Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Annu Rev Neurosci ; 43: 55-72, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-31874067

RESUMO

Although Lorente de No' recognized the anatomical distinction of the hippocampal Cornu Ammonis (CA) 2 region, it had, until recently, been assigned no unique function. Its location between the key players of the circuit, CA3 and CA1, which along with the entorhinal cortex and dentate gyrus compose the classic trisynaptic circuit, further distracted research interest. However, the connectivity of CA2 pyramidal cells, together with unique patterns of gene expression, hints at a much larger contribution to hippocampal information processing than has been ascribed. Here we review recent advances that have identified new roles for CA2 in hippocampal centric processing, together with specialized functions in social memory and, potentially, as a broadcaster of novelty. These new data, together with CA2's role in disease, justify a closer look at how this small region exerts its influence and how it might best be exploited to understand and treat disease-related circuit dysfunctions.


Assuntos
Região CA2 Hipocampal/fisiologia , Hipocampo/fisiologia , Memória/fisiologia , Vias Neurais/fisiologia , Animais , Córtex Entorrinal/fisiologia , Humanos , Rede Nervosa/fisiologia
2.
Cell ; 149(1): 188-201, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22365813

RESUMO

Adult-born granule cells (GCs), a minor population of cells in the hippocampal dentate gyrus, are highly active during the first few weeks after functional integration into the neuronal network, distinguishing them from less active, older adult-born GCs and the major population of dentate GCs generated developmentally. To ascertain whether young and old GCs perform distinct memory functions, we created a transgenic mouse in which output of old GCs was specifically inhibited while leaving a substantial portion of young GCs intact. These mice exhibited enhanced or normal pattern separation between similar contexts, which was reduced following ablation of young GCs. Furthermore, these mutant mice exhibited deficits in rapid pattern completion. Therefore, pattern separation requires adult-born young GCs but not old GCs, and older GCs contribute to the rapid recall by pattern completion. Our data suggest that as adult-born GCs age, their function switches from pattern separation to rapid pattern completion.


Assuntos
Envelhecimento , Giro Denteado/citologia , Giro Denteado/fisiologia , Animais , Pareamento Cromossômico , Giro Denteado/crescimento & desenvolvimento , Proteínas de Fluorescência Verde/genética , Hipocampo/fisiologia , Memória , Camundongos , Camundongos Transgênicos
3.
Nature ; 586(7828): 270-274, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32999460

RESUMO

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.


Assuntos
Hipocampo/citologia , Hipocampo/fisiologia , Memória/fisiologia , Vias Neurais/fisiologia , Animais , Região CA2 Hipocampal/citologia , Região CA2 Hipocampal/fisiologia , Cognição , Giro Denteado/citologia , Giro Denteado/fisiologia , Feminino , Hipotálamo Posterior/citologia , Hipotálamo Posterior/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Interação Social
4.
Mol Psychiatry ; 28(5): 1932-1945, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36882500

RESUMO

The BTBR T+Itpr3tf/J (BTBR/J) strain is one of the most valid models of idiopathic autism, serving as a potent forward genetics tool to dissect the complexity of autism. We found that a sister strain with an intact corpus callosum, BTBR TF/ArtRbrc (BTBR/R), showed more prominent autism core symptoms but moderate ultrasonic communication/normal hippocampus-dependent memory, which may mimic autism in the high functioning spectrum. Intriguingly, disturbed epigenetic silencing mechanism leads to hyperactive endogenous retrovirus (ERV), a mobile genetic element of ancient retroviral infection, which increases de novo copy number variation (CNV) formation in the two BTBR strains. This feature makes the BTBR strain a still evolving multiple-loci model toward higher ASD susceptibility. Furthermore, active ERV, analogous to virus infection, evades the integrated stress response (ISR) of host defense and hijacks the transcriptional machinery during embryonic development in the BTBR strains. These results suggest dual roles of ERV in the pathogenesis of ASD, driving host genome evolution at a long-term scale and managing cellular pathways in response to viral infection, which has immediate effects on embryonic development. The wild-type Draxin expression in BTBR/R also makes this substrain a more precise model to investigate the core etiology of autism without the interference of impaired forebrain bundles as in BTBR/J.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Retrovirus Endógenos , Gravidez , Feminino , Humanos , Animais , Camundongos , Retrovirus Endógenos/genética , Variações do Número de Cópias de DNA , Transtorno Autístico/etiologia , Prosencéfalo/metabolismo , Corpo Caloso/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/complicações , Camundongos Endogâmicos
5.
Hippocampus ; 33(3): 252-265, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36594707

RESUMO

Dynamic interactions between the subregions of the hippocampus are required for the encoding and consolidation of memory. While the interplay and contributions of the CA1 and CA3 regions are well understood, we continue to learn more about how CA2 differentially contributes to the organization of network function. For example, CA2 place cells have been reported to be less spatially tuned during exploration, but uniquely capable of coding place while an animal stops. Here we applied chemogenetics to transiently silence CA2 pyramidal cells and found that CA2 influences hippocampal dynamics in a state-dependent manner. We find that during rest, CA2 inhibition reduces synchronization across regions (CA1, CA2, CA3) and frequency bands (low-gamma- and ripple-band). Moreover, during new learning CA1 place field formation is slower in the absence of CA2 transmission and during pausing, CA1 pyramidal cells are less excitable without CA2 drive. On the network level, a novel convolutional neural network (SpikeDecoder) was employed to show subregion and state-dependent changes in spatial coding that agree with our observations on the single cell level. Together these data suggest additional novel roles for CA2 in governing and differentiating hippocampal dynamics under discrete behavioral states.


Assuntos
Região CA1 Hipocampal , Hipocampo , Animais , Hipocampo/fisiologia , Região CA1 Hipocampal/fisiologia , Células Piramidais
6.
Hum Mol Genet ; 30(18): 1762-1772, 2021 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-34104969

RESUMO

A report of a family of Darier's disease with mood disorders drew attention when the causative gene was identified as ATP2A2 (or SERCA2), which encodes a Ca2+ pump on the endoplasmic reticulum (ER) membrane and is important for intracellular Ca2+ signaling. Recently, it was found that loss-of-function mutations of ATP2A2 confer a risk of neuropsychiatric disorders including depression, bipolar disorder and schizophrenia. In addition, a genome-wide association study found an association between ATP2A2 and schizophrenia. However, the mechanism of how ATP2A2 contributes to vulnerability to these mental disorders is unknown. Here, we analyzed Atp2a2 heterozygous brain-specific conditional knockout (hetero cKO) mice. The ER membranes prepared from the hetero cKO mouse brain showed decreased Ca2+ uptake activity. In Atp2a2 heterozygous neurons, decays of cytosolic Ca2+ level were slower than control neurons after depolarization. The hetero cKO mice showed altered behavioral responses to novel environments and impairments in fear memory, suggestive of enhanced dopamine signaling. In vivo dialysis demonstrated that extracellular dopamine levels in the NAc were indeed higher in the hetero cKO mice. These results altogether indicate that the haploinsufficiency of Atp2a2 in the brain causes prolonged cytosolic Ca2+ transients, which possibly results in enhanced dopamine signaling, a common feature of mood disorders and schizophrenia. These findings elucidate how ATP2A2 mutations causing a dermatological disease may exert their pleiotropic effects on the brain and confer a risk for mental disorders.


Assuntos
Comportamento Animal , Encéfalo/enzimologia , Doença de Darier , Dopamina/metabolismo , Mutação com Perda de Função , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Transdução de Sinais , Animais , Doença de Darier/enzimologia , Doença de Darier/genética , Dopamina/genética , Camundongos , Camundongos Knockout , Especificidade de Órgãos/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
7.
Mol Psychiatry ; 27(8): 3343-3354, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35491410

RESUMO

Immune dysregulation plays a key role in the pathogenesis of autism. Changes occurring at the systemic level, from brain inflammation to disturbed innate/adaptive immune in the periphery, are frequently observed in patients with autism; however, the intrinsic mechanisms behind them remain elusive. We hypothesize a common etiology may lie in progenitors of different types underlying widespread immune dysregulation. By single-cell RNA sequencing (sc-RNA seq), we trace the developmental origins of immune dysregulation in a mouse model of idiopathic autism. It is found that both in aorta-gonad-mesonephros (AGM) and yolk sac (YS) progenitors, the dysregulation of HDAC1-mediated epigenetic machinery alters definitive hematopoiesis during embryogenesis and downregulates the expression of the AP-1 complex for microglia development. Subsequently, these changes result in the dysregulation of the immune system, leading to gut dysbiosis and hyperactive microglia in the brain. We further confirm that dysregulated immune profiles are associated with specific microbiota composition, which may serve as a biomarker to identify autism of immune-dysregulated subtypes. Our findings elucidate a shared mechanism for the origin of immune dysregulation from the brain to the gut in autism and provide new insight to dissecting the heterogeneity of autism, as well as the therapeutic potential of targeting immune-dysregulated autism subtypes.


Assuntos
Transtorno Autístico , Camundongos , Animais , Transtorno Autístico/genética , Mesonefro , Saco Vitelino/fisiologia , Gônadas , Epigênese Genética/genética , Modelos Animais de Doenças
8.
Glia ; 70(5): 961-974, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35084774

RESUMO

Glutamatergic transmission prompts K+ efflux through postsynaptic NMDA receptors. The ensuing hotspot of extracellular K+ elevation depolarizes presynaptic terminal, boosting glutamate release, but whether this also affects glutamate uptake in local astroglia has remained an intriguing question. Here, we find that the pharmacological blockade, or conditional knockout, of postsynaptic NMDA receptors suppresses use-dependent increase in the amplitude and duration of the astrocytic glutamate transporter current (IGluT ), whereas blocking astrocytic K+ channels prevents the duration increase only. Glutamate spot-uncaging reveals that astrocyte depolarization, rather than extracellular K+ rises per se, is required to reduce the amplitude and duration of IGluT . Biophysical simulations confirm that local transient elevations of extracellular K+ can inhibit local glutamate uptake in fine astrocytic processes. Optical glutamate sensor imaging and a two-pathway test relate postsynaptic K+ efflux to enhanced extrasynaptic glutamate signaling. Thus, repetitive glutamatergic transmission triggers a feedback loop in which postsynaptic K+ efflux can transiently facilitate presynaptic release while reducing local glutamate uptake.


Assuntos
Ácido Glutâmico , Receptores de N-Metil-D-Aspartato , Animais , Astrócitos , Ratos , Ratos Sprague-Dawley , Sinapses
9.
J Neurosci ; 40(25): 4936-4944, 2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32414785

RESUMO

Hippocampus receives dense serotonergic input specifically from raphe nuclei. However, what information is carried by this input and its impact on behavior has not been fully elucidated. Here we used in vivo two-photon imaging of activity of hippocampal median raphe projection fibers in behaving male and female mice and identified two distinct populations: one linked to reward delivery and the other to locomotion. Local optogenetic manipulation of these fibers confirmed a functional role for these projections in the modulation of reward-induced behavior. The diverse function of serotonergic inputs suggests a key role in integrating locomotion and reward information into the hippocampal CA1.SIGNIFICANCE STATEMENT Information constantly flows in the hippocampus, but only some of it is captured as a memory. One potential process that discriminates which information should be remembered is concomitance with reward. In this work, we report a neuromodulatory pathway, which delivers reward signal as well as locomotion signal to the hippocampal CA1. We found that the serotonergic system delivers heterogeneous input that may be integrated by the hippocampus to support its mnemonic functions. It is dynamically involved in regulating behavior through interaction with the hippocampus. Our results suggest that the serotonergic system interacts with the hippocampus in a dynamic and behaviorally specific manner to regulate reward-related information processing.


Assuntos
Comportamento Animal/fisiologia , Hipocampo/fisiologia , Locomoção/fisiologia , Vias Neurais/fisiologia , Recompensa , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Serotoninérgicos/fisiologia
10.
EMBO J ; 34(21): 2652-70, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26423604

RESUMO

Paternal behavior is not innate but arises through social experience. After mating and becoming fathers, male mice change their behavior toward pups from infanticide to paternal care. However, the precise brain areas and circuit mechanisms connecting these social behaviors are largely unknown. Here we demonstrated that the c-Fos expression pattern in the four nuclei of the preoptic-bed nuclei of stria terminalis (BST) region could robustly discriminate five kinds of previous social behavior of male mice (parenting, infanticide, mating, inter-male aggression, solitary control). Specifically, neuronal activation in the central part of the medial preoptic area (cMPOA) and rhomboid nucleus of the BST (BSTrh) retroactively detected paternal and infanticidal motivation with more than 95% accuracy. Moreover, cMPOA lesions switched behavior in fathers from paternal to infanticidal, while BSTrh lesions inhibited infanticide in virgin males. The projections from cMPOA to BSTrh were largely GABAergic. Optogenetic or pharmacogenetic activation of cMPOA attenuated infanticide in virgin males. Taken together, this study identifies the preoptic-BST nuclei underlying social motivations in male mice and reveals unexpected complexity in the circuit connecting these nuclei.


Assuntos
Comportamento Paterno , Área Pré-Óptica/fisiologia , Animais , Comportamento Animal , Mapeamento Encefálico , Neurônios GABAérgicos/metabolismo , Masculino , Camundongos , Área Pré-Óptica/citologia , Proteínas Proto-Oncogênicas c-fos/metabolismo
11.
Angew Chem Int Ed Engl ; 58(27): 9262-9268, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31087740

RESUMO

Cargo transport along axons, a physiological process mediated by motor proteins, is essential for neuronal function and survival. A current limitation in the study of axonal transport is the lack of a robust imaging technique with a high spatiotemporal resolution to visualize and quantify the movement of motor proteins in real-time and in different depth planes. Herein, we present a dynamic imaging technique that fully exploits the characteristics of upconversion nanoparticles. This technique can be used as a microscopic probe for the quantitative in situ tracking of retrograde transport neurons with single-particle resolution in multilayered cultures. This study may provide a powerful tool to reveal dynamic neuronal activity and intra-axonal transport function as well as any associated neurodegenerative diseases resulting from mutation or impairment in the axonal transport machinery.


Assuntos
Nanopartículas Metálicas/química , Proteínas Motores Moleculares/metabolismo , Neurônios/metabolismo , Animais , Axônios/química , Axônios/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Reprogramação Celular , Dineínas/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Raios Infravermelhos , Camundongos , Microscopia de Fluorescência , Neurônios/citologia , Transporte Proteico , Ratos
12.
J Neurosci ; 34(8): 3056-66, 2014 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-24553945

RESUMO

Contextual learning involves associating cues with an environment and relating them to past experience. Previous data indicate functional specialization within the hippocampal circuit: the dentate gyrus (DG) is crucial for discriminating similar contexts, whereas CA3 is required for associative encoding and recall. Here, we used Arc/H1a catFISH imaging to address the contribution of the largely overlooked CA2 region to contextual learning by comparing ensemble codes across CA3, CA2, and CA1 in mice exposed to familiar, altered, and novel contexts. Further, to manipulate the quality of information arriving in CA2 we used two hippocampal mutant mouse lines, CA3-NR1 KOs and DG-NR1 KOs, that result in hippocampal CA3 neuronal activity that is uncoupled from the animal's sensory environment. Our data reveal largely coherent responses across the CA axis in control mice in purely novel or familiar contexts; however, in the mutant mice subject to these protocols the CA2 response becomes uncoupled from CA1 and CA3. Moreover, we show in wild-type mice that the CA2 ensemble is more sensitive than CA1 and CA3 to small changes in overall context. Our data suggest that CA2 may be tuned to remap in response to any conflict between stored and current experience.


Assuntos
Região CA2 Hipocampal/fisiologia , Aprendizagem/fisiologia , Animais , Comportamento Animal/fisiologia , Sinais (Psicologia) , Proteínas do Citoesqueleto/fisiologia , Meio Ambiente , Hipocampo/fisiologia , Processamento de Imagem Assistida por Computador , Hibridização in Situ Fluorescente , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/fisiologia , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/fisiologia , RNA/biossíntese , RNA/genética , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Sensação/fisiologia
13.
J Neurosci ; 34(33): 11007-15, 2014 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-25122900

RESUMO

The ability to associate the consumption of a taste with its positive or negative consequences is fundamental to survival and influences the behavior of species ranging from invertebrate to human. As a result, for both research and clinical reasons, there has been a great effort to understand the neuronal circuits, as well as the cellular and molecular mechanisms, underlying taste learning. From a neuroanatomical perspective, the contributions of the cortex and amygdala are well documented; however, the literature is riddled with conflicting results regarding the role of the hippocampus in different facets of taste learning. Here, we use conditional genetics in mice to block NMDA receptor-dependent plasticity individually in each of the three major hippocampal subfields, CA1, CA3, and the dentate gyrus, via deletion of the NR1 subunit. Across the CA1, CA3, and dentate gyrus NR1 knock-out lines, we uncover a pattern of differential deficits that establish the dispensability of hippocampal plasticity in incidental taste learning, the requirement of CA1 plasticity for associative taste learning, and a specific requirement for plasticity in the dentate gyrus when there is a long temporal gap between the taste and its outcome. Together, these data establish that the hippocampus is involved in associative taste learning and suggest an episodic component to this type of memory.


Assuntos
Aprendizagem por Associação/fisiologia , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Percepção Gustatória/fisiologia , Paladar/fisiologia , Animais , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
14.
Hippocampus ; 25(1): 38-50, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25139366

RESUMO

Stress alters the function of many physiological processes throughout the body, including in the brain. A neural circuit particularly vulnerable to the effects of stress is the hippocampus, a key component of the episodic and spatial memory system in both humans and rodents. Earlier studies have provided snapshots of morphological, molecular, physiological and behavioral changes in the hippocampus following either acute or repeated stress. However, the cumulative impact of repeated stress on in vivo hippocampal physiology remains unexplored. Here we report the stress-induced modulation of the spatially receptive fields of the hippocampal CA1 'place cells' as mice explore familiar and novel tracks after 5 and 10 days of immobilization stress. We find that similar to what has been observed following acute stress, five days of repeated stress results in decreased excitability of CA1 pyramidal cells. Following ten days of chronic stress, however, this decreased hippocampal excitability is no longer evident, suggesting adaptation may have occurred. In addition to these changes in neuronal excitability, we find deficient context discrimination, wherein both short-term and chronic stress impair the ability of the hippocampus to unambiguously distinguish novel and familiar environments. These results suggest that a loss of network flexibility may underlie some of the behavioral deficits accompanying chronic stress.


Assuntos
Região CA1 Hipocampal/fisiopatologia , Discriminação Psicológica/fisiologia , Generalização Psicológica/fisiologia , Memória Espacial/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Comportamento Animal/fisiologia , Região CA1 Hipocampal/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo
15.
J Neurosci ; 33(20): 8909-21, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23678132

RESUMO

Lateral habenula (LHb) has attracted growing interest as a regulator of serotonergic and dopaminergic neurons in the CNS. However, it remains unclear how the LHb modulates brain states in animals. To identify the neural substrates that are under the influence of LHb regulation, we examined the effects of rat LHb lesions on the hippocampal oscillatory activity associated with the transition of brain states. Our results showed that the LHb lesion shortened the theta activity duration both in anesthetized and sleeping rats. Furthermore, this inhibitory effect of LHb lesion on theta maintenance depended upon an intact serotonergic median raphe, suggesting that LHb activity plays an essential role in maintaining hippocampal theta oscillation via the serotonergic raphe. Multiunit recording of sleeping rats further revealed that firing of LHb neurons showed significant phase-locking activity at each theta oscillation cycle in the hippocampus. LHb neurons showing activity that was coordinated with that of the hippocampal theta were localized in the medial LHb division, which receives afferents from the diagonal band of Broca (DBB), a pacemaker region for the hippocampal theta oscillation. Thus, our findings indicate that the DBB may pace not only the hippocampus, but also the LHb, during rapid eye movement sleep. Since serotonin is known to negatively regulate theta oscillation in the hippocampus, phase-locking activity of the LHb neurons may act, under the influence of the DBB, to maintain the hippocampal theta oscillation by modulating the activity of serotonergic neurons.


Assuntos
Potenciais de Ação/fisiologia , Sincronização de Fases em Eletroencefalografia/fisiologia , Habenula/citologia , Hipocampo/fisiologia , Neurônios/fisiologia , Ritmo Teta/fisiologia , Animais , Mapeamento Encefálico , Toxina da Cólera , Eletroencefalografia , Eletrólise , Eletromiografia , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Habenula/lesões , Masculino , Vias Neurais/fisiologia , RNA Mensageiro , Ratos , Ratos Long-Evans , Sono/fisiologia , Estilbamidinas , Vigília/fisiologia
16.
Neurosci Biobehav Rev ; 158: 105559, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38246230

RESUMO

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that impacts a variety of cognitive and behavioral domains. While a genetic component of ASD has been well-established, none of the numerous syndromic genes identified in humans accounts for more than 1% of the clinical patients. Due to this large number of target genes, numerous mouse models of the disorder have been generated. However, the focus on distinct brain circuits, behavioral phenotypes and diverse experimental approaches has made it difficult to synthesize the overwhelming number of model animal studies into concrete throughlines that connect the data across levels of investigation. Here we chose to focus on one circuit, the hippocampus, and one hypothesis, a shift in excitatory/inhibitory balance, to examine, from the level of the tripartite synapse up to the level of in vivo circuit activity, the key commonalities across disparate models that can illustrate a path towards a better mechanistic understanding of ASD's impact on hippocampal circuit function.


Assuntos
Transtorno do Espectro Autista , Animais , Camundongos , Humanos , Transtorno do Espectro Autista/genética , Sinapses , Hipocampo , Modelos Animais de Doenças
17.
Nat Commun ; 15(1): 2190, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38467602

RESUMO

The precise temporal coordination of neural activity is crucial for brain function. In the hippocampus, this precision is reflected in the oscillatory rhythms observed in CA1. While it is known that a balance between excitatory and inhibitory activity is necessary to generate and maintain these oscillations, the differential contribution of feedforward and feedback inhibition remains ambiguous. Here we use conditional genetics to chronically silence CA1 pyramidal cell transmission, ablating the ability of these neurons to recruit feedback inhibition in the local circuit, while recording physiological activity in mice. We find that this intervention leads to local pathophysiological events, with ripple amplitude and intrinsic frequency becoming significantly larger and spatially triggered local population spikes locked to the trough of the theta oscillation appearing during movement. These phenotypes demonstrate that feedback inhibition is crucial in maintaining local sparsity of activation and reveal the key role of lateral inhibition in CA1 in shaping circuit function.


Assuntos
Hipocampo , Células Piramidais , Camundongos , Animais , Retroalimentação , Hipocampo/fisiologia , Células Piramidais/fisiologia , Neurônios , Região CA1 Hipocampal/fisiologia , Interneurônios/fisiologia , Potenciais de Ação/fisiologia
18.
Sci Adv ; 10(2): eadk4741, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38198539

RESUMO

Adult neurogenesis confers the hippocampus with unparalleled neural plasticity, essential for intricate cognitive functions. The specific influence of sparse newborn neurons (NBNs) in modulating neural activities and subsequently steering behavior, however, remains obscure. Using an engineered NBN-tetanus toxin mouse model (NBN-TeTX), we noninvasively silenced NBNs, elucidating their crucial role in impulse inhibition and cognitive flexibility as evidenced through Morris water maze reversal learning and Go/Nogo task in operant learning. Task-based functional MRI (tb-fMRI) paired with operant learning revealed dorsal hippocampal hyperactivation during the Nogo task in male NBN-TeTX mice, suggesting that hippocampal hyperexcitability might underlie the observed behavioral deficits. Additionally, resting-state fMRI (rs-fMRI) exhibited enhanced functional connectivity between the dorsal and ventral dentate gyrus following NBN silencing. Further investigations into the activities of PV+ interneurons and mossy cells highlighted the indispensability of NBNs in maintaining the hippocampal excitation/inhibition balance. Our findings emphasize that the neural plasticity driven by NBNs extensively modulates the hippocampus, sculpting inhibitory control and cognitive flexibility.


Assuntos
Cognição , Neurônios , Masculino , Animais , Camundongos , Aprendizagem , Interneurônios , Transmissão Sináptica
19.
Proc Natl Acad Sci U S A ; 107(4): 1618-23, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20080609

RESUMO

To investigate the role of M1 muscarininc acetylcholine receptors (m1 receptors) in metabotropic glutamate receptor (mGluR)-mediated long-term depression (LTD), we produced mouse lines in which deletion of the m1 gene is restricted to the forebrain (FB-m1KO) or hippocampal CA3 pyramidal neurons (CA3-m1KO). Stimulation in FB-m1KO hippocampal slices resulted in excitatory postsynaptic potentials and long-term synaptic plasticity (long-term potentiation and LTD) similar to controls. The mice were deficient in (S)-3,5-dihydroxyphenylglycine hydrate (DHPG)-induced mGluR LTD, which correlated with a presynaptic increase in the release of neurotransmitters. Protein kinase C (PKC) activity, which is downstream from both mGluRs and m1 receptors, was reduced in CA3 but not in CA1. The presynaptic requirement of m1 receptors was confirmed by the lack of DHPG-induced mGluR LTD in the CA1 of slices from CA3-m1KO mice. mGluR LTD was rescued by stimulating PKC activity pharmacologically in CA3-m1KO mice. These data confirm a role for PKC activation in presynaptic induction of mGluR LTD and distinguish between the roles of mGluRs and m1 receptors.


Assuntos
Hipocampo/metabolismo , Depressão Sináptica de Longo Prazo , Receptor Muscarínico M1/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Camundongos , Camundongos Knockout , Proteína Quinase C/metabolismo , Receptor Muscarínico M1/deficiência , Receptores de N-Metil-D-Aspartato/metabolismo
20.
Learn Mem ; 19(4): 164-9, 2012 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-22419815

RESUMO

Recent studies focusing on the memory for temporal order have reported that CA1 plays a critical role in the memory for the sequences of events, in addition to its well-described role in spatial navigation. In contrast, CA3 was found to principally contribute to the memory for the association of items with spatial or contextual information in tasks focusing on spatial memory. Other studies have shown that NMDA signaling in the hippocampus is critical to memory performance in studies that have investigated spatial and temporal order memory independently. However, the role of NMDA signaling separately in CA1 and CA3 in memory that combines both spatial and temporal processing demands (episodic memory) has not been examined. Here we investigated the effect of the deletion of the NR1 subunit of the NMDA receptor in CA1 or CA3 on the spatial and the temporal aspects of episodic memory, using a behavioral task that allows for these two aspects of memory to be evaluated distinctly within the same task. Under these conditions, NMDA signaling in CA1 specifically contributes to the spatial aspect of memory function and is not required to support the memory for temporal order of events.


Assuntos
Região CA1 Hipocampal/fisiologia , Memória Episódica , N-Metilaspartato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/fisiologia , Animais , Região CA3 Hipocampal/fisiologia , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/análise
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa