Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Cell ; 186(1): 209-229.e26, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36608654

RESUMO

Transcription factors (TFs) regulate gene programs, thereby controlling diverse cellular processes and cell states. To comprehensively understand TFs and the programs they control, we created a barcoded library of all annotated human TF splice isoforms (>3,500) and applied it to build a TF Atlas charting expression profiles of human embryonic stem cells (hESCs) overexpressing each TF at single-cell resolution. We mapped TF-induced expression profiles to reference cell types and validated candidate TFs for generation of diverse cell types, spanning all three germ layers and trophoblasts. Targeted screens with subsets of the library allowed us to create a tailored cellular disease model and integrate mRNA expression and chromatin accessibility data to identify downstream regulators. Finally, we characterized the effects of combinatorial TF overexpression by developing and validating a strategy for predicting combinations of TFs that produce target expression profiles matching reference cell types to accelerate cellular engineering efforts.


Assuntos
Diferenciação Celular , Fatores de Transcrição , Humanos , Cromatina , Regulação da Expressão Gênica , Células-Tronco Embrionárias Humanas/metabolismo , Fatores de Transcrição/metabolismo , Atlas como Assunto
3.
Nature ; 583(7818): 819-824, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32699411

RESUMO

The thalamic reticular nucleus (TRN), the major source of thalamic inhibition, regulates thalamocortical interactions that are critical for sensory processing, attention and cognition1-5. TRN dysfunction has been linked to sensory abnormality, attention deficit and sleep disturbance across multiple neurodevelopmental disorders6-9. However, little is known about the organizational principles that underlie its divergent functions. Here we performed an integrative study linking single-cell molecular and electrophysiological features of the mouse TRN to connectivity and systems-level function. We found that cellular heterogeneity in the TRN is characterized by a transcriptomic gradient of two negatively correlated gene-expression profiles, each containing hundreds of genes. Neurons in the extremes of this transcriptomic gradient express mutually exclusive markers, exhibit core or shell-like anatomical structure and have distinct electrophysiological properties. The two TRN subpopulations make differential connections with the functionally distinct first-order and higher-order thalamic nuclei to form molecularly defined TRN-thalamus subnetworks. Selective perturbation of the two subnetworks in vivo revealed their differential role in regulating sleep. In sum, our study provides a comprehensive atlas of TRN neurons at single-cell resolution and links molecularly defined subnetworks to the functional organization of thalamocortical circuits.


Assuntos
Redes Reguladoras de Genes , Núcleos Talâmicos/citologia , Núcleos Talâmicos/metabolismo , Animais , Análise por Conglomerados , Feminino , Perfilação da Expressão Gênica , Hibridização in Situ Fluorescente , Metaloendopeptidases/metabolismo , Camundongos , Vias Neurais , Neurônios/metabolismo , Osteopontina/metabolismo , Técnicas de Patch-Clamp , RNA-Seq , Análise de Célula Única , Sono/genética , Sono/fisiologia , Núcleos Talâmicos/fisiologia , Transcriptoma
4.
Nature ; 574(7778): 413-417, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31597963

RESUMO

A longstanding goal in neuroscience has been to image membrane voltage across a population of individual neurons in an awake, behaving mammal. Here we describe a genetically encoded fluorescent voltage indicator, SomArchon, which exhibits millisecond response times and is compatible with optogenetic control, and which increases the sensitivity, signal-to-noise ratio, and number of neurons observable several-fold over previously published fully genetically encoded reagents1-8. Under conventional one-photon microscopy, SomArchon enables the routine population analysis of around 13 neurons at once, in multiple brain regions (cortex, hippocampus, and striatum) of head-fixed, awake, behaving mice. Using SomArchon, we detected both positive and negative responses of striatal neurons during movement, as previously reported by electrophysiology but not easily detected using modern calcium imaging techniques9-11, highlighting the power of voltage imaging to reveal bidirectional modulation. We also examined how spikes relate to the subthreshold theta oscillations of individual hippocampal neurons, with SomArchon showing that the spikes of individual neurons are more phase-locked to their own subthreshold theta oscillations than to local field potential theta oscillations. Thus, SomArchon reports both spikes and subthreshold voltage dynamics in awake, behaving mice.


Assuntos
Biomarcadores Ambientais , Hipocampo/citologia , Neurônios/fisiologia , Imagem Óptica/métodos , Vigília/fisiologia , Potenciais de Ação/fisiologia , Animais , Biomarcadores Ambientais/genética , Hipocampo/diagnóstico por imagem , Camundongos , Optogenética
5.
Nature ; 530(7591): 481-4, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26886798

RESUMO

Because autism spectrum disorders are neurodevelopmental disorders and patients typically display symptoms before the age of three, one of the key questions in autism research is whether the pathology is reversible in adults. Here we investigate the developmental requirement of Shank3 in mice, a prominent monogenic autism gene that is estimated to contribute to approximately 1% of all autism spectrum disorder cases. SHANK3 is a postsynaptic scaffold protein that regulates synaptic development, function and plasticity by orchestrating the assembly of postsynaptic density macromolecular signalling complex. Disruptions of the Shank3 gene in mouse models have resulted in synaptic defects and autistic-like behaviours including anxiety, social interaction deficits, and repetitive behaviour. We generated a novel Shank3 conditional knock-in mouse model, and show that re-expression of the Shank3 gene in adult mice led to improvements in synaptic protein composition, spine density and neural function in the striatum. We also provide behavioural evidence that certain behavioural abnormalities including social interaction deficit and repetitive grooming behaviour could be rescued, while anxiety and motor coordination deficit could not be recovered in adulthood. Together, these results reveal the profound effect of post-developmental activation of Shank3 expression on neural function, and demonstrate a certain degree of continued plasticity in the adult diseased brain.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Fatores Etários , Envelhecimento/genética , Animais , Ansiedade/genética , Transtorno do Espectro Autista/psicologia , Espinhas Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Asseio Animal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos , Transtornos das Habilidades Motoras/genética , Transtornos das Habilidades Motoras/fisiopatologia , Neostriado/citologia , Neostriado/metabolismo , Neostriado/patologia , Plasticidade Neuronal/genética , Densidade Pós-Sináptica/química , Densidade Pós-Sináptica/metabolismo , Desempenho Psicomotor , Comportamento Social
6.
Dev Biol ; 468(1-2): 93-100, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32976839

RESUMO

Fragile X mental retardation 1 (FMR1) encodes the RNA binding protein FMRP. Loss of FMRP drives Fragile X syndrome (FXS), the leading inherited cause of intellectual disability and a leading monogenic cause of autism. While cortical hyperexcitability is a hallmark of FXS, the reported phenotypes and underlying mechanisms, including alterations in synaptic transmission and ion channel properties, are heterogeneous and at times contradictory. Here, we report the generation of new isogenic FMR1y/+ and FMR1y/- human pluripotent stem cell (hPSC) lines using CRISPR-Cas9 to facilitate the study of how complete FMRP loss, independent of genetic background, drives molecular and cellular alterations relevant for FXS. After differentiating these stem cell tools into excitatory neurons, we systematically assessed the impact of FMRP loss on intrinsic membrane and synaptic properties over time. Using whole-cell patch clamp analyses, we found that FMR1y/- neurons overall showed an increased intrinsic membrane excitability compared to age-matched FMR1y/+ controls, with no discernable alternations in synaptic transmission. Surprisingly, longitudinal analyses of cell intrinsic defects revealed that a majority of significant changes emerged early following in vitro differentiation and some were not stable over time. Collectively, this study provides a new isogenic hPSC model which can be further leveraged by the scientific community to investigate basic mechanisms of FMR1 gene function relevant for FXS. Moreover, our results suggest that precocious changes in the intrinsic membrane properties during early developmental could be a critical cellular pathology ultimately contributing to cortical hyperexcitability in FXS.


Assuntos
Diferenciação Celular , Membrana Celular/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Células-Tronco Embrionárias Humanas/metabolismo , Potenciais da Membrana , Neurônios/metabolismo , Transmissão Sináptica , Linhagem Celular , Membrana Celular/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Humanos
7.
Proc Natl Acad Sci U S A ; 113(46): E7287-E7296, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27803317

RESUMO

Rett syndrome (RTT) arises from loss-of-function mutations in methyl-CpG binding protein 2 gene (Mecp2), but fundamental aspects of its physiological mechanisms are unresolved. Here, by whole-cell recording of synaptic responses in MeCP2 mutant mice in vivo, we show that visually driven excitatory and inhibitory conductances are both reduced in cortical pyramidal neurons. The excitation-to-inhibition (E/I) ratio is increased in amplitude and prolonged in time course. These changes predict circuit-wide reductions in response reliability and selectivity of pyramidal neurons to visual stimuli, as confirmed by two-photon imaging. Targeted recordings reveal that parvalbumin-expressing (PV+) interneurons in mutant mice have reduced responses. PV-specific MeCP2 deletion alone recapitulates effects of global MeCP2 deletion on cortical circuits, including reduced pyramidal neuron responses and reduced response reliability and selectivity. Furthermore, MeCP2 mutant mice show reduced expression of the cation-chloride cotransporter KCC2 (K+/Cl- exporter) and a reduced KCC2/NKCC1 (Na+/K+/Cl- importer) ratio. Perforated patch recordings demonstrate that the reversal potential for GABA is more depolarized in mutant mice, but is restored by application of the NKCC1 inhibitor bumetanide. Treatment with recombinant human insulin-like growth factor-1 restores responses of PV+ and pyramidal neurons and increases KCC2 expression to normalize the KCC2/NKCC1 ratio. Thus, loss of MeCP2 in the brain alters both excitation and inhibition in brain circuits via multiple mechanisms. Loss of MeCP2 from a specific interneuron subtype contributes crucially to the cell-specific and circuit-wide deficits of RTT. The joint restoration of inhibition and excitation in cortical circuits is pivotal for functionally correcting the disorder.


Assuntos
Córtex Cerebral/fisiologia , Interneurônios/fisiologia , Células Piramidais/fisiologia , Síndrome de Rett/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Fator de Crescimento Insulin-Like I/farmacologia , Interneurônios/efeitos dos fármacos , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parvalbuminas , Células Piramidais/efeitos dos fármacos , Proteínas Recombinantes
8.
J Neurosci ; 36(7): 2247-60, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26888934

RESUMO

Intellectual disability is a common neurodevelopmental disorder characterized by impaired intellectual and adaptive functioning. Both environmental insults and genetic defects contribute to the etiology of intellectual disability. Copy number variations of SORBS2 have been linked to intellectual disability. However, the neurobiological function of SORBS2 in the brain is unknown. The SORBS2 gene encodes ArgBP2 (Arg/c-Abl kinase binding protein 2) protein in non-neuronal tissues and is alternatively spliced in the brain to encode nArgBP2 protein. We found nArgBP2 colocalized with F-actin at dendritic spines and growth cones in cultured hippocampal neurons. In the mouse brain, nArgBP2 was highly expressed in the cortex, amygdala, and hippocampus, and enriched in the outer one-third of the molecular layer in dentate gyrus. Genetic deletion of Sorbs2 in mice led to reduced dendritic complexity and decreased frequency of AMPAR-miniature spontaneous EPSCs in dentate gyrus granule cells. Behavioral characterization revealed that Sorbs2 deletion led to a reduced acoustic startle response, and defective long-term object recognition memory and contextual fear memory. Together, our findings demonstrate, for the first time, an important role for nArgBP2 in neuronal dendritic development and excitatory synaptic transmission, which may thus inform exploration of neurobiological basis of SORBS2 deficiency in intellectual disability. SIGNIFICANCE STATEMENT: Copy number variations of the SORBS2 gene are linked to intellectual disability, but the neurobiological mechanisms are unknown. We found that nArgBP2, the only neuronal isoform encoded by SORBS2, colocalizes with F-actin at neuronal dendritic growth cones and spines. nArgBP2 is highly expressed in the cortex, amygdala, and dentate gyrus in the mouse brain. Genetic deletion of Sorbs2 in mice leads to impaired dendritic complexity and reduced excitatory synaptic transmission in dentate gyrus granule cells, accompanied by behavioral deficits in acoustic startle response and long-term memory. This is the first study of Sorbs2 function in the brain, and our findings may facilitate the study of neurobiological mechanisms underlying SORBS2 deficiency in the development of intellectual disability.


Assuntos
Encéfalo/crescimento & desenvolvimento , Dendritos/patologia , Memória , Proteínas dos Microfilamentos/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Comportamento Animal , DNA/genética , Espinhas Dendríticas/patologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Cones de Crescimento/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Memória de Longo Prazo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Proteínas de Ligação a RNA , Reconhecimento Psicológico , Reflexo de Sobressalto/genética
9.
Nature ; 472(7344): 437-42, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21423165

RESUMO

Autism spectrum disorders (ASDs) comprise a range of disorders that share a core of neurobehavioural deficits characterized by widespread abnormalities in social interactions, deficits in communication as well as restricted interests and repetitive behaviours. The neurological basis and circuitry mechanisms underlying these abnormal behaviours are poorly understood. SHANK3 is a postsynaptic protein, whose disruption at the genetic level is thought to be responsible for the development of 22q13 deletion syndrome (Phelan-McDermid syndrome) and other non-syndromic ASDs. Here we show that mice with Shank3 gene deletions exhibit self-injurious repetitive grooming and deficits in social interaction. Cellular, electrophysiological and biochemical analyses uncovered defects at striatal synapses and cortico-striatal circuits in Shank3 mutant mice. Our findings demonstrate a critical role for SHANK3 in the normal development of neuronal connectivity and establish causality between a disruption in the Shank3 gene and the genesis of autistic-like behaviours in mice.


Assuntos
Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Neostriado/fisiopatologia , Animais , Comportamento Compulsivo/genética , Feminino , Deleção de Genes , Asseio Animal , Masculino , Camundongos , Proteínas dos Microfilamentos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neostriado/patologia , Proteínas do Tecido Nervoso , Vias Neurais , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Comportamento Autodestrutivo/genética , Comportamento Autodestrutivo/fisiopatologia , Comportamento Social , Sinapses/metabolismo , Sinapses/patologia
10.
Neuron ; 112(3): 441-457.e6, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-37992714

RESUMO

Social isolation is a risk factor for multiple mood disorders. Specifically, social isolation can remodel the brain, causing behavioral abnormalities, including sociability impairments. Here, we investigated social behavior impairment in mice following chronic social isolation stress (CSIS) and conducted a screening of susceptible brain regions using functional readouts. CSIS enhanced synaptic inhibition in the anterior cingulate cortex (ACC), particularly at inhibitory synapses of cholecystokinin (CCK)-expressing interneurons. This enhanced synaptic inhibition in the ACC was characterized by CSIS-induced loss of presynaptic cannabinoid type-1 receptors (CB1Rs), resulting in excessive axonal calcium influx. Activation of CCK-expressing interneurons or conditional knockdown of CB1R expression in CCK-expressing interneurons specifically reproduced social impairment. In contrast, optogenetic activation of CB1R or administration of CB1R agonists restored sociability in CSIS mice. These results suggest that the CB1R may be an effective therapeutic target for preventing CSIS-induced social impairments by restoring synaptic inhibition in the ACC.


Assuntos
Canabinoides , Giro do Cíngulo , Animais , Masculino , Camundongos , Canabinoides/metabolismo , Canabinoides/farmacologia , Giro do Cíngulo/metabolismo , Interneurônios/fisiologia , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Isolamento Social , Sinapses/fisiologia
11.
Cell Rep Med ; 5(5): 101534, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38670100

RESUMO

Thalamocortical (TC) circuits are essential for sensory information processing. Clinical and preclinical studies of autism spectrum disorders (ASDs) have highlighted abnormal thalamic development and TC circuit dysfunction. However, mechanistic understanding of how TC dysfunction contributes to behavioral abnormalities in ASDs is limited. Here, our study on a Shank3 mouse model of ASD reveals TC neuron hyperexcitability with excessive burst firing and a temporal mismatch relationship with slow cortical rhythms during sleep. These TC electrophysiological alterations and the consequent sensory hypersensitivity and sleep fragmentation in Shank3 mutant mice are causally linked to HCN2 channelopathy. Restoring HCN2 function early in postnatal development via a viral approach or lamotrigine (LTG) ameliorates sensory and sleep problems. A retrospective case series also supports beneficial effects of LTG treatment on sensory behavior in ASD patients. Our study identifies a clinically relevant circuit mechanism and proposes a targeted molecular intervention for ASD-related behavioral impairments.


Assuntos
Transtorno do Espectro Autista , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Proteínas do Tecido Nervoso , Tálamo , Animais , Tálamo/metabolismo , Tálamo/patologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Camundongos , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Transtorno do Espectro Autista/patologia , Lamotrigina/farmacologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Canalopatias/genética , Canalopatias/metabolismo , Canalopatias/patologia , Humanos , Modelos Animais de Doenças , Masculino , Neurônios/metabolismo , Feminino , Camundongos Endogâmicos C57BL , Mutação/genética , Sono/fisiologia , Sono/efeitos dos fármacos , Sono/genética , Canais de Potássio
12.
J Biol Chem ; 286(25): 22456-68, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21540179

RESUMO

Collybistin promotes submembrane clustering of gephyrin and is essential for the postsynaptic localization of gephyrin and γ-aminobutyric acid type A (GABA(A)) receptors at GABAergic synapses in hippocampus and amygdala. Four collybistin isoforms are expressed in brain neurons; CB2 and CB3 differ in the C terminus and occur with and without the Src homology 3 (SH3) domain. We have found that in transfected hippocampal neurons, all collybistin isoforms (CB2(SH3+), CB2(SH3-), CB3(SH3+), and CB3(SH3-)) target to and concentrate at GABAergic postsynapses. Moreover, in non-transfected neurons, collybistin concentrates at GABAergic synapses. Hippocampal neurons co-transfected with CB2(SH3-) and gephyrin developed very large postsynaptic gephyrin and GABA(A) receptor clusters (superclusters). This effect was accompanied by a significant increase in the amplitude of miniature inhibitory postsynaptic currents. Co-transfection with CB2(SH3+) and gephyrin induced the formation of many (supernumerary) non-synaptic clusters. Transfection with gephyrin alone did not affect cluster number or size, but gephyrin potentiated the clustering effect of CB2(SH3-) or CB2(SH3+). Co-transfection with CB2(SH3-) or CB2(SH3+) and gephyrin did not affect the density of presynaptic GABAergic terminals contacting the transfected cells, indicating that collybistin is not synaptogenic. Nevertheless, the synaptic superclusters induced by CB2(SH3-) and gephyrin were accompanied by enlarged presynaptic GABAergic terminals. The enhanced clustering of gephyrin and GABA(A) receptors induced by collybistin isoforms was not accompanied by enhanced clustering of neuroligin 2. Moreover, during the development of GABAergic synapses, the clustering of gephyrin and GABA(A) receptors preceded the clustering of neuroligin 2. We propose a model in which the SH3- isoforms play a major role in the postsynaptic accumulation of GABA(A) receptors and in GABAergic synaptic strength.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Receptores de GABA-A/metabolismo , Sinapses/metabolismo , Animais , Proteínas de Transporte/metabolismo , Fatores de Troca do Nucleotídeo Guanina/química , Células HEK293 , Humanos , Potenciais Pós-Sinápticos Inibidores , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Fatores de Troca de Nucleotídeo Guanina Rho , Transfecção , Ácido gama-Aminobutírico/metabolismo , Domínios de Homologia de src
13.
Sci Adv ; 8(49): eade1136, 2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36475786

RESUMO

Ionic conductivity and membrane capacitance are two foundational parameters that govern neuron excitability. Conventional optogenetics has emerged as a powerful tool to temporarily manipulate membrane ionic conductivity in intact biological systems. However, no analogous method exists for precisely manipulating cell membrane capacitance to enable long-lasting modulation of neuronal excitability. Genetically targetable chemical assembly of conductive and insulating polymers can modulate cell membrane capacitance, but further development of this technique has been hindered by poor spatiotemporal control of the polymer deposition and cytotoxicity from the widely diffused peroxide. We address these issues by harnessing genetically targetable photosensitizer proteins to assemble electrically functional polymers in neurons with precise spatiotemporal control. Using whole-cell patch-clamp recordings, we demonstrate that this optogenetic polymerization can achieve stepwise modulation of both neuron membrane capacitance and intrinsic excitability. Furthermore, cytotoxicity can be limited by controlling light exposure, demonstrating a promising new method for precisely modulating cell excitability.

14.
J Neurosci ; 29(16): 5116-26, 2009 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-19386907

RESUMO

We previously reported greater GABAA receptor-mediated tonic currents in D2+ striatopallidal than D1+ striatonigral medium spiny neurons (MSNs) are mediated by alpha5-subunit-containing receptors. Here, we used whole-cell recordings in slices from bacterial artificial chromosome transgenic mice to investigate the link between subunit composition, phosphorylation, and dopamine receptor activation. Whole-cell recordings in slices from delta-subunit knock-out mice demonstrate that while MSNs in wild-type mice do express delta-subunit-containing receptors, this receptor subtype is not responsible for tonic conductance observed in the acute slice preparation. We assessed the contribution of the beta1- and beta3-subunits expressed in MSNs by their sensitivity to etomidate, an agonist selective for beta2- or beta3-subunit-containing GABAA receptors. Although etomidate produced substantial tonic current in D2+ neurons, there was no effect in D1+ neurons. However, with internal PKA application or dopamine modulation, D1+ neurons expressed tonic conductance and responded to etomidate application. Our results suggest that distinct phosphorylation of beta3-subunits may cause larger tonic current in D2+ striatopallidal MSNs, and proper intracellular conditions can reveal tonic current in D1+ cells.


Assuntos
Corpo Estriado/fisiologia , Dopamina/fisiologia , Condução Nervosa/fisiologia , Neurônios/fisiologia , Receptores de GABA-A/fisiologia , Transmissão Sináptica , Animais , Linhagem Celular , Corpo Estriado/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/fisiologia , Receptores de GABA-A/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética
15.
Mol Cell Neurosci ; 42(1): 45-55, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19463950

RESUMO

Neuroligins (NLGs) are postsynaptic cell adhesion molecules that are thought to function in synaptogenesis. To investigate the role of NLGs on synaptic transmission once the synapse is formed, we transfected neuroligin-2 (NLG-2) in cultured mouse cerebellar granule cells (CGCs), and recorded GABA(A) (gamma-aminobutyric acid) receptor mediated miniature postsynaptic currents (mIPSCs). NLG-2 transfected cells had mIPSCs with faster decay than matching GFP expressing controls at young culture ages (days in vitro, DIV7-8). Down-regulation of NLG-2 by the isoform specific shRNA-NLG-2 resulted in an opposite effect. We and others have shown that the switch of alpha subunits of GABA(A)Rs from alpha2/3 to alpha1 underlies developmental speeding of the IPSC decay in various CNS regions, including the cerebellum. To assess whether the reduced decay time of mIPSCs by NLG-2 is due to the recruitment of more alpha1 containing GABA(A)Rs at the synapses, we examined the prolongation of current decay by the Zolpidem, which has been shown to preferentially enhance the activity of alpha1 subunit-containing GABA channel. The application of Zolpidem resulted in a significantly greater prolongation kinetics of synaptic currents in NLG-2 over-expressing cells than control cells, suggesting that NLG-2 over-expression accelerates synapse maturation by promoting incorporation of the alpha1 subunit-containing GABA(A)Rs at postsynaptic sites in immature cells. In addition, the effect of NLG-2 on the speeding of decay time course of synaptic currents was abolished when we used CGC cultures from alpha1-/- mice. Lastly, to exclude the possibility that the fast decay of mIPSCs induced by NLG-2 could be also due to the impacts of NLG-2 on the GABA transient in synaptic cleft, we measured the sensitivity of mIPSCs to the fast-off competitive antagonists TPMPA. We found that TPMPA similarly inhibits mIPSCs in control and NLG-2 over-expressing CGCs both at young age (DIV8) and old age (DIV14) of cultures. However, we confirm our previous finding of a greater inhibition of mIPSCs in young (DIV8) than more mature (DIV14) cultures. Together, our results suggest that NLG-2 does not alter uniquantal GABA release, and the fast decay of mIPSC induced by NLG-2 is due to the differential expression of postsynaptic GABA(A) receptor subtypes. Taken all together, we propose that NLG-2 plays important functional role in inhibitory synapse development and maturation.


Assuntos
Cerebelo/citologia , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Sinapses/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Moléculas de Adesão Celular Neuronais , Células Cultivadas , Técnicas de Cocultura/métodos , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Glutamato Descarboxilase/genética , Proteínas de Fluorescência Verde/genética , Humanos , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp/métodos , Ácidos Fosfínicos/farmacologia , Subunidades Proteicas/genética , Piridinas/farmacologia , Interferência de RNA/fisiologia , Receptores de GABA-A/deficiência , Receptores de GABA-A/genética , Transfecção/métodos , Zolpidem
16.
Mol Cell Neurosci ; 42(4): 466-83, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19796685

RESUMO

Cell adhesion molecules have been implicated as key organizers of synaptic structures, but there is still a need to determine how these molecules facilitate neurotransmitter receptor recruitment to developing synapses. Here, we identify erythrocyte protein band 4.1-like 3 (protein 4.1B) as an intracellular effector molecule of Synaptic Cell Adhesion Molecule 1 (SynCAM1) that is sufficient to recruit NMDA-type receptors (NMDARs) to SynCAM1 adhesion sites in COS7 cells. Protein 4.1B in conjunction with SynCAM1 also increased the frequency of NMDAR-mediated mEPSCs and area of presynaptic contact in an HEK293 cell/ neuron co-culture assay. Studies in cultured hippocampal neurons reveal that manipulation of protein 4.1B expression levels specifically affects NMDAR-mediated activity and localization. Finally, further experimentation in COS7 cells show that SynCAM1 may also interact with protein 4.1N to specifically effect AMPA type receptor (AMPAR) recruitment. Thus, SynCAM1 may recruit both AMPARs and NMDARs by independent mechanisms during synapse formation.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Bioensaio/métodos , Biomarcadores/metabolismo , Células COS , Adesão Celular/fisiologia , Moléculas de Adesão Celular , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Chlorocebus aethiops , Técnicas de Cocultura , Hipocampo/citologia , Humanos , Imunoglobulinas , Proteínas de Membrana/genética , Proteínas dos Microfilamentos , Microesferas , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/fisiologia , Proteínas Supressoras de Tumor/genética
17.
Eur J Paediatr Neurol ; 24: 129-133, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31928904

RESUMO

The four voltage-gated sodium channels SCN1/2/3/8A have been associated with heterogeneous types of developmental disorders, each presenting with disease specific temporal and cell type specific gene expression. Using single-cell RNA sequencing transcriptomic data from humans and mice, we observe that SCN1A is predominantly expressed in inhibitory neurons. In contrast, SCN2/3/8A are profoundly expressed in excitatory neurons with SCN2/3A starting prenatally, followed by SCN1/8A neonatally. In contrast to previous observations from low resolution RNA screens, we observe that all four genes are expressed in both excitatory and inhibitory neurons, however, exhibit differential expression strength. These findings provide molecular evidence, at single-cell resolution, to support the hypothesis that the excitatory/inhibitory (E/I) neuronal expression ratios of sodium channels are important regulatory mechanisms underlying brain homeostasis and neurological diseases. Modulating the E/I expression balance within cell types of sodium channels could serve as a potential strategy to develop targeted treatment for NaV-associated neuronal developmental disorders.


Assuntos
Encéfalo/metabolismo , Deficiências do Desenvolvimento/metabolismo , Neurônios/metabolismo , Canais de Sódio Disparados por Voltagem/metabolismo , Animais , Deficiências do Desenvolvimento/genética , Humanos , Camundongos , Canais de Sódio Disparados por Voltagem/genética
18.
Neuron ; 107(1): 38-51.e8, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32353253

RESUMO

Optogenetics is among the most widely employed techniques to manipulate neuronal activity. However, a major drawback is the need for invasive implantation of optical fibers. To develop a minimally invasive optogenetic method that overcomes this challenge, we engineered a new step-function opsin with ultra-high light sensitivity (SOUL). We show that SOUL can activate neurons located in deep mouse brain regions via transcranial optical stimulation and elicit behavioral changes in SOUL knock-in mice. Moreover, SOUL can be used to modulate neuronal spiking and induce oscillations reversibly in macaque cortex via optical stimulation from outside the dura. By enabling external light delivery, our new opsin offers a minimally invasive tool for manipulating neuronal activity in rodent and primate models with fewer limitations on the depth and size of target brain regions and may further facilitate the development of minimally invasive optogenetic tools for the treatment of neurological disorders.


Assuntos
Opsinas , Optogenética/métodos , Animais , Encéfalo/fisiologia , Macaca , Camundongos , Modelos Animais , Neurônios/fisiologia
19.
Transl Psychiatry ; 10(1): 29, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-32066662

RESUMO

CACNA1I, a schizophrenia risk gene, encodes a subtype of voltage-gated T-type calcium channel CaV3.3. We previously reported that a patient-derived missense de novo mutation (R1346H) of CACNA1I impaired CaV3.3 channel function. Here, we generated CaV3.3-RH knock-in animals, along with mice lacking CaV3.3, to investigate the biological impact of R1346H (RH) variation. We found that RH mutation altered cellular excitability in the thalamic reticular nucleus (TRN), where CaV3.3 is abundantly expressed. Moreover, RH mutation produced marked deficits in sleep spindle occurrence and morphology throughout non-rapid eye movement (NREM) sleep, while CaV3.3 haploinsufficiency gave rise to largely normal spindles. Therefore, mice harboring the RH mutation provide a patient derived genetic model not only to dissect the spindle biology but also to evaluate the effects of pharmacological reagents in normalizing sleep spindle deficits. Importantly, our analyses highlighted the significance of characterizing individual spindles and strengthen the inferences we can make across species over sleep spindles. In conclusion, this study established a translational link between a genetic allele and spindle deficits during NREM observed in schizophrenia patients, representing a key step toward testing the hypothesis that normalizing spindles may be beneficial for schizophrenia patients.


Assuntos
Canais de Cálcio Tipo T , Esquizofrenia , Animais , Eletroencefalografia , Humanos , Camundongos , Esquizofrenia/genética , Sono , Sono REM
20.
Nat Neurosci ; 23(12): 1629-1636, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32807948

RESUMO

Recent success in identifying gene-regulatory elements in the context of recombinant adeno-associated virus vectors has enabled cell-type-restricted gene expression. However, within the cerebral cortex these tools are largely limited to broad classes of neurons. To overcome this limitation, we developed a strategy that led to the identification of multiple new enhancers to target functionally distinct neuronal subtypes. By investigating the regulatory landscape of the disease gene Scn1a, we discovered enhancers selective for parvalbumin (PV) and vasoactive intestinal peptide-expressing interneurons. Demonstrating the functional utility of these elements, we show that the PV-specific enhancer allowed for the selective targeting and manipulation of these neurons across vertebrate species, including humans. Finally, we demonstrate that our selection method is generalizable and characterizes additional PV-specific enhancers with exquisite specificity within distinct brain regions. Altogether, these viral tools can be used for cell-type-specific circuit manipulation and hold considerable promise for use in therapeutic interventions.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Interneurônios/fisiologia , Animais , Callithrix , Córtex Cerebral/citologia , Feminino , Humanos , Macaca mulatta , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Neurônios , Parvalbuminas/fisiologia , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Peptídeo Intestinal Vasoativo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA