Regulatory-Associated Protein of mTOR (Raptor), to directly manipulate mTORC1 in PV INs. We found that homozygous, but not heterozygous, conditional knock-out of Rptor resulted in a decrease in CA1 PV INs of mTORC1 signaling via its downstream effector S6 phosphorylation assessed by immunofluorescence. In whole-cell recordings from hippocampal slices, repetitive firing of CA1 PV INs was impaired in mice with either homozygous or heterozygous conditional knock-out of Rptor. High frequency stimulation of Schaffer collateral inputs that induce LTPIE in PV INs of control mice failed to do so in mice with either heterozygous or homozygous conditional knock-out of Rptor in PV INs. At the behavioral level, mice with homozygous or heterozygous conditional knock-out of Rptor showed similar long-term contextual fear memory or contextual fear memory discrimination relative to control mice. Thus, mTORC1 activity in CA1 PV INs regulates repetitive firing and LTPIE but not consolidation of long-term contextual fear memory and context discrimination. Our results indicate that mTORC1 plays cell-specific roles in synaptic plasticity of hippocampal inhibitory interneurons that are differentially involved in hippocampus-dependent learning and memory.


Subject(s)
CA1 Region, Hippocampal , Fear , Hippocampus , Interneurons , Long-Term Potentiation , Mechanistic Target of Rapamycin Complex 1 , Memory , Parvalbumins , Animals , CA1 Region, Hippocampal/metabolism , Fear/physiology , Hippocampus/metabolism , Interneurons/metabolism , Long-Term Potentiation/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Memory/physiology , Mice , Parvalbumins/metabolism , Synapses/metabolism
15.
iScience ; 25(5): 104259, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35521524

ABSTRACT

Hippocampal somatostatin (SOM) cells are dendrite-projecting inhibitory interneurons. CA1 SOM cells receive major excitatory inputs from pyramidal cells (PC-SOM synapses) which show mGluR1a- and mTORC1-mediated long-term potentiation (LTP). PC-SOM synapse LTP contributes to CA1 network metaplasticity and memory consolidation, but whether it is sufficient to regulate these processes remains unknown. Here we used optogenetic stimulation of CA1 pyramidal cells and whole-cell recordings in slices to show that optogenetic theta-burst stimulation (TBSopto) produces LTP at PC-SOM synapses. At the network level, we found that TBSopto differentially regulates metaplasticity of pyramidal cell inputs: enhancing LTP at Schaffer collateral synapses and depressing LTP at temporo-ammonic synapses. At the behavioral level, we uncovered that in vivo TBSopto regulates learning-induced LTP at PC-SOM synapses, as well as contextual fear memory. Thus, LTP of PC-SOM synapses is a long-term feedback mechanism controlling pyramidal cell synaptic plasticity, sufficient to regulate memory consolidation.

16.
Mol Brain ; 14(1): 130, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34429141

ABSTRACT

Somatostatin-expressing interneurons (SOM-INs) are a major subpopulation of GABAergic cells in CA1 hippocampus that receive excitation from pyramidal cells (PCs), and, in turn, provide feedback inhibition onto PC dendrites. Excitatory synapses onto SOM-INs show a Hebbian long-term potentiation (LTP) mediated by type 1a metabotropic glutamate receptors (mGluR1a) that is implicated in hippocampus-dependent learning. The neuropeptide somatostatin (SST) is also critical for hippocampal long-term synaptic plasticity, as well as learning and memory. SST effects on hippocampal PCs are well documented, but its actions on inhibitory interneurons remain largely undetermined. In the present work, we investigate the involvement of SST in long-term potentiation of CA1 SOM-IN excitatory synapses using pharmacological approaches targeting the somatostatinergic system and whole cell recordings in slices from transgenic mice expressing eYFP in SOM-INs. We report that application of exogenous SST14 induces long-term potentiation of excitatory postsynaptic potentials in SOM-INs via somatostatin type 1-5 receptors (SST1-5Rs) but does not affect synapses of PC or parvalbumin-expressing interneurons. Hebbian LTP in SOM-INs was prevented by inhibition of SSTRs and by depletion of SST by cysteamine treatment, suggesting a critical role of endogenous SST in LTP. LTP of SOM-IN excitatory synapses induced by SST14 was independent of NMDAR and mGluR1a, activity-dependent, and prevented by blocking GABAA receptor function. Our results indicate that endogenous SST may contribute to Hebbian LTP at excitatory synapses of SOM-INs by controlling GABAA inhibition, uncovering a novel role for SST in regulating long-term synaptic plasticity in somatostatinergic cells that may be important for hippocampus-dependent memory processes.


Subject(s)
CA1 Region, Hippocampal/drug effects , Excitatory Postsynaptic Potentials/drug effects , GABAergic Neurons/drug effects , Interneurons/drug effects , Long-Term Potentiation/drug effects , Somatostatin/physiology , Synapses/drug effects , Animals , Bacterial Proteins , Cysteamine/pharmacology , Female , GABA-A Receptor Antagonists/pharmacology , GABAergic Neurons/metabolism , Gene Knock-In Techniques , Genes, Reporter , Humans , Interneurons/metabolism , Luminescent Proteins , Male , Memory/physiology , Mice , Mice, Transgenic , Peptides, Cyclic/pharmacology , Receptors, Metabotropic Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Somatostatin/drug effects , Receptors, Somatostatin/physiology , Somatostatin/pharmacology , Synapses/physiology
17.
Front Neural Circuits ; 15: 687558, 2021.
Article in English | MEDLINE | ID: mdl-34149368

ABSTRACT

A distinctive feature of the hippocampal structure is the diversity of inhibitory interneurons. These complex inhibitory interconnections largely contribute to the tight modulation of hippocampal circuitry, as well as to the formation and coordination of neuronal assemblies underlying learning and memory. Inhibitory interneurons provide more than a simple transitory inhibition of hippocampal principal cells (PCs). The synaptic plasticity of inhibitory neurons provides long-lasting changes in the hippocampal network and is a key component of memory formation. The dendrite targeting interneurons expressing the peptide somatostatin (SOM) are particularly interesting in this regard because they display unique long-lasting synaptic changes leading to metaplastic regulation of hippocampal networks. In this article, we examine the actions of the neuropeptide SOM on hippocampal cells, synaptic plasticity, learning, and memory. We address the different subtypes of hippocampal SOM interneurons. We describe the long-term synaptic plasticity that takes place at the excitatory synapses of SOM interneurons, its singular induction and expression mechanisms, as well as the consequences of these changes on the hippocampal network, learning, and memory. We also review evidence that astrocytes provide cell-specific dynamic regulation of inhibition of PC dendrites by SOM interneurons. Finally, we cover how, in mouse models of Alzheimer's disease (AD), dysfunction of plasticity of SOM interneuron excitatory synapses may also contribute to cognitive impairments in brain disorders.


Subject(s)
Interneurons , Somatostatin , Animals , Hippocampus/metabolism , Interneurons/metabolism , Mice , Neuronal Plasticity , Somatostatin/metabolism , Synapses/metabolism
18.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34156977

ABSTRACT

The majority of patients affected with lysosomal storage disorders (LSD) exhibit neurological symptoms. For mucopolysaccharidosis type IIIC (MPSIIIC), the major burdens are progressive and severe neuropsychiatric problems and dementia, primarily thought to stem from neurodegeneration. Using the MPSIIIC mouse model, we studied whether clinical manifestations preceding massive neurodegeneration arise from synaptic dysfunction. Reduced levels or abnormal distribution of multiple synaptic proteins were revealed in cultured hippocampal and CA1 pyramidal MPSIIIC neurons. These defects were rescued by virus-mediated gene correction. Dendritic spines were reduced in pyramidal neurons of mouse models of MPSIIIC and other (Tay-Sachs, sialidosis) LSD as early as at P10. MPSIIIC neurons also presented alterations in frequency and amplitude of miniature excitatory and inhibitory postsynaptic currents, sparse synaptic vesicles, reduced postsynaptic densities, disorganized microtubule networks, and partially impaired axonal transport of synaptic proteins. Furthermore, postsynaptic densities were reduced in postmortem cortices of human MPS patients, suggesting that the pathology is a common hallmark for neurological LSD. Together, our results demonstrate that lysosomal storage defects cause early alterations in synaptic structure and abnormalities in neurotransmission originating from impaired synaptic vesicular transport, and they suggest that synaptic defects could be targeted to treat behavioral and cognitive defects in neurological LSD patients.


Subject(s)
Lysosomal Storage Diseases/metabolism , Mucopolysaccharidosis III , Pyramidal Cells , Secretory Vesicles/metabolism , Synaptic Transmission/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cells, Cultured , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Disease Progression , Drug Discovery , Hippocampus/pathology , Mice , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/psychology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Protein Transport , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
19.
Mol Psychiatry ; 26(12): 7225-7246, 2021 12.
Article in English | MEDLINE | ID: mdl-34127816

ABSTRACT

CACNA1A deletions cause epilepsy, ataxia, and a range of neurocognitive deficits, including inattention, impulsivity, intellectual deficiency and autism. To investigate the underlying mechanisms, we generated mice carrying a targeted Cacna1a deletion restricted to parvalbumin-expressing (PV) neurons (PVCre;Cacna1ac/+) or to cortical pyramidal cells (PC) (Emx1Cre;Cacna1ac/+). GABA release from PV-expressing GABAergic interneurons (PV-INs) is reduced in PVCre;Cacna1ac/+ mutants, resulting in impulsivity, cognitive rigidity and inattention. By contrast, the deletion of Cacna1a in PCs does not impact cortical excitability or behaviour in Emx1Cre;Cacna1ac/+ mutants. A targeted Cacna1a deletion in the orbitofrontal cortex (OFC) results in reversal learning deficits while a medial prefrontal cortex (mPFC) deletion impairs selective attention. These deficits can be rescued by the selective chemogenetic activation of cortical PV-INs in the OFC or mPFC of PVCre;Cacna1ac/+ mutants. Thus, Cacna1a haploinsufficiency disrupts perisomatic inhibition in frontal cortical circuits, leading to a range of potentially reversible neurocognitive deficits.


Subject(s)
Calcium Channels, N-Type/metabolism , Interneurons , Neurodevelopmental Disorders , Parvalbumins , Animals , Interneurons/metabolism , Mice , Neurons/metabolism , Parvalbumins/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism
20.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Article in English | MEDLINE | ID: mdl-33876772

ABSTRACT

The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) integrates multiple signals to regulate critical cellular processes such as mRNA translation, lipid biogenesis, and autophagy. Germline and somatic mutations in mTOR and genes upstream of mTORC1, such as PTEN, TSC1/2, AKT3, PIK3CA, and components of GATOR1 and KICSTOR complexes, are associated with various epileptic disorders. Increased mTORC1 activity is linked to the pathophysiology of epilepsy in both humans and animal models, and mTORC1 inhibition suppresses epileptogenesis in humans with tuberous sclerosis and animal models with elevated mTORC1 activity. However, the role of mTORC1-dependent translation and the neuronal cell types mediating the effect of enhanced mTORC1 activity in seizures remain unknown. The eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and 2 (4E-BP2) are translational repressors downstream of mTORC1. Here we show that the ablation of 4E-BP2, but not 4E-BP1, in mice increases the sensitivity to pentylenetetrazole (PTZ)- and kainic acid (KA)-induced seizures. We demonstrate that the deletion of 4E-BP2 in inhibitory, but not excitatory neurons, causes an increase in the susceptibility to PTZ-induced seizures. Moreover, mice lacking 4E-BP2 in parvalbumin, but not somatostatin or VIP inhibitory neurons exhibit a lowered threshold for seizure induction and reduced number of parvalbumin neurons. A mouse model harboring a human PIK3CA mutation that enhances the activity of the PI3K-AKT pathway (Pik3caH1047R-Pvalb ) selectively in parvalbumin neurons shows susceptibility to PTZ-induced seizures. Our data identify 4E-BP2 as a regulator of epileptogenesis and highlight the central role of increased mTORC1-dependent translation in parvalbumin neurons in the pathophysiology of epilepsy.


Subject(s)
Epilepsy/metabolism , Eukaryotic Initiation Factors/metabolism , Neurons/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Epilepsy/genetics , Epilepsy/physiopathology , Eukaryotic Initiation Factors/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Neural Inhibition , Neurons/physiology , Parvalbumins/genetics , Parvalbumins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...