Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Nature ; 599(7883): 96-101, 2021 11.
Article in English | MEDLINE | ID: mdl-34616037

ABSTRACT

Social memory-the ability to recognize and remember familiar conspecifics-is critical for the survival of an animal in its social group1,2. The dorsal CA2 (dCA2)3-5 and ventral CA1 (vCA1)6 subregions of the hippocampus, and their projection targets6,7, have important roles in social memory. However, the relevant extrahippocampal input regions remain poorly defined. Here we identify the medial septum (MS) as a dCA2 input region that is critical for social memory and reveal that modulation of the MS by serotonin (5-HT) bidirectionally controls social memory formation, thereby affecting memory stability. Novel social interactions increase activity in dCA2-projecting MS neurons and induce plasticity at glutamatergic synapses from MS neurons onto dCA2 pyramidal neurons. The activity of dCA2-projecting MS cells is enhanced by the neuromodulator 5-HT acting on 5-HT1B receptors. Moreover, optogenetic manipulation of median raphe 5-HT terminals in the MS bidirectionally regulates social memory stability. This work expands our understanding of the neural mechanisms by which social interactions lead to social memory and provides evidence that 5-HT has a critical role in promoting not only prosocial behaviours8,9, but also social memory, by influencing distinct target structures.


Subject(s)
Memory/physiology , Neural Pathways , Septal Nuclei/physiology , Serotonin/metabolism , Social Behavior , Animals , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/physiology , Female , Glutamic Acid/metabolism , Male , Mice , Neuronal Plasticity , Optogenetics , Pyramidal Cells/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Septal Nuclei/cytology , Synapses/metabolism
2.
Int J Mol Sci ; 22(7)2021 Apr 04.
Article in English | MEDLINE | ID: mdl-33916620

ABSTRACT

Somatostatin receptor subtype 4 (SST4) has been shown to mediate analgesic, antidepressant and anti-inflammatory functions without endocrine actions; therefore, it is proposed to be a novel target for drug development. To overcome the species differences of SST4 receptor expression and function between humans and mice, we generated an SST4 humanized mouse line to serve as a translational animal model for preclinical research. A transposon vector containing the hSSTR4 and reporter gene construct driven by the hSSTR4 regulatory elements were created. The vector was randomly inserted in Sstr4-deficient mice. hSSTR4 expression was detected by bioluminescent in vivo imaging of the luciferase reporter predominantly in the brain. RT-qPCR confirmed the expression of the human gene in the brain and various peripheral tissues consistent with the in vivo imaging. RNAscope in situ hybridization revealed the presence of hSSTR4 transcripts in glutamatergic excitatory neurons in the CA1 and CA2 regions of the hippocampus; in the GABAergic interneurons in the granular layer of the olfactory bulb and in both types of neurons in the primary somatosensory cortex, piriform cortex, prelimbic cortex and amygdala. This novel SST4 humanized mouse line might enable us to investigate the differences of human and mouse SST4 receptor expression and function and assess the effects of SST4 receptor agonist drug candidates.


Subject(s)
CA1 Region, Hippocampal/metabolism , CA2 Region, Hippocampal/metabolism , Gene Expression Regulation , Neurons/metabolism , Receptors, Somatostatin/biosynthesis , Animals , CA1 Region, Hippocampal/cytology , CA2 Region, Hippocampal/cytology , Humans , Mice , Mice, Transgenic , Receptors, Somatostatin/genetics
3.
Mol Psychiatry ; 26(1): 350-364, 2021 01.
Article in English | MEDLINE | ID: mdl-31745235

ABSTRACT

Mineralocorticoid receptors (MRs) in the brain play a role in learning and memory, neuronal differentiation, and regulation of the stress response. Within the hippocampus, the highest expression of MRs is in area CA2. CA2 pyramidal neurons have a distinct molecular makeup resulting in a plasticity-resistant phenotype, distinguishing them from neurons in CA1 and CA3. Thus, we asked whether MRs regulate CA2 neuron properties and CA2-related behaviors. Using three conditional knockout methods at different stages of development, we found a striking decrease in multiple molecular markers for CA2, an effect mimicked by chronic antagonism of MRs. Furthermore, embryonic deletion of MRs disrupted afferent inputs to CA2 and enabled synaptic potentiation of the normally LTP-resistant synaptic currents in CA2. We also found that CA2-targeted MR knockout was sufficient to disrupt social behavior and alter behavioral responses to novelty. Altogether, these results demonstrate an unappreciated role for MRs in controlling CA2 pyramidal cell identity and in facilitating CA2-dependent behaviors.


Subject(s)
Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Receptors, Mineralocorticoid/metabolism , Animals , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/metabolism , Female , Male , Mice , Mice, Knockout , Neuronal Plasticity , Phenotype , Receptors, Mineralocorticoid/deficiency , Receptors, Mineralocorticoid/genetics
4.
Nature ; 586(7828): 270-274, 2020 10.
Article in English | MEDLINE | ID: mdl-32999460

ABSTRACT

The ability to recognize information that is incongruous with previous experience is critical for survival. Novelty signals have therefore evolved in the mammalian brain to enhance attention, perception and memory1,2. Although the importance of regions such as the ventral tegmental area3,4 and locus coeruleus5 in broadly signalling novelty is well-established, these diffuse monoaminergic transmitters have yet to be shown to convey specific information on the type of stimuli that drive them. Whether distinct types of novelty, such as contextual and social novelty, are differently processed and routed in the brain is unknown. Here we identify the supramammillary nucleus (SuM) as a novelty hub in the hypothalamus6. The SuM region is unique in that it not only responds broadly to novel stimuli, but also segregates and selectively routes different types of information to discrete cortical targets-the dentate gyrus and CA2 fields of the hippocampus-for the modulation of mnemonic processing. Using a new transgenic mouse line, SuM-Cre, we found that SuM neurons that project to the dentate gyrus are activated by contextual novelty, whereas the SuM-CA2 circuit is preferentially activated by novel social encounters. Circuit-based manipulation showed that divergent novelty channelling in these projections modifies hippocampal contextual or social memory. This content-specific routing of novelty signals represents a previously unknown mechanism that enables the hypothalamus to flexibly modulate select components of cognition.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Memory/physiology , Neural Pathways/physiology , Animals , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/physiology , Cognition , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Female , Hypothalamus, Posterior/cytology , Hypothalamus, Posterior/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Social Interaction
5.
Nature ; 587(7833): 264-269, 2020 11.
Article in English | MEDLINE | ID: mdl-32968277

ABSTRACT

The consolidation of spatial memory depends on the reactivation ('replay') of hippocampal place cells that were active during recent behaviour. Such reactivation is observed during sharp-wave ripples (SWRs)-synchronous oscillatory electrical events that occur during non-rapid-eye-movement (non-REM) sleep1-8 and whose disruption impairs spatial memory3,5,6,8. Although the hippocampus also encodes a wide range of non-spatial forms of declarative memory, it is not yet known whether SWRs are necessary for such memories. Moreover, although SWRs can arise from either the CA3 or the CA2 region of the hippocampus7,9, the relative importance of SWRs from these regions for memory consolidation is unknown. Here we examine the role of SWRs during the consolidation of social memory-the ability of an animal to recognize and remember a member of the same species-focusing on CA2 because of its essential role in social memory10-12. We find that ensembles of CA2 pyramidal neurons that are active during social exploration of previously unknown conspecifics are reactivated during SWRs. Notably, disruption or enhancement of CA2 SWRs suppresses or prolongs social memory, respectively. Thus, SWR-mediated reactivation of hippocampal firing related to recent experience appears to be a general mechanism for binding spatial, temporal and sensory information into high-order memory representations, including social memory.


Subject(s)
CA2 Region, Hippocampal/physiology , Memory/physiology , Sleep/physiology , Social Interaction , Animals , CA2 Region, Hippocampal/anatomy & histology , CA2 Region, Hippocampal/cytology , Male , Memory Consolidation/physiology , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Optogenetics , Pyramidal Cells/physiology
6.
Behav Brain Res ; 379: 112355, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31730784

ABSTRACT

Ovarian steroids modulate the neuronal structure and function during the estrous cycle, contrasting peak effects during the proestrus cycle and low effects during the metestrus cycle. An ovariectomy (OVX) decreases gonadal hormones and tests the effects of substitutive therapies. We studied female rats with a normal estrous cycle and we also studied the effects of systemic progesterone (P4, 4.0 mg/kg) or its reduced metabolite allopregnanolone (ALLO, 4.0 mg/kg, both for 10 days) in females who had had an OVX 16.5 weeks prior to the study (long-term OVX) with the novel object recognition test (NORT) for associative memory. The dendritic shape and spine density in Golgi-impregnated basal dendrites (stratum oriens) of hippocampal pyramidal neurons was also studied. Proestrus females had a better performance than metestrus or OVX females in short-term memory (tested 1 h after the acquisition phase). Proestrus and metestrus females showed better results than OVX females for long-term memory (24 h after the initial phase). Both P4 and ALLO recovered the cognitive impairment induced by long-term OVX. Also, proestrus females had a higher density of dendritic spines than metestrus females, OVX reduced the density of spines when compared to intact females, whereas both P4 and ALLO treatments increased the dendritic spine density, number of dendritic branches along the dendritic length, and branching order compared to vehicle. These data add the dendrites of the stratum oriens as an additional site for naturally occurring changes in spine density during the estrous cycle and evidence the actions of progestins in both behavioral recovery and the structural dendritic rearrangement of hippocampal pyramidal neurons in long-term OVX female rats.


Subject(s)
CA1 Region, Hippocampal , CA2 Region, Hippocampal , Cognitive Dysfunction , Dendritic Spines , Estrous Cycle/metabolism , Learning , Ovariectomy/adverse effects , Pregnanolone/metabolism , Pregnanolone/pharmacology , Progesterone/metabolism , Progesterone/pharmacology , Pyramidal Cells , Animals , Association Learning/drug effects , Association Learning/physiology , Behavior, Animal/physiology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/drug effects , CA2 Region, Hippocampal/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Dendritic Spines/drug effects , Disease Models, Animal , Female , Learning/drug effects , Learning/physiology , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Pregnanolone/administration & dosage , Progesterone/administration & dosage , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Rats, Wistar , Recognition, Psychology/physiology
7.
Cell Rep ; 27(1): 86-98.e3, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943417

ABSTRACT

The hippocampus is critical for the formation of episodic memory. It is, therefore, important to understand intra-hippocampal circuitry, especially in the often overlooked area CA2. Using specific transgenic mouse lines combined with opto- and chemogenetics, we show that local plasticity of parvalbumin-expressing interneurons in area CA2 allows CA3 input to recruit CA2 pyramidal neurons (PNs), thereby increasing the excitatory drive between CA3 and CA1. CA2 PNs provide both stronger excitation and larger feed-forward inhibition onto deep, compared with superficial, CA1 PNs. This feed-forward inhibition, largely mediated by parvalbumin-expressing interneurons, normalizes the excitatory drive onto deep and superficial CA1 PNs. Finally, we identify a target of CA2 in area CA1, i.e., CA1 PNs, whose soma are located in stratum radiatum. These data provide insight into local hippocampal circuitry and reveal how localized plasticity can potentially control information flow in the larger hippocampal network.


Subject(s)
CA2 Region, Hippocampal/physiology , Hippocampus/physiology , Interneurons/physiology , Neuronal Plasticity/physiology , Parvalbumins/metabolism , Synaptic Transmission/physiology , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , CA2 Region, Hippocampal/cytology , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/physiology , Hippocampus/cytology , Interneurons/metabolism , Male , Memory, Episodic , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/physiology
8.
Hippocampus ; 29(2): 78-92, 2019 02.
Article in English | MEDLINE | ID: mdl-30067288

ABSTRACT

Excitatory synaptic inputs from specific brain regions are often targeted to distinct dendritic arbors on hippocampal pyramidal neurons. Recent work has suggested that CA2 pyramidal neurons respond robustly and preferentially to excitatory input into the stratum lacunosum moleculare (SLM), with a relatively modest response to Schaffer collateral excitatory input into stratum radiatum (SR) in acute mouse hippocampal slices, but the extent to which this difference may be explained by morphology is unknown. In an effort to replicate these findings and to better understand the role of dendritic morphology in shaping responses from proximal and distal synaptic sites, we measured excitatory postsynaptic currents and action potentials in CA2 pyramidal cells in response to SR and SLM stimulation and subsequently analyzed confocal images of the filled cells. We found that, in contrast to previous reports, SR stimulation evoked substantial responses in all recorded CA2 pyramidal cells. Strikingly, however, we found that not all neurons responded to SLM stimulation, and in those neurons that did, responses evoked by SLM and SR were comparable in size and effectiveness in inducing action potentials. In a comprehensive morphometric analysis of CA2 pyramidal cell apical dendrites, we found that the neurons that were unresponsive to SLM stimulation were the same ones that lacked substantial apical dendritic arborization in the SLM. Neurons responsive to both SR and SLM stimulation had roughly equal amounts of dendritic branching in each layer. Remarkably, our study in mouse CA2 generally replicates the work characterizing the diversity of CA2 pyramidal cells in the guinea pig hippocampus. We conclude, then, that like in guinea pig, mouse CA2 pyramidal cells have a diverse apical dendrite morphology that is likely to be reflective of both the amount and source of excitatory input into CA2 from the entorhinal cortex and CA3.


Subject(s)
CA2 Region, Hippocampal/physiology , Dendrites/physiology , Entorhinal Cortex/physiology , Excitatory Postsynaptic Potentials/physiology , Pyramidal Cells/physiology , Synapses/physiology , Animals , CA2 Region, Hippocampal/cytology , Entorhinal Cortex/cytology , Mice , Mice, Inbred C57BL , Organ Culture Techniques
9.
Curr Opin Neurobiol ; 54: 194-199, 2019 02.
Article in English | MEDLINE | ID: mdl-30120016

ABSTRACT

Synaptic plasticity in the hippocampus is thought to play a vital role in both the refinement of neuronal circuits during development and in learning in the mature brain. Synapses in hippocampal area CA1 are known for a robust capacity for long-term potentiation (LTP), whereas synapses in the stratum radiatum of hippocampal area CA2 are particularly resistant to such changes. Although we have yet to fully understand the mechanisms behind this resistance to plasticity, a number of genes and extracellular matrix components highly expressed in CA2 appear to function as molecular brakes on plasticity and develop postnatally in the rodent brain. Curiously, the developmental profile of several CA2-enriched molecules is suggestive of a still undefined critical window of plasticity in the hippocampus.


Subject(s)
CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Synapses/physiology , Animals , Humans
10.
Nature ; 564(7735): 213-218, 2018 12.
Article in English | MEDLINE | ID: mdl-30518859

ABSTRACT

Although the hippocampus is known to be important for declarative memory, it is less clear how hippocampal output regulates motivated behaviours, such as social aggression. Here we report that pyramidal neurons in the CA2 region of the hippocampus, which are important for social memory, promote social aggression in mice. This action depends on output from CA2 to the lateral septum, which is selectively enhanced immediately before an attack. Activation of the lateral septum by CA2 recruits a circuit that disinhibits a subnucleus of the ventromedial hypothalamus that is known to trigger attack. The social hormone arginine vasopressin enhances social aggression by acting on arginine vasopressin 1b receptors on CA2 presynaptic terminals in the lateral septum to facilitate excitatory synaptic transmission. In this manner, release of arginine vasopressin in the lateral septum, driven by an animal's internal state, may serve as a modulatory control that determines whether CA2 activity leads to declarative memory of a social encounter and/or promotes motivated social aggression.


Subject(s)
Aggression/physiology , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/physiology , Neural Inhibition , Neural Pathways/physiology , Septal Nuclei/cytology , Septal Nuclei/physiology , Social Behavior , Animals , Arginine Vasopressin/metabolism , Clozapine/analogs & derivatives , Clozapine/pharmacology , Excitatory Postsynaptic Potentials , Female , Male , Memory/physiology , Mice , Mice, Inbred BALB C , Motivation , Presynaptic Terminals/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Pyramidal Cells/metabolism , Receptors, Vasopressin/metabolism , Synaptic Transmission , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/physiology
11.
J Proteome Res ; 17(4): 1700-1711, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29518331

ABSTRACT

Regulator of G Protein Signaling 14 (RGS14) is a complex scaffolding protein that integrates G protein and MAPK signaling pathways. In the adult mouse brain, RGS14 is predominantly expressed in hippocampal CA2 neurons where it naturally inhibits synaptic plasticity and hippocampus-dependent learning and memory. However, the signaling proteins that RGS14 natively engages to regulate plasticity are unknown. Here, we show that RGS14 exists in a high-molecular-weight protein complex in brain. To identify RGS14 neuronal interacting partners, endogenous RGS14 immunoprecipitated from mouse brain was subjected to mass spectrometry and proteomic analysis. We find that RGS14 interacts with key postsynaptic proteins that regulate plasticity. Gene ontology analysis reveals the most enriched RGS14 interactors have functional roles in actin-binding, calmodulin(CaM)-binding, and CaM-dependent protein kinase (CaMK) activity. We validate these findings using biochemical assays that identify interactions with two previously unknown binding partners. We report that RGS14 directly interacts with Ca2+/CaM and is phosphorylated by CaMKII in vitro. Lastly, we detect that RGS14 associates with CaMKII and CaM in hippocampal CA2 neurons. Taken together, these findings demonstrate that RGS14 is a novel CaM effector and CaMKII phosphorylation substrate thereby providing new insight into mechanisms by which RGS14 controls plasticity in CA2 neurons.


Subject(s)
Brain Chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Hippocampus/chemistry , RGS Proteins/metabolism , Animals , CA2 Region, Hippocampal/cytology , Calcium/metabolism , Hippocampus/metabolism , Mice , Neuronal Plasticity , Neurons/metabolism , Phosphorylation , Protein Binding , Proteomics
12.
J Neurosci ; 38(5): 1218-1231, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29279308

ABSTRACT

Oxytocin (OXT) receptors (OXTRs) are prominently expressed in hippocampal CA2 and CA3 pyramidal neurons, but little is known about its physiological function. As the functional necessity of hippocampal CA2 for social memory processing, we tested whether CA2 OXTRs may contribute to long-term social recognition memory (SRM) formation. Here, we found that conditional deletion of Oxtr from forebrain (Oxtr-/-) or CA2/CA3a-restricted excitatory neurons in adult male mice impaired the persistence of long-term SRM but had no effect on sociability and preference for social novelty. Conditional deletion of CA2/CA3a Oxtr showed no changes in anxiety-like behavior assessed using the open-field, elevated plus maze and novelty-suppressed feeding tests. Application of a highly selective OXTR agonist [Thr4,Gly7]-OXT to hippocampal slices resulted in an acute and lasting potentiation of excitatory synaptic responses in CA2 pyramidal neurons that relied on N-methyl-d-aspartate receptor activation and calcium/calmodulin-dependent protein kinase II activity. In addition, Oxtr-/- mice displayed a defect in the induction of long-term potentiation, but not long-term depression, at the synapses between the entorhinal cortex and CA2 pyramidal neurons. Furthermore, Oxtr deletion led to a reduced complexity of basal dendritic arbors of CA2 pyramidal neurons, but caused no alteration in the density of apical dendritic spines. Considering that the methodologies we have used to delete Oxtr do not rule out targeting the neighboring CA3a region, these findings suggest that OXTR signaling in the CA2/CA3a is crucial for the persistence of long-term SRM.SIGNIFICANCE STATEMENT Oxytocin receptors (OXTRs) are abundantly expressed in hippocampal CA2 and CA3 regions, but there are little known about their physiological function. Taking advantage of the conditional Oxtr knock-out mice, the present study highlights the importance of OXTR signaling in the induction of long-term potentiation at the synapses between the entorhinal cortex and CA2 pyramidal neurons and the persistence of long-term social recognition memory. Thus, OXTRs in the CA2/CA3a may provide a new target for therapeutic approaches to the treatment of social cognition deficits, which are often observed in patients with neuropsychiatric disorders.


Subject(s)
CA2 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Receptors, Oxytocin/genetics , Receptors, Oxytocin/physiology , Recognition, Psychology/physiology , Social Behavior , Animals , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/metabolism , Dendrites/drug effects , Dendrites/ultrastructure , Entorhinal Cortex/cytology , Entorhinal Cortex/metabolism , Entorhinal Cortex/physiology , Excitatory Postsynaptic Potentials/drug effects , Gene Deletion , Long-Term Potentiation/genetics , Male , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Oxytocin/analogs & derivatives , Oxytocin/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, Oxytocin/agonists
13.
eNeuro ; 4(4)2017.
Article in English | MEDLINE | ID: mdl-28856240

ABSTRACT

The CA1 region of the hippocampus plays a critical role in spatial and contextual memory, and has well-established circuitry, function and plasticity. In contrast, the properties of the flanking CA2 pyramidal neurons (PNs), important for social memory, and lacking CA1-like plasticity, remain relatively understudied. In particular, little is known regarding the expression of voltage-gated K+ (Kv) channels and the contribution of these channels to the distinct properties of intrinsic excitability, action potential (AP) waveform, firing patterns and neurotransmission between CA1 and CA2 PNs. In the present study, we used multiplex fluorescence immunolabeling of mouse brain sections, and whole-cell recordings in acute mouse brain slices, to define the role of heterogeneous expression of Kv2 family Kv channels in CA1 versus CA2 pyramidal cell excitability. Our results show that the somatodendritic delayed rectifier Kv channel subunits Kv2.1, Kv2.2, and their auxiliary subunit AMIGO-1 have region-specific differences in expression in PNs, with the highest expression levels in CA1, a sharp decrease at the CA1-CA2 boundary, and significantly reduced levels in CA2 neurons. PNs in CA1 exhibit a robust contribution of Guangxitoxin-1E-sensitive Kv2-based delayed rectifier current to AP shape and after-hyperpolarization potential (AHP) relative to that seen in CA2 PNs. Our results indicate that robust Kv2 channel expression confers a distinct pattern of intrinsic excitability to CA1 PNs, potentially contributing to their different roles in hippocampal network function.


Subject(s)
Action Potentials/physiology , CA1 Region, Hippocampal/metabolism , CA2 Region, Hippocampal/metabolism , Pyramidal Cells/metabolism , Shab Potassium Channels/metabolism , Action Potentials/drug effects , Animals , Arthropod Proteins/pharmacology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , CA2 Region, Hippocampal/cytology , CA2 Region, Hippocampal/drug effects , Female , Gene Expression , Immunohistochemistry , Male , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Shab Potassium Channels/antagonists & inhibitors , Spider Venoms/pharmacology , Tissue Culture Techniques
14.
Neuron ; 95(5): 1089-1102.e5, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28823730

ABSTRACT

Input-timing-dependent plasticity (ITDP) is a circuit-based synaptic learning rule by which paired activation of entorhinal cortical (EC) and Schaffer collateral (SC) inputs to hippocampal CA1 pyramidal neurons (PNs) produces a long-term enhancement of SC excitation. We now find that paired stimulation of EC and SC inputs also induces ITDP of SC excitation of CA2 PNs. However, whereas CA1 ITDP results from long-term depression of feedforward inhibition (iLTD) as a result of activation of CB1 endocannabinoid receptors on cholecystokinin-expressing interneurons, CA2 ITDP results from iLTD through activation of δ-opioid receptors on parvalbumin-expressing interneurons. Furthermore, whereas CA1 ITDP has been previously linked to enhanced specificity of contextual memory, we find that CA2 ITDP is associated with enhanced social memory. Thus, ITDP may provide a general synaptic learning rule for distinct forms of hippocampal-dependent memory mediated by distinct hippocampal regions.


Subject(s)
CA2 Region, Hippocampal/physiology , Memory/physiology , Neuronal Plasticity/physiology , Animals , CA2 Region, Hippocampal/cytology , Entorhinal Cortex/physiology , Interneurons/metabolism , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Transgenic , Neural Inhibition/physiology , Parvalbumins/metabolism , Pyramidal Cells/physiology , Receptors, Opioid, delta/metabolism , Social Behavior , Time Factors
15.
Neuron ; 94(3): 642-655.e9, 2017 May 03.
Article in English | MEDLINE | ID: mdl-28472661

ABSTRACT

Hippocampal CA2 pyramidal cells project into both the neighboring CA1 and CA3 subfields, leaving them well positioned to influence network physiology and information processing for memory and space. While recent work has suggested unique roles for CA2, including encoding position during immobility and generating ripple oscillations, an interventional examination of the integrative functions of these connections has yet to be reported. Here we demonstrate that CA2 recruits feedforward inhibition in CA3 and that chronic genetically engineered shutdown of CA2-pyramidal-cell synaptic transmission consequently results in increased excitability of the recurrent CA3 network. In behaving mice, this led to spatially triggered episodes of network-wide hyperexcitability during exploration accompanied by the emergence of high-frequency discharges during rest. These findings reveal CA2 as a regulator of network processing in hippocampus and suggest that CA2-mediated inhibition in CA3 plays a key role in establishing the dynamic excitatory and inhibitory balance required for proper network function.


Subject(s)
CA2 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Nerve Net/physiology , Neural Inhibition/physiology , Pyramidal Cells/physiology , Synaptic Transmission/physiology , Theta Rhythm/physiology , Animals , CA2 Region, Hippocampal/cytology , CA3 Region, Hippocampal/cytology , Hippocampus/physiology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Tetanus Toxin/genetics , Tetanus Toxin/metabolism
16.
Cereb Cortex ; 27(11): 5284-5293, 2017 11 01.
Article in English | MEDLINE | ID: mdl-27733539

ABSTRACT

GABAergic dysfunction in hippocampus, a key feature of schizophrenia (SZ), may contribute to cognitive impairment in this disorder. In stratum oriens (SO) of sector CA3/2 of the human hippocampus, a network of genes involved in the regulation of glutamic acid decarboxylase GAD67 has been identified. Several of the genes in this network including epigenetic factors histone deacetylase 1 (HDAC1) and death-associated protein 6 (DAXX), the GABAergic enzyme GAD65 as well as the kainate receptor (KAR) subunits GluR6 and 7 show significant changes in expression in this area in SZ. We have tested whether HDAC1 and DAXX regulate GAD67, GAD65, or GluR in the intact rodent hippocampus. Stereotaxic injections of lentiviral vectors bearing shRNAi sequences for HDAC1 and DAXX were delivered into the SO of CA3/2, followed by laser microdissection of individual transduced GABA neurons. Quantitative PCR (QPCR) analyses demonstrated that inhibition of HDAC1 and DAXX increased expression of GAD67, GAD65, and GluR6 mRNA. Inhibition of DAXX, but not HDAC1 resulted in a significant increase in GluR7 mRNA. Our data support the hypothesis that HDAC1 and DAXX play a central role in coordinating the expression of genes in the GAD67 regulatory pathway in the SO of CA3/2.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CA2 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/metabolism , Epigenesis, Genetic , Glutamate Decarboxylase/metabolism , Histone Deacetylase 1/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , CA2 Region, Hippocampal/cytology , CA3 Region, Hippocampal/cytology , Cell Line , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Male , Molecular Chaperones , Neural Pathways/cytology , Neural Pathways/metabolism , Nuclear Proteins/antagonists & inhibitors , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism
17.
Hippocampus ; 26(12): 1593-1607, 2016 12.
Article in English | MEDLINE | ID: mdl-27650887

ABSTRACT

It is well-established that the feed-forward connected main hippocampal areas, CA3, CA2, and CA1 work cooperatively during spatial navigation and memory. These areas are similar in terms of the prevalent types of neurons; however, they display different spatial coding and oscillatory dynamics. Understanding the temporal dynamics of these operations requires simultaneous recordings from these regions. However, simultaneous recordings from multiple regions and subregions in behaving animals have become possible only recently. We performed large-scale silicon probe recordings simultaneously spanning across all layers of CA1, CA2, and CA3 regions in rats during spatial navigation and sleep and compared their behavior-dependent spiking, oscillatory dynamics and functional connectivity. The accuracy of place cell spatial coding increased progressively from distal to proximal CA1, suddenly dropped in CA2, and increased again from CA3a toward CA3c. These variations can be attributed in part to the different entorhinal inputs to each subregions, and the differences in theta modulation of CA1, CA2, and CA3 neurons. We also found that neurons in the subregions showed differences in theta modulation, phase precession, state-dependent changes in firing rates and functional connectivity among neurons of these regions. Our results indicate that a combination of intrinsic properties together with distinct intra- and extra-hippocampal inputs may account for the subregion-specific modulation of spiking dynamics and spatial tuning of neurons during behavior. © 2016 Wiley Periodicals, Inc.


Subject(s)
CA1 Region, Hippocampal/physiology , CA2 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Neurons/physiology , Space Perception/physiology , Action Potentials , Animals , CA1 Region, Hippocampal/cytology , CA2 Region, Hippocampal/cytology , CA3 Region, Hippocampal/cytology , Electrodes, Implanted , Entorhinal Cortex/cytology , Entorhinal Cortex/physiology , Immunohistochemistry , Male , Motor Activity/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/cytology , Rats, Long-Evans , Signal Processing, Computer-Assisted , Spatial Navigation/physiology , Theta Rhythm/physiology
18.
J Neurosci ; 36(23): 6312-20, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27277807

ABSTRACT

UNLABELLED: Long-term potentiation of excitatory synapses on pyramidal neurons in the stratum radiatum rarely occurs in hippocampal area CA2. Here, we present evidence that perineuronal nets (PNNs), a specialized extracellular matrix typically localized around inhibitory neurons, also surround mouse CA2 pyramidal neurons and envelop their excitatory synapses. CA2 pyramidal neurons express mRNA transcripts for the major PNN component aggrecan, identifying these neurons as a novel source for PNNs in the hippocampus. We also found that disruption of PNNs allows synaptic potentiation of normally plasticity-resistant excitatory CA2 synapses; thus, PNNs play a role in restricting synaptic plasticity in area CA2. Finally, we found that postnatal development of PNNs on CA2 pyramidal neurons is modified by early-life enrichment, suggesting that the development of circuits containing CA2 excitatory synapses are sensitive to manipulations of the rearing environment. SIGNIFICANCE STATEMENT: Perineuronal nets (PNNs) are thought to play a major role in restricting synaptic plasticity during postnatal development, and are altered in several models of neurodevelopmental disorders, such as schizophrenia and Rett syndrome. Although PNNs have been predominantly studied in association with inhibitory neurons throughout the brain, we describe a dense expression of PNNs around excitatory pyramidal neurons in hippocampal area CA2. We also provide insight into a previously unrecognized role for PNNs in restricting plasticity at excitatory synapses and raise the possibility of an early critical period of hippocampal plasticity that may ultimately reveal a key mechanism underlying learning and memory impairments of PNN-associated neurodevelopmental disorders.


Subject(s)
CA2 Region, Hippocampal/cytology , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation, Developmental/physiology , Nerve Net/physiology , Pyramidal Cells/physiology , Satellite Cells, Perineuronal/physiology , Animals , Animals, Newborn , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/drug effects , Nerve Net/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure , Satellite Cells, Perineuronal/drug effects
19.
Nat Rev Neurosci ; 17(2): 89-102, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26806628

ABSTRACT

Hippocampal area CA2 has several features that distinguish it from CA1 and CA3, including a unique gene expression profile, failure to display long-term potentiation and relative resistance to cell death. A recent increase in interest in the CA2 region, combined with the development of new methods to define and manipulate its neurons, has led to some exciting new discoveries on the properties of CA2 neurons and their role in behaviour. Here, we review these findings and call attention to the idea that the definition of area CA2 ought to be revised in light of gene expression data.


Subject(s)
CA2 Region, Hippocampal/physiology , Learning/physiology , Neuronal Plasticity/physiology , Social Behavior , Animals , CA2 Region, Hippocampal/cytology , Humans , Nerve Net/physiology , Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...