Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
PLoS Biol ; 21(7): e3002221, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37498809

RESUMO

Investigation of brain function has been fueled by an accelerating development of novel technologies and tools. This Perspective looks at the unprecedented neurotechnological progress of the past 2 decades and discusses future strategies to elucidate brain function.


Assuntos
Neurociências , Encéfalo , Previsões , Tecnologia
2.
PLoS Biol ; 19(4): e3001146, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33793545

RESUMO

General anesthesia is characterized by reversible loss of consciousness accompanied by transient amnesia. Yet, long-term memory impairment is an undesirable side effect. How different types of general anesthetics (GAs) affect the hippocampus, a brain region central to memory formation and consolidation, is poorly understood. Using extracellular recordings, chronic 2-photon imaging, and behavioral analysis, we monitor the effects of isoflurane (Iso), medetomidine/midazolam/fentanyl (MMF), and ketamine/xylazine (Keta/Xyl) on network activity and structural spine dynamics in the hippocampal CA1 area of adult mice. GAs robustly reduced spiking activity, decorrelated cellular ensembles, albeit with distinct activity signatures, and altered spine dynamics. CA1 network activity under all 3 anesthetics was different to natural sleep. Iso anesthesia most closely resembled unperturbed activity during wakefulness and sleep, and network alterations recovered more readily than with Keta/Xyl and MMF. Correspondingly, memory consolidation was impaired after exposure to Keta/Xyl and MMF, but not Iso. Thus, different anesthetics distinctly alter hippocampal network dynamics, synaptic connectivity, and memory consolidation, with implications for GA strategy appraisal in animal research and clinical settings.


Assuntos
Anestésicos/efeitos adversos , Hipocampo/efeitos dos fármacos , Consolidação da Memória/efeitos dos fármacos , Coluna Vertebral/efeitos dos fármacos , Anestesia/efeitos adversos , Anestésicos/farmacologia , Animais , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Feminino , Fentanila/efeitos adversos , Fentanila/farmacologia , Hipocampo/citologia , Hipocampo/fisiologia , Isoflurano/efeitos adversos , Isoflurano/farmacologia , Ketamina/efeitos adversos , Ketamina/farmacologia , Masculino , Medetomidina/efeitos adversos , Medetomidina/farmacologia , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Midazolam/efeitos adversos , Midazolam/farmacologia , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Coluna Vertebral/fisiologia , Xilazina/efeitos adversos , Xilazina/farmacologia
3.
J Neurosci ; 42(4): 601-618, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34844990

RESUMO

Precise information flow from the hippocampus (HP) to prefrontal cortex (PFC) emerges during early development and accounts for cognitive processing throughout life. On flip side, this flow is selectively impaired in mental illness. In mouse models of psychiatric risk mediated by gene-environment interaction (GE), the prefrontal-hippocampal coupling is disrupted already shortly after birth. While this impairment relates to local miswiring in PFC and HP, it might be also because of abnormal connectivity between the two brain areas. Here, we test this hypothesis by combining in vivo electrophysiology and optogenetics with in-depth tracing of projections and monitor the morphology and function of hippocampal afferents in the PFC of control and GE mice of either sex throughout development. We show that projections from the hippocampal CA1 area preferentially target layer 5/6 pyramidal neurons and interneurons, and to a lesser extent layer 2/3 neurons of prelimbic cortex (PL), a subdivision of PFC. In neonatal GE mice, sparser axonal projections from CA1 pyramidal neurons with decreased release probability reach the PL. Their ability to entrain layer 5/6 oscillatory activity and firing is decreased. These structural and functional deficits of hippocampal-prelimbic connectivity persist, yet are less prominent in prejuvenile GE mice. Thus, besides local dysfunction of HP and PL, weaker connectivity between the two brain areas is present in GE mice throughout development.SIGNIFICANCE STATEMENT Poor cognitive performance in mental disorders comes along with prefrontal-hippocampal dysfunction. Recent data from mice that model the psychiatric risk mediated by gene-environment (GE) interaction identified the origin of deficits during early development, when the local circuits in both areas are compromised. Here, we show that sparser and less efficient connectivity as well as cellular dysfunction are the substrate of the weaker excitatory drive from hippocampus (HP) to prefrontal cortex (PFC) as well as of poorer oscillatory coupling between the two brain areas in these mice. While the structural and functional connectivity deficits persist during the entire development, their magnitude decreases with age. The results add experimental evidence for the developmental miswiring hypothesis of psychiatric disorders.


Assuntos
Interação Gene-Ambiente , Hipocampo/crescimento & desenvolvimento , Transtornos Mentais/genética , Transtornos Mentais/fisiopatologia , Rede Nervosa/crescimento & desenvolvimento , Córtex Pré-Frontal/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Hipocampo/química , Masculino , Transtornos Mentais/psicologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/química , Córtex Pré-Frontal/química , Fatores de Risco
4.
J Physiol ; 601(16): 3605-3630, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37434507

RESUMO

Cognitive processing relies on the functional refinement of the limbic circuitry during the first two weeks of life. During this developmental period, when the auditory, somatosensory and visual systems are still largely immature, the sense of olfaction acts as 'door to the world', providing an important source of environmental inputs. However, it is unknown whether early olfactory processing shapes the activity in the limbic circuitry during neonatal development. Here, we address this question by combining simultaneous in vivo recordings from the olfactory bulb (OB), lateral entorhinal cortex (LEC), hippocampus (HP) and prefrontal cortex (PFC) with olfactory stimulation as well as opto- and chemogenetic manipulations of mitral/tufted cells in the OB of non-anaesthetized neonatal mice of both sexes. We show that the neonatal OB synchronizes the limbic circuity in the beta frequency range. Moreover, it drives neuronal and network activity in LEC, as well as subsequently, HP and PFC via long-range projections from mitral cells to HP-projecting LEC neurons. Thus, OB activity shapes the communication within limbic circuits during neonatal development. KEY POINTS: During early postnatal development, oscillatory activity in the olfactory bulb synchronizes the limbic circuit. Olfactory stimulation boosts firing and beta synchronization along the olfactory bulb-lateral entorhinal cortex-hippocampal-prefrontal pathway. Mitral cells drive neuronal and network activity in the lateral entorhinal cortex (LEC), as well as subsequently, the hippocampus (HP) and the prefrontal cortex (PFC) via long-range projections from mitral cells to HP-projecting LEC neurons. Inhibition of vesicle release on LEC targeting mitral cell axons reveals direct involvement of LEC in the olfactory bulb-driven oscillatory entrainment of the limbic circuitry.


Assuntos
Bulbo Olfatório , Olfato , Camundongos , Animais , Masculino , Feminino , Olfato/fisiologia , Bulbo Olfatório/fisiologia , Córtex Entorrinal/fisiologia , Hipocampo , Neurônios/fisiologia
5.
J Physiol ; 601(4): 847-857, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36647326

RESUMO

Cognitive deficits in mental disorders result from dysfunctional activity in large-scale brain networks centred around the hippocampus and the prefrontal cortex. Dysfunctional activity emerges early during development and precedes the cognitive disabilities. The prefrontal-hippocampal network is driven by a prominent input from the lateral entorhinal cortex. We have previously shown that during early development, the entorhinal drive of the prefrontal-hippocampal network is impaired in a mouse model of mental disorders, yet the cellular substrate of this impairment is still poorly understood. Here, we address this question by a detailed characterization of projection neurons across the layers of the lateral entorhinal cortex in immune-challenged Disc1+/- mice at the beginning of the second postnatal week. We found that the activity and morphology of neurons in layers 2b and 3, which project to the hippocampus, are impaired. Neurons in layer 2b show increased spike-frequency adaptation, whereas neurons in layer 3 have reduced dendritic complexity but increased spike density. These findings identify the developmental alterations of entorhinal-hippocampal communication that underlie network dysfunction in immune-challenged Disc1+/- mice. KEY POINTS: Neonatal immune-challenged Disc1+/- mice show layer-specific changes in the lateral entorhinal cortex. Entorhinal layer 2b pyramidal neurons have increased spike-frequency adaptation. Reduced dendritic complexity but increased spine density characterize layer 3 pyramidal neurons.


Assuntos
Córtex Entorrinal , Hipocampo , Camundongos , Animais , Córtex Entorrinal/fisiologia , Hipocampo/fisiologia , Neurônios/fisiologia , Células Piramidais , Córtex Pré-Frontal , Proteínas do Tecido Nervoso
6.
Mol Psychiatry ; 27(11): 4707-4721, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36123424

RESUMO

The precise development of the neocortex is a prerequisite for higher cognitive and associative functions. Despite numerous advances that have been made in understanding neuronal differentiation and cortex development, our knowledge regarding the impact of specific genes associated with neurodevelopmental disorders on these processes is still limited. Here, we show that Taok2, which is encoded in humans within the autism spectrum disorder (ASD) susceptibility locus 16p11.2, is essential for neuronal migration. Overexpression of de novo mutations or rare variants from ASD patients disrupts neuronal migration in an isoform-specific manner. The mutated TAOK2α variants but not the TAOK2ß variants impaired neuronal migration. Moreover, the TAOK2α isoform colocalizes with microtubules. Consequently, neurons lacking Taok2 have unstable microtubules with reduced levels of acetylated tubulin and phosphorylated JNK1. Mice lacking Taok2 develop gross cortical and cortex layering abnormalities. Moreover, acute Taok2 downregulation or Taok2 knockout delayed the migration of upper-layer cortical neurons in mice, and the expression of a constitutively active form of JNK1 rescued these neuronal migration defects. Finally, we report that the brains of the Taok2 KO and 16p11.2 del Het mouse models show striking anatomical similarities and that the heterozygous 16p11.2 microdeletion mouse model displayed reduced levels of phosphorylated JNK1 and neuronal migration deficits, which were ameliorated upon the introduction of TAOK2α in cortical neurons and in the developing cortex of those mice. These results delineate the critical role of TAOK2 in cortical development and its contribution to neurodevelopmental disorders, including ASD.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Neocórtex , Proteínas Serina-Treonina Quinases , Animais , Humanos , Camundongos , Transtorno do Espectro Autista/genética , Transtorno Autístico/genética , Modelos Animais de Doenças , Microtúbulos/genética , Microtúbulos/metabolismo , Neocórtex/metabolismo , Neurogênese/genética , Neurogênese/fisiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
7.
J Neurosci ; 41(5): 813-822, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33431633

RESUMO

The sensory and cognitive abilities of the mammalian neocortex are underpinned by intricate columnar and laminar circuits formed from an array of diverse neuronal populations. One approach to determining how interactions between these circuit components give rise to complex behavior is to investigate the rules by which cortical circuits are formed and acquire functionality during development. This review summarizes recent research on the development of the neocortex, from genetic determination in neural stem cells through to the dynamic role that specific neuronal populations play in the earliest circuits of neocortex, and how they contribute to emergent function and cognition. While many of these endeavors take advantage of model systems, consideration will also be given to advances in our understanding of activity in nascent human circuits. Such cross-species perspective is imperative when investigating the mechanisms underlying the dysfunction of early neocortical circuits in neurodevelopmental disorders, so that one can identify targets amenable to therapeutic intervention.


Assuntos
Neocórtex/citologia , Neocórtex/crescimento & desenvolvimento , Rede Nervosa/citologia , Rede Nervosa/crescimento & desenvolvimento , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Animais , Humanos , Lógica
8.
PLoS Biol ; 17(1): e2006994, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30703080

RESUMO

Although the developmental principles of sensory and cognitive processing have been extensively investigated, their synergy has been largely neglected. During early life, most sensory systems are still largely immature. As a notable exception, the olfactory system is functional at birth, controlling mother-offspring interactions and neonatal survival. Here, we elucidate the structural and functional principles underlying the communication between olfactory bulb (OB) and lateral entorhinal cortex (LEC)-the gatekeeper of limbic circuitry-during neonatal development. Combining optogenetics, pharmacology, and electrophysiology in vivo with axonal tracing, we show that mitral cell-dependent discontinuous theta bursts in OB drive network oscillations and time the firing in LEC of anesthetized mice via axonal projections confined to upper cortical layers. Acute pharmacological silencing of OB activity diminishes entorhinal oscillations, whereas odor exposure boosts OB-entorhinal coupling at fast frequencies. Chronic impairment of olfactory sensory neurons disrupts OB-entorhinal activity. Thus, OB activity shapes the maturation of entorhinal circuits.


Assuntos
Bulbo Olfatório/fisiologia , Córtex Olfatório/fisiologia , Olfato/fisiologia , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Fenômenos Eletrofisiológicos/fisiologia , Córtex Entorrinal/metabolismo , Córtex Entorrinal/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Odorantes , Córtex Olfatório/metabolismo , Optogenética/métodos , Ritmo Teta/fisiologia
9.
Cereb Cortex ; 31(2): 1240-1258, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33037815

RESUMO

Disrupted-in-schizophrenia 1 (DISC1) gene represents an intracellular hub of developmental processes. When combined with early environmental stressors, such as maternal immune activation, but not in the absence of thereof, whole-brain DISC1 knock-down leads to memory and executive deficits as result of impaired prefrontal-hippocampal communication throughout development. While synaptic dysfunction in neonatal prefrontal cortex (PFC) has been recently identified as one source of abnormal long-range coupling, the contribution of hippocampus (HP) is still unknown. Here, we aim to fill this knowledge gap by combining in vivo electrophysiology and optogenetics with morphological and behavioral assessment of immune-challenged mice with DISC1 knock-down either in the whole brain (GE) or restricted to pyramidal neurons in hippocampal CA1 area (GHPE). We found abnormal network activity, sharp-waves, and neuronal firing in CA1 that complement the deficits in upper layer of PFC. Moreover, optogenetic activating CA1 pyramidal neurons fails to activate the prefrontal local circuits. These deficits that persist till prejuvenile age relate to dendrite sparsification and loss of spines of CA1 pyramidal neurons. As a long-term consequence, DISC1 knock-down in HP leads to poorer recognition memory at prejuvenile age. Thus, DISC1-controlled developmental processes in HP in immune-challenged mice are critical for circuit function and cognitive behavior.


Assuntos
Cognição/fisiologia , Comportamento Exploratório/fisiologia , Técnicas de Silenciamento de Genes/métodos , Hipocampo/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/deficiência , Córtex Pré-Frontal/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Feminino , Hipocampo/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/imunologia , Córtex Pré-Frontal/imunologia , Gravidez , Células Piramidais/fisiologia
10.
J Neurosci ; 40(21): 4203-4218, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32312886

RESUMO

The accessory olfactory system controls social and sexual behavior. In the mouse accessory olfactory bulb, the first central stage of information processing along the accessory olfactory pathway, projection neurons (mitral cells) display infra-slow oscillatory discharge with remarkable periodicity. The physiological mechanisms that underlie this default output state, however, remain controversial. Moreover, whether such rhythmic infra-slow activity patterns exist in awake behaving mice and whether such activity reflects the functional organization of the accessory olfactory bulb circuitry remain unclear. Here, we hypothesize that mitral cell ensembles form synchronized microcircuits that subdivide the accessory olfactory bulb into segregated functional clusters. We use a miniature microscope to image the Ca2+ dynamics within the apical dendritic compartments of large mitral cell ensembles in vivo We show that infra-slow periodic patterns of concerted neural activity, indeed, reflect the idle state of accessory olfactory bulb output in awake male and female mice. Ca2+ activity profiles are distinct and glomerulus-specific. Confocal time-lapse imaging in acute slices reveals that groups of mitral cells assemble into microcircuits that exhibit correlated Ca2+ signals. Moreover, electrophysiological profiling of synaptic connectivity indicates functional coupling between mitral cells. Our results suggest that both intrinsically rhythmogenic neurons and neurons entrained by fast synaptic drive are key elements in organizing the accessory olfactory bulb into functional microcircuits, each characterized by a distinct default pattern of infra-slow rhythmicity.SIGNIFICANCE STATEMENT Information processing in the accessory olfactory bulb (AOB) plays a central role in conspecific chemosensory communication. Surprisingly, many basic physiological principles that underlie neuronal signaling in the AOB remain elusive. Here, we show that AOB projection neurons (mitral cells) form parallel synchronized ensembles both in vitro and in vivo Infra-slow synchronous oscillatory activity within AOB microcircuits thus adds a new dimension to chemosensory coding along the accessory olfactory pathway.


Assuntos
Rede Nervosa/fisiologia , Neurônios/fisiologia , Bulbo Olfatório/fisiologia , Condutos Olfatórios/fisiologia , Potenciais de Ação/fisiologia , Animais , Camundongos
11.
J Neurosci ; 39(7): 1222-1235, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30617212

RESUMO

Compromised brain development has been hypothesized to account for mental illness. This concept was underpinned by the function of the molecule disrupted-in-schizophrenia 1 (DISC1), which represents an intracellular hub of developmental processes and has been related to cognitive dysfunction in psychiatric disorders. Mice with whole-brain DISC1 knock-down show impaired prefrontal-hippocampal function and cognitive abilities throughout development and at adulthood, especially when combined with early environmental stressors, such as maternal immune activation (MIA). However, the contribution of abnormal DISC1-driven maturation of either prefrontal cortex (PFC) or hippocampus (HP) to these deficits is still unknown. Here, we use in utero electroporation to restrict the DISC1 knock-down to prefrontal layer II/III pyramidal neurons during perinatal development and expose these mice to MIA as an environmental stressor (dual-hit GPFCE mice, both sexes). Combining in vivo electrophysiology and neuroanatomy with behavioral testing, we show that GPFCE mice at neonatal age have abnormal patterns of oscillatory activity and firing in PFC, but not HP. Abnormal firing rates in PFC of GPFCE mice relate to sparser dendritic arborization and lower spine density. Moreover, the long-range coupling within prefrontal-hippocampal networks is decreased at this age. The transient prefrontal DISC1 knock-down was sufficient to permanently perturb the prefrontal-hippocampal communication and caused poorer recognition memory performance at pre-juvenile age. Thus, developmental dysfunction of prefrontal circuitry causes long-lasting disturbances related to mental illness.SIGNIFICANCE STATEMENT Hypofrontality is considered a main cause of cognitive deficits in mental disorders, yet the underlying mechanisms are still largely unknown. During development, long before the emergence of disease symptoms, the functional coupling within the prefrontal-hippocampal network, which is the core brain circuit involved in cognitive processing, is reduced. To assess to which extent impaired prefrontal development contributes to the early dysfunction, immune-challenged mice with transient DISC1 knock-down confined to PFC were investigated in their prefrontal-hippocampal communication throughout development by in vivo electrophysiology and behavioral testing. We show that perturbing developmental processes of prefrontal layer II/III pyramidal neurons is sufficient to diminish prefrontal-hippocampal coupling and decrease the cognitive performance throughout development.


Assuntos
Disfunção Cognitiva/genética , Proteínas do Tecido Nervoso/genética , Animais , Comportamento Animal/fisiologia , Disfunção Cognitiva/psicologia , Comportamento Exploratório/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/crescimento & desenvolvimento , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Células Piramidais/ultraestrutura , Reconhecimento Psicológico/fisiologia
12.
J Physiol ; 598(24): 5753-5769, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32926437

RESUMO

KEY POINTS: During early postnatal development, mitral cells show either irregular bursting or non-bursting firing patterns Bursting mitral cells preferentially fire during theta bursts in the neonatal olfactory bulb, being locked to the theta phase Bursting mitral cells preferentially fire during theta bursts in the neonatal lateral entorhinal cortex and are temporally related to both respiration rhythm- and theta phase Bursting mitral cells act as a cellular substrate of the olfactory drive that promotes the oscillatory entrainment of entorhinal networks ABSTRACT: Shortly after birth, the olfactory system provides not only the main source of environmental inputs to blind, deaf, non-whisking and motorically-limited rodents, but also the drive boosting the functional entrainment of limbic circuits. However, the cellular substrate of this early communication remains largely unknown. Here, we combine in vivo and in vitro patch-clamp and extracellular recordings to reveal the contribution of mitral cell (MC) firing to early patterns of network activity in both the neonatal olfactory bulb (OB) and the lateral entorhinal cortex (LEC), the gatekeeper of limbic circuits. We show that MCs predominantly fire either in an irregular bursting or non-bursting pattern during discontinuous theta events in the OB. However, the temporal spike-theta phase coupling is stronger for bursting than non-bursting MCs. In line with the direct OB-to-LEC projections, both bursting and non-bursting discharge augments during co-ordinated patterns of entorhinal activity, albeit with higher magnitude for bursting MCs. For these neurons, temporal coupling to the discontinuous theta events in the LEC is stronger. Thus, bursting MCs might drive the entrainment of the OB-LEC network during neonatal development.


Assuntos
Bulbo Olfatório , Olfato , Potenciais de Ação , Animais , Animais Recém-Nascidos , Córtex Entorrinal , Camundongos
13.
Mol Psychiatry ; 24(9): 1329-1350, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-29467497

RESUMO

Atypical brain connectivity is a major contributor to the pathophysiology of neurodevelopmental disorders (NDDs) including autism spectrum disorders (ASDs). TAOK2 is one of several genes in the 16p11.2 microdeletion region, but whether it contributes to NDDs is unknown. We performed behavioral analysis on Taok2 heterozygous (Het) and knockout (KO) mice and found gene dosage-dependent impairments in cognition, anxiety, and social interaction. Taok2 Het and KO mice also have dosage-dependent abnormalities in brain size and neural connectivity in multiple regions, deficits in cortical layering, dendrite and synapse formation, and reduced excitatory neurotransmission. Whole-genome and -exome sequencing of ASD families identified three de novo mutations in TAOK2 and functional analysis in mice and human cells revealed that all the mutations impair protein stability, but they differentially impact kinase activity, dendrite growth, and spine/synapse development. Mechanistically, loss of Taok2 activity causes a reduction in RhoA activation, and pharmacological enhancement of RhoA activity rescues synaptic phenotypes. Together, these data provide evidence that TAOK2 is a neurodevelopmental disorder risk gene and identify RhoA signaling as a mediator of TAOK2-dependent synaptic development.


Assuntos
Transtorno do Espectro Autista/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Adulto , Animais , Ansiedade/genética , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/patologia , Transtorno do Espectro Autista/psicologia , Criança , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Disfunção Cognitiva/psicologia , Dendritos/metabolismo , Dendritos/patologia , Feminino , Humanos , Relações Interpessoais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Transtornos do Neurodesenvolvimento/psicologia , Neurogênese , Fenótipo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Transmissão Sináptica , Sequenciamento do Exoma
14.
Eur J Neurosci ; 50(6): 3072-3084, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31087437

RESUMO

Despite inherent difficulties to translate human cognitive phenotype into animals, a large number of animal models for psychiatric disorders, such as schizophrenia, have been developed over the last decades. To which extent they reproduce common patterns of dysfunction related to mental illness and abnormal processes of maturation is still largely unknown. While the devastating symptoms of disease are firstly detectable in adulthood, they are considered to reflect profound miswiring of brain circuitry as result of abnormal development. To reveal whether different disease models share common dysfunction early in life, we investigate the prefrontal-hippocampal communication at neonatal age in (a) mice mimicking the abnormal genetic background (22q11.2 microdeletion, DISC1 knockdown), (b) mice mimicking the challenge by environmental stressors (maternal immune activation during pregnancy), (c) mice mimicking the combination of both aetiologies (dual-hit models) and pharmacological mouse models. Simultaneous extracellular recordings in vivo from all layers of prelimbic subdivision (PL) of prefrontal cortex (PFC) and CA1 area of intermediate/ventral hippocampus (i/vHP) show that network oscillations have a more fragmented structure and decreased power mainly in neonatal mice that mimic both genetic and environmental aetiology of disease. These mice also show layer-specific firing deficits in PL. Similar early network dysfunction was present in mice with 22q11.2 microdeletion. The abnormal activity patterns are accompanied by weaker synchrony and directed interactions within prefrontal-hippocampal networks. Thus, only severe genetic defects or combined genetic environmental stressors are disruptive enough for reproducing the early network miswiring in mental disorders.


Assuntos
Hipocampo/fisiopatologia , Transtornos Mentais/fisiopatologia , Rede Nervosa/fisiopatologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiopatologia , Potenciais de Ação/fisiologia , Animais , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Hipocampo/crescimento & desenvolvimento , Camundongos , Rede Nervosa/crescimento & desenvolvimento , Córtex Pré-Frontal/crescimento & desenvolvimento
15.
PLoS Biol ; 13(11): e1002304, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26600123

RESUMO

Optimal behavior relies on the combination of inputs from multiple senses through complex interactions within neocortical networks. The ontogeny of this multisensory interplay is still unknown. Here, we identify critical factors that control the development of visual-tactile processing by combining in vivo electrophysiology with anatomical/functional assessment of cortico-cortical communication and behavioral investigation of pigmented rats. We demonstrate that the transient reduction of unimodal (tactile) inputs during a short period of neonatal development prior to the first cross-modal experience affects feed-forward subcortico-cortical interactions by attenuating the cross-modal enhancement of evoked responses in the adult primary somatosensory cortex. Moreover, the neonatal manipulation alters cortico-cortical interactions by decreasing the cross-modal synchrony and directionality in line with the sparsification of direct projections between primary somatosensory and visual cortices. At the behavioral level, these functional and structural deficits resulted in lower cross-modal matching abilities. Thus, neonatal unimodal experience during defined developmental stages is necessary for setting up the neuronal networks of multisensory processing.


Assuntos
Neocórtex/fisiopatologia , Rede Nervosa/fisiopatologia , Transtornos de Sensação/etiologia , Privação Sensorial , Córtex Somatossensorial/fisiopatologia , Distúrbios Somatossensoriais/etiologia , Animais , Animais Recém-Nascidos , Comportamento Animal , Potenciais Somatossensoriais Evocados , Comportamento Exploratório , Feminino , Masculino , Neocórtex/patologia , Rede Nervosa/patologia , Neurônios/patologia , Ratos Endogâmicos BN , Reconhecimento Psicológico , Transtornos de Sensação/patologia , Transtornos de Sensação/fisiopatologia , Córtex Somatossensorial/patologia , Distúrbios Somatossensoriais/patologia , Distúrbios Somatossensoriais/fisiopatologia , Tato , Percepção do Tato , Vibrissas/lesões , Percepção Visual
16.
Cereb Cortex ; 27(2): 1011-1026, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26637449

RESUMO

The fate of the subplate (SP) is still a matter of debate. The SP and layer 6 (which is ontogenetically the oldest and innermost neocortical lamina) develop coincidentally. Yet, the function of sublamina 6B is largely unknown. It has been suggested that it consists partly of neurons from the transient SP, however, experimental evidence for this hypothesis is still missing. To obtain first insights into the neuronal complement of layer 6B in the somatosensory rat barrel cortex, we used biocytin stainings of SP neurons (aged 0-4 postnatal days, PND) and layer 6B neurons (PND 11-35) obtained during in vitro whole-cell patch-clamp recordings. Neurons were reconstructed for a quantitative characterization of their axonal and dendritic morphology. An unsupervised cluster analysis revealed that the SP and layer 6B consist of heterogeneous but comparable neuronal cell populations. Both contain 5 distinct spine-bearing cell types whose relative fractions change with increasing age. Pyramidal cells were more prominent in layer 6B, whereas non-pyramidal neurons were less frequent. Because of the high morphological similarity of SP and layer 6B neurons, we suggest that layer 6B consists of persistent non-pyramidal neurons from the SP and cortical L6B pyramidal neurons.


Assuntos
Neocórtex/anatomia & histologia , Animais , Animais Recém-Nascidos , Axônios/fisiologia , Axônios/ultraestrutura , Polaridade Celular , Dendritos/fisiologia , Dendritos/ultraestrutura , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Processamento de Imagem Assistida por Computador , Neocórtex/citologia , Neocórtex/diagnóstico por imagem , Neurônios/fisiologia , Técnicas de Patch-Clamp , Células Piramidais/fisiologia , Células Piramidais/ultraestrutura , Ratos , Ratos Wistar , Córtex Somatossensorial/anatomia & histologia , Córtex Somatossensorial/citologia , Córtex Somatossensorial/crescimento & desenvolvimento
17.
J Neurosci ; 36(13): 3676-90, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27030754

RESUMO

Precise information flow during mnemonic and executive tasks requires the coactivation of adult prefrontal and hippocampal networks in oscillatory rhythms. This interplay emerges early in life, most likely as an anticipatory template of later cognitive performance. At neonatal age, hippocampal theta bursts drive the generation of prefrontal theta-gamma oscillations. In the absence of direct reciprocal interactions, the question arises of which feedback mechanisms control the early entrainment of prefrontal-hippocampal networks. Here, we demonstrate that prefrontal-hippocampal activity couples with discontinuous theta oscillations and neuronal firing in both lateral entorhinal cortex and ventral midline thalamic nuclei of neonatal rats. However, these two brain areas have different contributions to the neonatal long-range communication. The entorhinal cortex mainly modulates the hippocampal activity via direct axonal projections. In contrast, thalamic theta bursts are controlled by the prefrontal cortex via mutual projections and contribute to hippocampal activity. Thus, the neonatal prefrontal cortex modulates the level of hippocampal activation by directed interactions with the ventral midline thalamus. Similar to the adult task-related communication, theta-band activity ensures the feedback control of long-range coupling in the developing brain. SIGNIFICANCE STATEMENT: Memories are encoded by finely tuned interactions within large-scale neuronal networks. This cognitive performance is not inherited, but progressively matures in relationship with the establishment of long-range coupling in the immature brain. The hippocampus initiates and unidirectionally drives the oscillatory entrainment of neonatal prefrontal cortex, yet feedback interactions that precisely control this early communication are still unresolved. Here, we identified distinct roles of entorhinal cortex and ventral midline thalamus for the functional development of prefrontal-hippocampal interactions. While entorhinal oscillations modulate the hippocampal activity by timing the neuronal firing via monosynaptic afferents, thalamic nuclei act as a relay station routing prefrontal activation back to hippocampus. Understanding the mechanisms of network maturation represents the prerequisite for assessing circuit dysfunction in neurodevelopmental disorders.


Assuntos
Mapeamento Encefálico , Córtex Entorrinal/fisiologia , Hipocampo/fisiologia , Vias Neurais/fisiologia , Córtex Pré-Frontal/fisiologia , Tálamo/fisiologia , Potenciais de Ação , Análise de Variância , Animais , Animais Recém-Nascidos , Masculino , Neurônios/fisiologia , Córtex Pré-Frontal/lesões , Ratos , Ratos Wistar , Estilbamidinas/metabolismo , Tálamo/lesões , Fatores de Tempo
18.
J Neurosci ; 36(11): 3127-44, 2016 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-26985025

RESUMO

The accessory olfactory system controls social and sexual behavior. However, key aspects of sensory signaling along the accessory olfactory pathway remain largely unknown. Here, we investigate patterns of spontaneous neuronal activity in mouse accessory olfactory bulb mitral cells, the direct neural link between vomeronasal sensory input and limbic output. Both in vitro and in vivo, we identify a subpopulation of mitral cells that exhibit slow stereotypical rhythmic discharge. In intrinsically rhythmogenic neurons, these periodic activity patterns are maintained in absence of fast synaptic drive. The physiological mechanism underlying mitral cell autorhythmicity involves cyclic activation of three interdependent ionic conductances: subthreshold persistent Na(+) current, R-type Ca(2+) current, and Ca(2+)-activated big conductance K(+) current. Together, the interplay of these distinct conductances triggers infraslow intrinsic oscillations with remarkable periodicity, a default output state likely to affect sensory processing in limbic circuits. SIGNIFICANCE STATEMENT: We show for the first time that some rodent accessory olfactory bulb mitral cells-the direct link between vomeronasal sensory input and limbic output-are intrinsically rhythmogenic. Driven by ≥ 3 distinct interdependent ionic conductances, infraslow intrinsic oscillations show remarkable periodicity both in vitro and in vivo. As a novel default state, infraslow autorhythmicity is likely to affect limbic processing of pheromonal information.


Assuntos
Potenciais de Ação/fisiologia , Neurônios/fisiologia , Bulbo Olfatório/citologia , Condutos Olfatórios/fisiologia , Periodicidade , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Fármacos Cardiovasculares/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Fosfolipases A2 do Grupo II , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Neurônios/classificação , Neurônios/efeitos dos fármacos , Pirimidinas/farmacologia , Venenos de Aranha/farmacologia , Valina/análogos & derivados , Valina/farmacologia , ômega-Agatoxina IVA/farmacologia
19.
Synapse ; 71(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28105686

RESUMO

The presumptive unisensory neocortical areas process multisensory information by oscillatory entrainment of neuronal networks via direct cortico-cortical projections. While neonatal unimodal experience has been identified as necessary for setting up the neuronal networks of multisensory processing, it is still unclear whether early cross-modal experience equally controls the ontogeny of multisensory processing. Here, we assess the development of visual-somatosensory interactions and their anatomical substrate by performing extracellular recordings of network activity in primary sensory cortices in vivo and assessing the cortico-cortical connectivity in pigmented rats. Similar to adult animals, juvenile rats with minimal cross-modal experience display supra-additive augmentation of evoked responses, time-dependent modulation of power and phase reset of network oscillations in response to cross-modal light and whisker stimulation. Moreover, the neuronal discharge of individual neurons is stronger coupled to theta and alpha network oscillations after visual-tactile stimuli. The adult-like multisensory processing of juvenile rats relies on abundant direct visual-somatosensory connections and thalamocortical feedforward interactions. Thus, cellular and network interactions ensuring multisensory processing emerge before cross-modal experience and refine during juvenile development.


Assuntos
Córtex Somatossensorial/fisiologia , Percepção do Tato , Percepção Visual , Animais , Potenciais Somatossensoriais Evocados , Potenciais Evocados Visuais , Retroalimentação Fisiológica , Feminino , Masculino , Ratos , Córtex Somatossensorial/crescimento & desenvolvimento , Córtex Visual/crescimento & desenvolvimento , Córtex Visual/fisiologia
20.
Cereb Cortex ; 26(11): 4265-4281, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27613435

RESUMO

Cognitive deficits represent a major burden of neuropsychiatric disorders and result in part from abnormal communication within hippocampal-prefrontal circuits. While it has been hypothesized that this network dysfunction arises during development, long before the first clinical symptoms, experimental evidence is still missing. Here, we show that pre-juvenile mice mimicking genetic and environmental risk factors of disease (dual-hit GE mice) have poorer recognition memory that correlates with augmented coupling by synchrony and stronger directed interactions between prefrontal cortex and hippocampus. The network dysfunction emerges already during neonatal development, yet it initially consists in a diminished hippocampal theta drive and consequently, a weaker and disorganized entrainment of local prefrontal circuits in discontinuous oscillatory activity in dual-hit GE mice when compared with controls. Thus, impaired maturation of functional communication within hippocampal-prefrontal networks switching from hypo- to hyper-coupling may represent a mechanism underlying the pathophysiology of cognitive deficits in neuropsychiatric disorders.


Assuntos
Transtornos Cognitivos , Deficiências do Desenvolvimento , Interação Gene-Ambiente , Hipocampo/fisiologia , Vias Neurais/fisiologia , Córtex Pré-Frontal/fisiopatologia , Animais , Animais Recém-Nascidos , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Deficiências do Desenvolvimento/induzido quimicamente , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/genética , Modelos Animais de Doenças , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/genética , Feminino , Hipocampo/efeitos dos fármacos , Indutores de Interferon/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/efeitos dos fármacos , Poli I-C/toxicidade , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA