Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
STAR Protoc ; 4(1): 101967, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36856768

RESUMO

Obtaining mechanistic insights into the disruptions of neuronal excitation and inhibition (E/I) balance in brain disorders has remained challenging. Here, we present a protocol for in vitro characterization of E/I balance. Using human induced pluripotent stem cells, we describe the generation of glutamatergic excitatory/GABAergic inhibitory neuronal co-cultures at defined ratios, followed by analyzing E/I network properties using immunocytochemistry and multi-electrode array recording. This approach allows for studying cell-type-specific contribution of disease genes to E/I balance in human neurons. For complete details on the use and execution of this protocol, please refer to Mossink et al. (2022)1 and Wang et al. (2022).2.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Técnicas de Cocultura , Neurônios GABAérgicos
2.
Mol Psychiatry ; 27(1): 1-18, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33972691

RESUMO

Activity in the healthy brain relies on a concerted interplay of excitation (E) and inhibition (I) via balanced synaptic communication between glutamatergic and GABAergic neurons. A growing number of studies imply that disruption of this E/I balance is a commonality in many brain disorders; however, obtaining mechanistic insight into these disruptions, with translational value for the patient, has typically been hampered by methodological limitations. Cadherin-13 (CDH13) has been associated with autism and attention-deficit/hyperactivity disorder. CDH13 localizes at inhibitory presynapses, specifically of parvalbumin (PV) and somatostatin (SST) expressing GABAergic neurons. However, the mechanism by which CDH13 regulates the function of inhibitory synapses in human neurons remains unknown. Starting from human-induced pluripotent stem cells, we established a robust method to generate a homogenous population of SST and MEF2C (PV-precursor marker protein) expressing GABAergic neurons (iGABA) in vitro, and co-cultured these with glutamatergic neurons at defined E/I ratios on micro-electrode arrays. We identified functional network parameters that are most reliably affected by GABAergic modulation as such, and through alterations of E/I balance by reduced expression of CDH13 in iGABAs. We found that CDH13 deficiency in iGABAs decreased E/I balance by means of increased inhibition. Moreover, CDH13 interacts with Integrin-ß1 and Integrin-ß3, which play opposite roles in the regulation of inhibitory synaptic strength via this interaction. Taken together, this model allows for standardized investigation of the E/I balance in a human neuronal background and can be deployed to dissect the cell-type-specific contribution of disease genes to the E/I balance.


Assuntos
Caderinas , Neurônios GABAérgicos , Parvalbuminas , Caderinas/metabolismo , Neurônios GABAérgicos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Integrinas/metabolismo , Parvalbuminas/metabolismo , Sinapses/metabolismo
3.
Neurobiol Dis ; 163: 105587, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34923109

RESUMO

Monoamine neurotransmitter abundance affects motor control, emotion, and cognitive function and is regulated by monoamine oxidases. Among these, Monoamine oxidase A (MAOA) catalyzes the degradation of dopamine, norepinephrine, and serotonin into their inactive metabolites. Loss-of-function mutations in the X-linked MAOA gene have been associated with Brunner syndrome, which is characterized by various forms of impulsivity, maladaptive externalizing behavior, and mild intellectual disability. Impaired MAOA activity in individuals with Brunner syndrome results in bioamine aberration, but it is currently unknown how this affects neuronal function, specifically in dopaminergic (DA) neurons. Here we generated human induced pluripotent stem cell (hiPSC)-derived DA neurons from three individuals with Brunner syndrome carrying different mutations and characterized neuronal properties at the single cell and neuronal network level in vitro. DA neurons of Brunner syndrome patients showed reduced synaptic density but exhibited hyperactive network activity. Intrinsic functional properties and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated synaptic transmission were not affected in DA neurons of individuals with Brunner syndrome. Instead, we show that the neuronal network hyperactivity is mediated by upregulation of the GRIN2A and GRIN2B subunits of the N-methyl-d-aspartate receptor (NMDAR), resulting in increased NMDAR-mediated currents. By correcting a MAOA missense mutation with CRISPR/Cas9 genome editing we normalized GRIN2A and GRIN2B expression, NMDAR function and neuronal population activity to control levels. Our data suggest that MAOA mutations in Brunner syndrome increase the activity of dopaminergic neurons through upregulation of NMDAR function, which may contribute to the etiology of Brunner syndrome associated phenotypes.


Assuntos
Transtornos Disruptivos, de Controle do Impulso e da Conduta/genética , Neurônios Dopaminérgicos/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/genética , Deficiência Intelectual/genética , Monoaminoxidase/deficiência , Monoaminoxidase/genética , Mutação , Polimorfismo de Nucleotídeo Único , Receptores de N-Metil-D-Aspartato/metabolismo , Agressão , Transtornos Disruptivos, de Controle do Impulso e da Conduta/metabolismo , Transtornos Disruptivos, de Controle do Impulso e da Conduta/fisiopatologia , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas , Deficiência Intelectual/metabolismo , Deficiência Intelectual/fisiopatologia , Masculino , Monoaminoxidase/metabolismo , Rede Nervosa/metabolismo , Rede Nervosa/fisiopatologia , Sinapses/metabolismo , Transmissão Sináptica/genética
4.
Stem Cell Reports ; 16(9): 2182-2196, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34329594

RESUMO

Micro-electrode arrays (MEAs) are increasingly used to characterize neuronal network activity of human induced pluripotent stem cell (hiPSC)-derived neurons. Despite their gain in popularity, MEA recordings from hiPSC-derived neuronal networks are not always used to their full potential in respect to experimental design, execution, and data analysis. Therefore, we benchmarked the robustness of MEA-derived neuronal activity patterns from ten healthy individual control lines, and uncover comparable network phenotypes. To achieve standardization, we provide recommendations on experimental design and analysis. With such standardization, MEAs can be used as a reliable platform to distinguish (disease-specific) network phenotypes. In conclusion, we show that MEAs are a powerful and robust tool to uncover functional neuronal network phenotypes from hiPSC-derived neuronal networks, and provide an important resource to advance the hiPSC field toward the use of MEAs for disease phenotyping and drug discovery.


Assuntos
Técnicas de Cultura de Células , Eletrodos , Estudos de Associação Genética/métodos , Dispositivos Lab-On-A-Chip , Análise em Microsséries/métodos , Neurônios/citologia , Neurônios/metabolismo , Potenciais de Ação , Animais , Diferenciação Celular , Células Cultivadas , Estudos de Associação Genética/instrumentação , Humanos , Camundongos , Análise em Microsséries/instrumentação , Rede Nervosa
5.
J Neural Eng ; 18(3): 036016, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33724235

RESUMO

OBJECTIVE: In ischemic stroke, treatments to protect neurons from irreversible damage are urgently needed. Studies in animal models have shown that neuroprotective treatments targeting neuronal silencing improve brain recovery, but in clinical trials none of these were effective in patients. This failure of translation poses doubts on the real efficacy of treatments tested and on the validity of animal models for human stroke. Here, we established a human neuronal model of the ischemic penumbra by using human induced pluripotent stem cells and we provided an in-depth characterization of neuronal responses to hypoxia and treatment strategies at the network level. APPROACH: We generated neurons from induced pluripotent stem cells derived from healthy donor and we cultured them on micro-electrode arrays. We measured the electrophysiological activity of human neuronal networks under controlled hypoxic conditions. We tested the effect of different treatment strategies on neuronal network functionality. MAIN RESULTS: Human neuronal networks are vulnerable to hypoxia reflected by a decrease in activity and synchronicity under low oxygen conditions. We observe that full, partial or absent recovery depend on the timing of re-oxygenation and we provide a critical time threshold that, if crossed, is associated with irreversible impairments. We found that hypoxic preconditioning improves resistance to a second hypoxic insult. Finally, in contrast to previously tested, ineffective treatments, we show that stimulatory treatments counteracting neuronal silencing during hypoxia, such as optogenetic stimulation, are neuroprotective. SIGNIFICANCE: We presented a human neuronal model of the ischemic penumbra and we provided insights that may offer the basis for novel therapeutic approaches for patients after stroke. The use of human neurons might improve drug discovery and translation of findings to patients and might open new perspectives for personalized investigations.


Assuntos
Isquemia Encefálica , Células-Tronco Pluripotentes Induzidas , Fármacos Neuroprotetores , Animais , Isquemia Encefálica/terapia , Humanos , Hipóxia , Neurônios
6.
Cell Mol Life Sci ; 78(6): 2517-2563, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33263776

RESUMO

Neurodevelopmental disorders (NDDs), including intellectual disability (ID) and autism spectrum disorders (ASD), are a large group of disorders in which early insults during brain development result in a wide and heterogeneous spectrum of clinical diagnoses. Mutations in genes coding for chromatin remodelers are overrepresented in NDD cohorts, pointing towards epigenetics as a convergent pathogenic pathway between these disorders. In this review we detail the role of NDD-associated chromatin remodelers during the developmental continuum of progenitor expansion, differentiation, cell-type specification, migration and maturation. We discuss how defects in chromatin remodelling during these early developmental time points compound over time and result in impaired brain circuit establishment. In particular, we focus on their role in the three largest cell populations: glutamatergic neurons, GABAergic neurons, and glia cells. An in-depth understanding of the spatiotemporal role of chromatin remodelers during neurodevelopment can contribute to the identification of molecular targets for treatment strategies.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Transtornos do Neurodesenvolvimento/patologia , Animais , Cromatina/química , DNA Helicases/genética , DNA Helicases/metabolismo , Epigênese Genética , Histonas/genética , Histonas/metabolismo , Humanos , Transtornos do Neurodesenvolvimento/metabolismo , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional/genética
7.
Cell Rep ; 31(3): 107538, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32320658

RESUMO

Epilepsy, intellectual and cortical sensory deficits, and psychiatric manifestations are the most frequent manifestations of mitochondrial diseases. How mitochondrial dysfunction affects neural structure and function remains elusive, mostly because of a lack of proper in vitro neuronal model systems with mitochondrial dysfunction. Leveraging induced pluripotent stem cell technology, we differentiated excitatory cortical neurons (iNeurons) with normal (low heteroplasmy) and impaired (high heteroplasmy) mitochondrial function on an isogenic nuclear DNA background from patients with the common pathogenic m.3243A > G variant of mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS). iNeurons with high heteroplasmy exhibited mitochondrial dysfunction, delayed neural maturation, reduced dendritic complexity, and fewer excitatory synapses. Micro-electrode array recordings of neuronal networks displayed reduced network activity and decreased synchronous network bursting. Impaired neuronal energy metabolism and compromised structural and functional integrity of neurons and neural networks could be the primary drivers of increased susceptibility to neuropsychiatric manifestations of mitochondrial disease.


Assuntos
Mitocôndrias/metabolismo , Neurônios/metabolismo , Animais , Diferenciação Celular , Humanos , Ratos , Ratos Wistar
8.
Cell Rep ; 30(1): 173-186.e6, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31914384

RESUMO

Pathogenic mutations in either one of the epigenetic modifiers EHMT1, MBD5, MLL3, or SMARCB1 have been identified to be causative for Kleefstra syndrome spectrum (KSS), a neurodevelopmental disorder with clinical features of both intellectual disability (ID) and autism spectrum disorder (ASD). To understand how these variants lead to the phenotypic convergence in KSS, we employ a loss-of-function approach to assess neuronal network development at the molecular, single-cell, and network activity level. KSS-gene-deficient neuronal networks all develop into hyperactive networks with altered network organization and excitatory-inhibitory balance. Interestingly, even though transcriptional data reveal distinct regulatory mechanisms, KSS target genes share similar functions in regulating neuronal excitability and synaptic function, several of which are associated with ID and ASD. Our results show that KSS genes mainly converge at the level of neuronal network communication, providing insights into the pathophysiology of KSS and phenotypically congruent disorders.


Assuntos
Transtorno Autístico/genética , Transtorno Autístico/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Rede Nervosa/metabolismo , Animais , Deleção Cromossômica , Cromossomos Humanos Par 9/genética , Anormalidades Craniofaciais/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Cardiopatias Congênitas/genética , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/deficiência , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Inibição Neural , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Ratos Wistar , Sinapses/metabolismo
9.
Nat Commun ; 10(1): 4928, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31666522

RESUMO

Kleefstra syndrome (KS) is a neurodevelopmental disorder caused by mutations in the histone methyltransferase EHMT1. To study the impact of decreased EHMT1 function in human cells, we generated excitatory cortical neurons from induced pluripotent stem (iPS) cells derived from KS patients. Neuronal networks of patient-derived cells exhibit network bursting with a reduced rate, longer duration, and increased temporal irregularity compared to control networks. We show that these changes are mediated by upregulation of NMDA receptor (NMDAR) subunit 1 correlating with reduced deposition of the repressive H3K9me2 mark, the catalytic product of EHMT1, at the GRIN1 promoter. In mice EHMT1 deficiency leads to similar neuronal network impairments with increased NMDAR function. Finally, we rescue the KS patient-derived neuronal network phenotypes by pharmacological inhibition of NMDARs. Summarized, we demonstrate a direct link between EHMT1 deficiency and NMDAR hyperfunction in human neurons, providing a potential basis for more targeted therapeutic approaches for KS.


Assuntos
Anormalidades Craniofaciais/genética , Cardiopatias Congênitas/genética , Histona-Lisina N-Metiltransferase/genética , Deficiência Intelectual/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Animais , Córtex Cerebral/citologia , Deleção Cromossômica , Cromossomos Humanos Par 9/genética , Cromossomos Humanos Par 9/metabolismo , Anormalidades Craniofaciais/metabolismo , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Cardiopatias Congênitas/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas , Deficiência Intelectual/metabolismo , Mutação com Perda de Função , Masculino , Camundongos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Cultura Primária de Células , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Regulação para Cima
10.
Cell Rep ; 10(3): 339-345, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25600869

RESUMO

The biogenesis of ribosomes and their coordination of protein translation consume an enormous amount of cellular energy. As such, it has been established that the inhibition of either process can extend eukaryotic lifespan. Here, we used next-generation sequencing to compare ribosome-associated RNAs from normal strains of Caenorhabditis elegans to those carrying the life-extending daf-2 mutation. We found a long noncoding RNA (lncRNA), transcribed telomeric sequence 1 (tts-1), on ribosomes of the daf-2 mutant. Depleting tts-1 in daf-2 mutants increases ribosome levels and significantly shortens their extended lifespan. We find tts-1 is also required for the longer lifespan of the mitochondrial clk-1 mutants but not the feeding-defective eat-2 mutants. In line with this, the clk-1 mutants express more tts-1 and fewer ribosomes than the eat-2 mutants. Our results suggest that the expression of tts-1 functions in different longevity pathways to reduce ribosome levels in a way that promotes life extension.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA