Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 148(20)2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34557899

RESUMO

The inhibitory GABAergic system in the brain is involved in the etiology of various psychiatric problems, including autism spectrum disorders (ASD), attention deficit hyperactivity disorder (ADHD) and others. These disorders are influenced not only by genetic but also by environmental factors, such as preterm birth, although the underlying mechanisms are not known. In a translational hyperoxia model, exposing mice pups at P5 to 80% oxygen for 48 h to mimic a steep rise of oxygen exposure caused by preterm birth from in utero into room air, we documented a persistent reduction of cortical mature parvalbumin-expressing interneurons until adulthood. Developmental delay of cortical myelin was observed, together with decreased expression of oligodendroglial glial cell-derived neurotrophic factor (GDNF), a factor involved in interneuronal development. Electrophysiological and morphological properties of remaining interneurons were unaffected. Behavioral deficits were observed for social interaction, learning and attention. These results demonstrate that neonatal oxidative stress can lead to decreased interneuron density and to psychiatric symptoms. The obtained cortical myelin deficit and decreased oligodendroglial GDNF expression indicate that an impaired oligodendroglial-interneuronal interplay contributes to interneuronal damage.


Assuntos
Lesões Encefálicas/metabolismo , Neurônios GABAérgicos/metabolismo , Hiperóxia/metabolismo , Interneurônios/metabolismo , Parvalbuminas/metabolismo , Nascimento Prematuro/metabolismo , Roedores/metabolismo , Animais , Linhagem Celular , Cognição/fisiologia , Modelos Animais de Doenças , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodendroglia/metabolismo , Comportamento Social
2.
J Neurosci ; 42(15): 3253-3270, 2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35241491

RESUMO

Anti-NMDA receptor (NMDAR) encephalitis is a severe neuropsychiatric disorder associated with autoantibodies against NMDARs, which cause a variety of symptoms from prominent psychiatric and cognitive manifestations to seizures and autonomic instability. Previous studies mainly focused on hippocampal effects of these autoantibodies, helping to explain mechanistic causes for cognitive impairment. However, antibodies' effects on higher cortical network function, where they could contribute to psychosis and/or seizures, have not been explored in detail until now. Here, we employed a patient-derived monoclonal antibody targeting the NR1 subunit of NMDAR and tested its effects on in vitro cultures of rodent cortical neurons, using imaging and electrophysiological techniques. We report that this hNR1 antibody drives cortical networks to a hyperexcitable state and disrupts mechanisms stabilizing network activity such as Npas4 signaling. Network hyperactivity is in part a result of a reduced synaptic output of inhibitory neurons, as indicated by a decreased inhibitory drive and levels of presynaptic inhibitory proteins, specifically in inhibitory-to-excitatory neuron synapses. Importantly, on a single-cell level hNR1 antibody selectively impairs NMDAR-mediated currents and synaptic transmission of cortical inhibitory neurons, yet has no effect on excitatory neurons, which contrasts with its effects on hippocampal neurons. Together, these findings provide a novel, cortex-specific mechanism of antibody-induced neuronal hyperexcitability, highlighting regional specificity underlying the pathology of autoimmune encephalitis.SIGNIFICANCE STATEMENT It is increasingly appreciated that the inadvertent activation of the immune system within CNS can underlie pathogenesis of neuropsychiatric disorders. Although the exact mechanisms remain elusive, autoantibodies derived from patients with autoimmune encephalitis pose a unique tool to study pathogenesis of neuropsychiatric states. Our analysis reveals that autoantibody against the NMDA receptor (NMDAR) has a distinct mechanism of action in the cortex, where it impairs function of inhibitory neurons leading to increased cortical network excitability, in contrast to previously described hippocampal synaptic mechanisms of information encoding, highlighting brain regional specificity. Notably, similar mechanism of NMDAR-mediated inhibitory hypofunction leading to cortical disinhibition has been suggested to underlie pathology of schizophrenia, hence our data provide new evidence for common mechanisms underlying neuropsychiatric disorders.


Assuntos
Encefalite , Receptores de N-Metil-D-Aspartato , Autoanticorpos/metabolismo , Doença de Hashimoto , Humanos , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/metabolismo
3.
Development ; 147(22)2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-33060132

RESUMO

The mammalian cortex is populated by neurons derived from neural progenitors located throughout the embryonic telencephalon. Excitatory neurons are derived from the dorsal telencephalon, whereas inhibitory interneurons are generated in its ventral portion. The transcriptional regulator PRDM16 is expressed by radial glia, neural progenitors present in both regions; however, its mechanisms of action are still not fully understood. It is unclear whether PRDM16 plays a similar role in neurogenesis in both dorsal and ventral progenitor lineages and, if so, whether it regulates common or unique networks of genes. Here, we show that Prdm16 expression in mouse medial ganglionic eminence (MGE) progenitors is required for maintaining their proliferative capacity and for the production of proper numbers of forebrain GABAergic interneurons. PRDM16 binds to cis-regulatory elements and represses the expression of region-specific neuronal differentiation genes, thereby controlling the timing of neuronal maturation. PRDM16 regulates convergent developmental gene expression programs in the cortex and MGE, which utilize both common and region-specific sets of genes to control the proliferative capacity of neural progenitors, ensuring the generation of correct numbers of cortical neurons.


Assuntos
Córtex Cerebral/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Células-Tronco Neurais/metabolismo , Fatores de Transcrição/metabolismo , Animais , Córtex Cerebral/citologia , Proteínas de Ligação a DNA/genética , Neurônios GABAérgicos/citologia , Interneurônios/citologia , Camundongos , Células-Tronco Neurais/citologia , Fatores de Transcrição/genética
4.
Neurobiol Dis ; 157: 105447, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34274461

RESUMO

Huntington's disease (HD) is a progressive, fatal neurodegenerative disorder characterized by motor, cognitive, and psychiatric disturbances. There is no known cure for HD, but its progressive nature allows for early therapeutic intervention. Currently, much of the research has focused on the striatum, however, there is evidence suggesting that disruption of thalamocortical circuits could underlie some of the early symptoms of HD. Loss of both cortical pyramidal neurons (CPNs) and thalamic neurons occurs in HD patients, and cognitive, somatosensory, and attention deficits precede motor abnormalities. However, the role of thalamocortical pathways in HD progression has been understudied. Here, we measured single unit activity and local field potentials (LFPs) from electrode arrays implanted in the thalamus and primary motor cortex of 4-5 month-old male and female Q175 mice. We assessed neuronal activity under baseline conditions as well as during presentation of rewards delivered via actuation of an audible solenoid valve. HD mice showed a significantly delayed licking response to the reward stimulus. At the same time, neuronal activation to the reward was delayed in thalamic neurons, CPNs and fast-spiking cortical interneurons (FSIs) of HD mice. In addition, thalamocortical coherence increased at lower frequencies in HD relative to wildtype mice. Together, these data provide evidence that impaired cortical and thalamic responses to reward stimuli, and impaired thalamocortical coherence, may play an important early role in motor, cognitive, and learning deficits in HD patients.


Assuntos
Doença de Huntington/fisiopatologia , Córtex Motor/fisiopatologia , Tálamo/fisiopatologia , Animais , Córtex Cerebral/fisiopatologia , Cognição , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Introdução de Genes , Interneurônios/fisiologia , Camundongos , Atividade Motora , Vias Neurais/fisiopatologia , Técnicas de Patch-Clamp , Células Piramidais/fisiologia
5.
Int J Mol Sci ; 22(17)2021 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-34502208

RESUMO

In the mammalian brain, cortical interneurons (INs) are a highly diverse group of cells. A key neurophysiological question concerns how each class of INs contributes to cortical circuit function and whether specific roles can be attributed to a selective cell type. To address this question, researchers are integrating knowledge derived from transcriptomic, histological, electrophysiological, developmental, and functional experiments to extensively characterise the different classes of INs. Our hope is that such knowledge permits the selective targeting of cell types for therapeutic endeavours. This review will focus on two of the main types of INs, namely the parvalbumin (PV+) or somatostatin (SOM+)-containing cells, and summarise the research to date on these classes.


Assuntos
Morte Celular , Córtex Cerebral/patologia , Interneurônios/patologia , Parvalbuminas/metabolismo , Somatostatina/metabolismo , Animais , Córtex Cerebral/metabolismo , Humanos , Interneurônios/metabolismo
6.
J Neurosci ; 39(39): 7648-7663, 2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31346031

RESUMO

Correlated electrical activity in neurons is a prominent characteristic of cortical microcircuits. Despite a growing amount of evidence concerning both spike-count and subthreshold membrane potential pairwise correlations, little is known about how different types of cortical neurons convert correlated inputs into correlated outputs. We studied pyramidal neurons and two classes of GABAergic interneurons of layer 5 in neocortical brain slices obtained from rats of both sexes, and we stimulated them with biophysically realistic correlated inputs, generated using dynamic clamp. We found that the physiological differences between cell types manifested unique features in their capacity to transfer correlated inputs. We used linear response theory and computational modeling to gain clear insights into how cellular properties determine both the gain and timescale of correlation transfer, thus tying single-cell features with network interactions. Our results provide further ground for the functionally distinct roles played by various types of neuronal cells in the cortical microcircuit.SIGNIFICANCE STATEMENT No matter how we probe the brain, we find correlated neuronal activity over a variety of spatial and temporal scales. For the cerebral cortex, significant evidence has accumulated on trial-to-trial covariability in synaptic inputs activation, subthreshold membrane potential fluctuations, and output spike trains. Although we do not yet fully understand their origin and whether they are detrimental or beneficial for information processing, we believe that clarifying how correlations emerge is pivotal for understanding large-scale neuronal network dynamics and computation. Here, we report quantitative differences between excitatory and inhibitory cells, as they relay input correlations into output correlations. We explain this heterogeneity by simple biophysical models and provide the most experimentally validated test of a theory for the emergence of correlations.


Assuntos
Interneurônios/fisiologia , Modelos Neurológicos , Neocórtex/fisiologia , Células Piramidais/fisiologia , Animais , Feminino , Técnicas In Vitro , Masculino , Ratos
7.
Semin Cell Dev Biol ; 76: 86-100, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28918121

RESUMO

This review will focus on early aspects of cortical interneurons (cIN) development from specification to migration and final positioning in the human cerebral cortex. These mechanisms have been largely studied in the mouse model, which provides unique possibilities of genetic analysis, essential to dissect the molecular and cellular events involved in cortical development. An important goal here is to discuss the conservation and the potential divergence of these mechanisms, with a particular interest for the situation in the human embryo. We will thus cover recent works, but also revisit older studies in the light of recent data to better understand the developmental mechanisms underlying cIN differentiation in human. Because cIN are implicated in severe developmental disorders, understanding the molecular and cellular mechanisms controlling their differentiation might clarify some causes and potential therapeutic approaches to these important clinical conditions.


Assuntos
Córtex Cerebral/embriologia , Interneurônios/fisiologia , Diferenciação Celular , Humanos
8.
Development ; 144(21): 3867-3878, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29089360

RESUMO

Cortical interneurons are a diverse group of neurons that project locally and are crucial for regulating information processing and flow throughout the cortex. Recent studies in mice have advanced our understanding of how these neurons are specified, migrate and mature. Here, we evaluate new findings that provide insights into the development of cortical interneurons and that shed light on when their fate is determined, on the influence that regional domains have on their development, and on the role that key transcription factors and other crucial regulatory genes play in these events. We focus on cortical interneurons that are derived from the medial ganglionic eminence, as most studies have examined this interneuron population. We also assess how these data inform our understanding of neuropsychiatric disease and discuss the potential role of cortical interneurons in cell-based therapies.


Assuntos
Córtex Cerebral/citologia , Interneurônios/citologia , Animais , Linhagem da Célula , Modelos Biológicos , Fatores de Tempo , Transcrição Gênica
9.
FASEB J ; 33(12): 14204-14220, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31665922

RESUMO

Polymorphic variants of the gene encoding for metabotropic glutamate receptor 3 (mGlu3) are linked to schizophrenia. Because abnormalities of cortical GABAergic interneurons lie at the core of the pathophysiology of schizophrenia, we examined whether mGlu3 receptors influence the developmental trajectory of cortical GABAergic transmission in the postnatal life. mGlu3-/- mice showed robust changes in the expression of interneuron-related genes in the prefrontal cortex (PFC), including large reductions in the expression of parvalbumin (PV) and the GluN1 subunit of NMDA receptors. The number of cortical cells enwrapped by perineuronal nets was increased in mGlu3-/- mice, suggesting that mGlu3 receptors shape the temporal window of plasticity of PV+ interneurons. Electrophysiological measurements of GABAA receptor-mediated responses revealed a more depolarized reversal potential of GABA currents in the somata of PFC pyramidal neurons in mGlu3-/- mice at postnatal d 9 associated with a reduced expression of the K+/Cl- symporter. Finally, adult mGlu3-/- mice showed lower power in electroencephalographic rhythms at 1-45 Hz in quiet wakefulness as compared with their wild-type counterparts. These findings suggest that mGlu3 receptors have a strong impact on the development of cortical GABAergic transmission and cortical neural synchronization mechanisms corroborating the concept that genetic variants of mGlu3 receptors may predispose to psychiatric disorders.-Imbriglio, T., Verhaeghe, R., Martinello, K., Pascarelli, M. T., Chece, G., Bucci, D., Notartomaso, S., Quattromani, M., Mascio, G., Scalabrì, F., Simeone, A., Maccari, S., Del Percio, C., Wieloch, T., Fucile, S., Babiloni, C., Battaglia, G., Limatola, C., Nicoletti, F., Cannella, M. Developmental abnormalities in cortical GABAergic system in mice lacking mGlu3 metabotropic glutamate receptors.


Assuntos
Córtex Cerebral/anormalidades , Embrião de Mamíferos/anormalidades , Neurônios GABAérgicos/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Biomarcadores , Córtex Cerebral/metabolismo , Feminino , Regulação da Expressão Gênica , Genes Homeobox , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro , Receptores de Glutamato Metabotrópico/genética
10.
Cereb Cortex ; 29(4): 1644-1658, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29912395

RESUMO

The delicate balance of excitation and inhibition is crucial for proper function of the cerebral cortex, relying on the accurate number and subtype composition of inhibitory gamma-aminobutyric (GABA)-expressing interneurons. Various intrinsic and extrinsic factors precisely orchestrate their multifaceted development including the long-range migration from the basal telencephalon to cortical targets as well as interneuron survival throughout the developmental period. Particularly expressed guidance receptors were described to channel the migration of cortical interneurons deriving from the medial ganglionic eminence (MGE) and the preoptic area (POA) along distinct routes. Hence, unveiling the regulatory genetic networks controlling subtype-specific gene expression profiles is key to understand interneuron-specific developmental programs and to reveal causes for associated disorders. In contrast to MGE-derived interneurons, little is known about the transcriptional networks in interneurons born in the POA. Here, we provide first evidence for the LIM-homeobox transcription factor LHX1 as a crucial key player in the post-mitotic development of POA-derived cortical interneurons. By transcriptional regulation of related genes, LHX1 modulates their survival as well as the subtype-specific expression of guidance receptors of the Eph/ephrin family, thereby affecting directional migration and layer distribution in the adult cortex.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Interneurônios/fisiologia , Proteínas com Homeodomínio LIM/fisiologia , Área Pré-Óptica/crescimento & desenvolvimento , Fatores de Transcrição/fisiologia , Animais , Movimento Celular , Sobrevivência Celular , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Efrina-B3/genética , Efrina-B3/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/citologia , Interneurônios/metabolismo , Proteínas com Homeodomínio LIM/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Área Pré-Óptica/citologia , Área Pré-Óptica/metabolismo , Receptor EphA4/genética , Receptor EphA4/fisiologia , Fatores de Transcrição/genética
11.
Proc Natl Acad Sci U S A ; 114(52): E11180-E11189, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29229852

RESUMO

Cortical interneurons (cINs) modulate excitatory neuronal activity by providing local inhibition. During fetal development, several cIN subtypes derive from the medial ganglionic eminence (MGE), a transient ventral telencephalic structure. While altered cIN development contributes to neurodevelopmental disorders, the inaccessibility of human fetal brain tissue during development has hampered efforts to define molecular networks controlling this process. Here, we modified protocols for directed differentiation of human embryonic stem cells, obtaining efficient, accelerated production of MGE-like progenitors and MGE-derived cIN subtypes with the expected electrophysiological properties. We defined transcriptome changes accompanying this process and integrated these data with direct transcriptional targets of NKX2-1, a transcription factor controlling MGE specification. This analysis defined NKX2-1-associated genes with enriched expression during MGE specification and cIN differentiation, including known and previously unreported transcription factor targets with likely roles in MGE specification, and other target classes regulating cIN migration and function. NKX2-1-associated peaks were enriched for consensus binding motifs for NKX2-1, LHX, and SOX transcription factors, suggesting roles in coregulating MGE gene expression. Among the NKX2-1 direct target genes with cIN-enriched expression was CHD2, which encodes a chromatin remodeling protein mutated to cause human epilepsies. Accordingly, CHD2 deficiency impaired cIN specification and altered later electrophysiological function, while CHD2 coassociated with NKX2-1 at cis-regulatory elements and was required for their transactivation by NKX2-1 in MGE-like progenitors. This analysis identified several aspects of gene-regulatory networks underlying human MGE specification and suggested mechanisms by which NKX2-1 acts with chromatin remodeling activities to regulate gene expression programs underlying cIN development.


Assuntos
Diferenciação Celular , Córtex Cerebral/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Interneurônios/metabolismo , Linhagem Celular , Córtex Cerebral/citologia , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias Humanas/citologia , Humanos , Interneurônios/citologia , Fator Nuclear 1 de Tireoide/genética , Fator Nuclear 1 de Tireoide/metabolismo
12.
Int J Mol Sci ; 20(19)2019 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-31581684

RESUMO

Progress in iPSC-based cellular systems provides new insights into human brain development and early neurodevelopmental deviations in psychiatric disorders. Among these, studies on schizophrenia (SCZ) take a prominent role owing to its high heritability and multifarious evidence that it evolves from a genetically induced vulnerability in brain development. Recent iPSC studies on patients with SCZ indicate that functional impairments of neural progenitor cells (NPCs) in monolayer culture extend to brain organoids by disrupting neocorticogenesis in an in vitro model. In addition, the formation of hippocampal circuit-like structures in vitro is impaired in patients with SCZ as is the case for glia development. Intriguingly, chimeric-mice experiments show altered oligodendrocyte and astrocyte development in vivo that highlights the importance of cell-cell interactions in the pathogenesis of early-onset SCZ. Likewise, cortical imbalances in excitatory-inhibitory signaling may result from a cell-autonomous defect in cortical interneuron (cIN) development. Overall, these findings indicate that genetic risk in SCZ impacts neocorticogenesis, hippocampal circuit formation, and the development of distinct glial and neuronal subtypes. In light of this remarkable progress, we discuss current limitations and further steps necessary to harvest the full potential of iPSC-based investigations on psychiatric disorders.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Transtornos Mentais/etiologia , Transtornos Mentais/metabolismo , Modelos Biológicos , Animais , Hipocampo/embriologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Células-Tronco Neurais/metabolismo , Neurogênese
13.
Development ; 142(7): 1267-78, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25804737

RESUMO

Medial ganglionic eminence (MGE)-derived GABAergic cortical interneurons (cINs) consist of multiple subtypes that are involved in many cortical functions. They also have a remarkable capacity to migrate, survive and integrate into cortical circuitry after transplantation into postnatal cortex. These features have engendered considerable interest in generating distinct subgroups of interneurons from pluripotent stem cells (PSCs) for the study of interneuron fate and function, and for the development of cell-based therapies. Although advances have been made, the capacity to generate highly enriched pools of subgroup fate-committed interneuron progenitors from PSCs has remained elusive. Previous studies have suggested that the two main MGE-derived interneuron subgroups--those expressing somatostatin (SST) and those expressing parvalbumin (PV)--are specified in the MGE from Nkx2.1-expressing progenitors at higher or lower levels of sonic hedgehog (Shh) signaling, respectively. To further explore the role of Shh and other factors in cIN fate determination, we generated a reporter line such that Nkx2.1-expressing progenitors express mCherry and postmitotic Lhx6-expressing MGE-derived interneurons express GFP. Manipulations of Shh exposure and time in culture influenced the subgroup fates of ESC-derived interneurons. Exposure to higher Shh levels, and collecting GFP-expressing precursors at 12 days in culture, resulted in the strongest enrichment for SST interneurons over those expressing PV, whereas the strongest enrichment for PV interneurons was produced by lower Shh and by collecting mCherry-expressing cells after 17 days in culture. These findings confirm that fate determination of cIN subgroups is crucially influenced by Shh signaling, and provide a system for the further study of interneuron fate and function.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias/citologia , Proteínas Hedgehog/metabolismo , Interneurônios/metabolismo , Parvalbuminas/metabolismo , Transdução de Sinais , Somatostatina/metabolismo , Potenciais de Ação , Animais , Padronização Corporal , Linhagem Celular , Separação Celular , Córtex Cerebral/citologia , Células-Tronco Embrionárias/metabolismo , Neurônios GABAérgicos/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Eminência Mediana/citologia , Camundongos , Mitose , Transplante de Células-Tronco , Telencéfalo/embriologia , Telencéfalo/metabolismo , Fatores de Tempo
14.
J Physiol ; 595(18): 6203-6217, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28513860

RESUMO

KEY POINTS: It has long been known that the somatosensory cortex gates sensory inputs from the contralateral side of the body. Here, we examined the contribution of the ipsilateral somatosensory cortex (iS1) to sensory gating during index finger voluntary activity. The amplitude of the P25/N33, but not other somatosensory evoked potential (SSEP) components, was reduced during voluntary activity compared with rest. Interhemispheric inhibition between S1s and intracortical inhibition in the S1 modulated the amplitude of the P25/N33. Note that changes in interhemispheric inhibition between S1s correlated with changes in cortical circuits in the ipsilateral motor cortex. Our findings suggest that cortical circuits, probably from somatosensory and motor cortex, contribute to sensory gating in the iS1 during voluntary activity in humans. ABSTRACT: An important principle in the organization of the somatosensory cortex is that it processes afferent information from the contralateral side of the body. The role of the ipsilateral somatosensory cortex (iS1) in sensory gating in humans remains largely unknown. Using electroencephalographic (EEG) recordings over the iS1 and electrical stimulation of the ulnar nerve at the wrist, we examined somatosensory evoked potentials (SSEPs; P14/N20, N20/P25 and P25/N33 components) and paired-pulse SSEPs between S1s (interhemispheric inhibition) and within (intracortical inhibition) the iS1 at rest and during tonic index finger voluntary activity. We found that the amplitude of the P25/N33, but not other SSEP components, was reduced during voluntary activity compared with rest. Interhemispheric inhibition increased the amplitude of the P25/N33 and intracortical inhibition reduced the amplitude of the P25/N33, suggesting a cortical origin for this effect. The P25/N33 receives inputs from the motor cortex, so we also examined the contribution of distinct sets of cortical interneurons by testing the effect of ulnar nerve stimulation on motor-evoked potentials (MEPs) elicited by transcranial magnetic stimulation over the ipsilateral motor cortex with the coil in the posterior-anterior (PA) and anterior-posterior (AP) orientation. Afferent input attenuated PA, but not AP, MEPs during voluntary activity compared with rest. Notably, changes in interhemispheric inhibition correlated with changes in PA MEPs. Our novel findings suggest that interhemispheric projections between S1s and intracortical circuits, probably from somatosensory and motor cortex, contribute to sensory gating in the iS1 during voluntary activity in humans.


Assuntos
Córtex Motor/fisiologia , Movimento , Filtro Sensorial , Córtex Somatossensorial/fisiologia , Adulto , Potencial Evocado Motor , Potenciais Somatossensoriais Evocados , Feminino , Dedos/inervação , Dedos/fisiologia , Lateralidade Funcional , Humanos , Masculino , Inibição Neural
15.
Development ; 141(2): 460-71, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24381199

RESUMO

Inhibitory interneurons control the flow of information and synchronization in the cerebral cortex at the circuit level. During embryonic development, multiple subtypes of cortical interneurons are generated in different regions of the ventral telencephalon, such as the medial and caudal ganglionic eminence (MGE and CGE), as well as the preoptic area (POA). These neurons then migrate over long distances towards their cortical target areas. Diverse families of diffusible and cell-bound signaling molecules, including the Eph/ephrin system, regulate and orchestrate interneuron migration. Ephrin A3 and A5, for instance, are expressed at the borders of the pathway of MGE-derived interneurons and prevent these cells from entering inappropriate regions via EphA4 forward signaling. We found that MGE-derived interneurons, in addition to EphA4, also express ephrin A and B ligands, suggesting Eph/ephrin forward and reverse signaling in the same cell. In vitro and in vivo approaches showed that EphA4-induced reverse signaling in MGE-derived interneurons promotes their migration and that this effect is mediated by ephrin A2 ligands. In EphA4 mutant mice, as well as after ephrin A2 knockdown using in utero electroporation, we found delayed interneuron migration at embryonic stages. Thus, besides functions in guiding MGE-derived interneurons to the cortex through forward signaling, here we describe a novel role of the ephrins in driving these neurons to their target via reverse signaling.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Efrina-A2/metabolismo , Interneurônios/fisiologia , Receptor EphA4/metabolismo , Telencéfalo/embriologia , Telencéfalo/metabolismo , Animais , Movimento Celular/fisiologia , Córtex Cerebral/citologia , Efrina-A2/antagonistas & inibidores , Efrina-A2/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor EphA4/genética , Transdução de Sinais , Telencéfalo/citologia
16.
Cereb Cortex ; 26(8): 3655-3668, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27288316

RESUMO

In the neocortex, inhibitory interneurons of the same subtype are electrically coupled with each other via dendritic gap junctions (GJs). The impact of multiple GJs on the biophysical properties of interneurons and thus on their input processing is unclear. The present experimentally based theoretical study examined GJs in L2/3 large basket cells (L2/3 LBCs) with 3 goals in mind: (1) To evaluate the errors due to GJs in estimating the cable properties of individual L2/3 LBCs and suggest ways to correct these errors when modeling these cells and the networks they form; (2) to bracket the GJ conductance value (0.05-0.25 nS) and membrane resistivity (10 000-40 000 Ω cm(2)) of L2/3 LBCs; these estimates are tightly constrained by in vitro input resistance (131 ± 18.5 MΩ) and the coupling coefficient (1-3.5%) of these cells; and (3) to explore the functional implications of GJs, and show that GJs: (i) dynamically modulate the effective time window for synaptic integration; (ii) improve the axon's capability to encode rapid changes in synaptic inputs; and (iii) reduce the orientation selectivity, linearity index, and phase difference of L2/3 LBCs. Our study provides new insights into the role of GJs and calls for caution when using in vitro measurements for modeling electrically coupled neuronal networks.


Assuntos
Junções Comunicantes/fisiologia , Interneurônios/fisiologia , Neocórtex/fisiologia , Sinapses/fisiologia , Animais , Axônios/fisiologia , Simulação por Computador , Dendritos/fisiologia , Potenciais da Membrana/fisiologia , Modelos Neurológicos , Ratos
17.
Cereb Cortex ; 26(1): 131-43, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25146370

RESUMO

Impaired sonic hedgehog (Shh) signaling is involved in the pathology of cortical formation found in neuropsychiatric disorders. However, its role in the specification of human cortical progenitors is not known. Here, we report that Shh is expressed in the human developing cortex at mid-gestation by radial glia cells (RGCs) and cortical neurons. We used RGC cultures, established from the dorsal (cortical) telencephalon of human brain at mid-gestation to study the effect of Shh signaling. Cortical RGCs in vitro maintained their regional characteristics, expressed components of Shh signaling, and differentiated into Nkx2.1, Lhx6, and calretinin-positive (CalR(+)) cells, potential cortical interneuron progenitors. Treatment with exogenous Shh increased the pool of Nkx2.1(+) progenitors, decreased Lhx6 expression, and suppressed the generation of CalR(+) cells. The blockade of endogenous Shh signaling increased the number of CalR(+) cells, but did not affect Nkx2.1 expression, implying the existence of parallel Shh-independent pathways for cortical Nkx2.1 regulation. These results support the idea that, during human brain development, Shh plays an important role in the specification of cortical progenitors. Since direct functional studies in humans are limited, the in vitro system that we established here could be of great interest for modeling the development of human cortical progenitors.


Assuntos
Cálcio/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Proteínas Hedgehog/metabolismo , Interneurônios/citologia , Telencéfalo/citologia , Adolescente , Adulto , Córtex Cerebral/citologia , Humanos , Proteínas do Tecido Nervoso/metabolismo , Adulto Jovem
18.
Cereb Cortex ; 25(7): 1842-57, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24451661

RESUMO

Complex and precisely orchestrated genetic programs contribute to the generation, migration, and maturation of cortical GABAergic interneurons (cIN). Yet, little is known about the signals that mediate the rapid alterations in gene expression that are required for cINs to transit through a series of developmental steps leading to their mature properties in the cortex. Here, we investigated the function of post-transcriptional regulation of gene expression by microRNAs on the development of cIN precursors. We find that conditional removal of the RNAseIII enzyme Dicer reduces the number of cINs in the adult mouse. Dicer is further necessary for the morphological and molecular maturation of cINs. Loss of mature miRNAs affects cINs development by impairing migration and differentiation of this cell type, while leaving proliferation of progenitors unperturbed. These developmental defects closely matched the abnormal expression of molecules involved in apoptosis and neuronal specification. In addition, we identified several miRNAs that are selectively upregulated in the postmitotic cINs, consistent with a role of miRNAs in the post-transcriptional control of the differentiation and apoptotic programs essential for cIN maturation. Thus, our results indicate that cIN progenitors require Dicer-dependent mechanisms to fine-tune the migration and maturation of cINs.


Assuntos
Sobrevivência Celular/fisiologia , Córtex Cerebral/fisiologia , RNA Helicases DEAD-box/metabolismo , Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , MicroRNAs/metabolismo , Ribonuclease III/metabolismo , Animais , Apoptose/fisiologia , Contagem de Células , Proliferação de Células/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , RNA Helicases DEAD-box/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/fisiologia , Proteínas Nucleares/metabolismo , Ribonuclease III/genética , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/metabolismo
19.
Cereb Cortex ; 25(12): 4854-68, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26420784

RESUMO

Neocortical GABAergic interneurons have a profound impact on cortical circuitry and its information processing capacity. Distinct subgroups of inhibitory interneurons can be distinguished by molecular markers, such as parvalbumin, somatostatin, and vasoactive intestinal polypeptide (VIP). Among these, VIP-expressing interneurons sparked a substantial interest since these neurons seem to operate disinhibitory circuit motifs found in all major neocortical areas. Several of these recent studies used transgenic Vip-ires-cre mice to specifically target the population of VIP-expressing interneurons. This makes it necessary to elucidate in detail the sensitivity and specificity of Cre expression for VIP neurons in these animals. Thus, we quantitatively compared endogenous tdTomato with Vip fluorescence in situ hybridization and αVIP immunohistochemistry in the barrel cortex of VIPcre/tdTomato mice in a layer-specific manner. We show that VIPcre/tdTomato mice are highly sensitive and specific for the entire population of VIP-expressing neurons. In the barrel cortex, approximately 13% of all GABAergic neurons are VIP expressing. Most VIP neurons are found in layer II/III (∼60%), whereas approximately 40% are found in the other layers of the barrel cortex. Layer II/III VIP neurons are significantly different from VIP neurons in layers IV-VI in several morphological and membrane properties, which suggest layer-dependent differences in functionality.


Assuntos
Interneurônios/citologia , Interneurônios/metabolismo , Interneurônios/fisiologia , Córtex Somatossensorial/citologia , Córtex Somatossensorial/metabolismo , Córtex Somatossensorial/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Potenciais de Ação , Animais , Axônios , Dendritos , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/fisiologia , Imuno-Histoquímica/métodos , Hibridização in Situ Fluorescente/métodos , Camundongos , Camundongos Transgênicos , Parvalbuminas/metabolismo , RNA Mensageiro/metabolismo , Somatostatina/metabolismo
20.
J Neurophysiol ; 114(1): 624-37, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25972585

RESUMO

Precise spike synchrony has been widely reported in the central nervous system, but its functional role in encoding, processing, and transmitting information is yet unresolved. Of particular interest is firing synchrony between inhibitory cortical interneurons, thought to drive various cortical rhythms such as gamma oscillations, the hallmark of cognitive states. Precise synchrony can arise between two interneurons connected electrically, through gap junctions, chemically, through fast inhibitory synapses, or dually, through both types of connections, but the properties of synchrony generated by these different modes of connectivity have never been compared in the same data set. In the present study we recorded in vitro from 152 homotypic pairs of two major subtypes of mouse neocortical interneurons: parvalbumin-containing, fast-spiking (FS) interneurons and somatostatin-containing (SOM) interneurons. We tested firing synchrony when the two neurons were driven to fire by long, depolarizing current steps and used a novel synchrony index to quantify the strength of synchrony, its temporal precision, and its dependence on firing rate. We found that SOM-SOM synchrony, driven solely by electrical coupling, was less precise than FS-FS synchrony, driven by inhibitory or dual coupling. Unlike SOM-SOM synchrony, FS-FS synchrony was strongly firing rate dependent and was not evident at the prototypical 40-Hz gamma frequency. Computer simulations reproduced these differences in synchrony without assuming any differences in intrinsic properties, suggesting that the mode of coupling is more important than the interneuron subtype. Our results provide novel insights into the mechanisms and properties of interneuron synchrony and point out important caveats in current models of cortical oscillations.


Assuntos
Potenciais de Ação/fisiologia , Interneurônios/fisiologia , Córtex Somatossensorial/fisiologia , Sinapses/fisiologia , Animais , Simulação por Computador , Camundongos Transgênicos , Modelos Neurológicos , Parvalbuminas/metabolismo , Técnicas de Patch-Clamp , Periodicidade , Córtex Somatossensorial/crescimento & desenvolvimento , Somatostatina/metabolismo , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA