Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Biol Chem ; 291(34): 18030-40, 2016 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-27382060

RESUMO

Glycine receptors are chloride-permeable, ligand-gated ion channels and contribute to the inhibition of neuronal firing in the central nervous system or to facilitation of neurotransmitter release if expressed at presynaptic sites. Recent structure-function studies have provided detailed insights into the mechanisms of channel gating, desensitization, and ion permeation. However, most of the work has focused only on comparing a few isoforms, and among studies, different cellular expression systems were used. Here, we performed a series of experiments using recombinantly expressed homomeric and heteromeric glycine receptor channels, including their splice variants, in the same cellular expression system to investigate and compare their electrophysiological properties. Our data show that the current-voltage relationships of homomeric channels formed by the α2 or α3 subunits change upon receptor desensitization from a linear to an inwardly rectifying shape, in contrast to their heteromeric counterparts. The results demonstrate that inward rectification depends on a single amino acid (Ala(254)) at the inner pore mouth of the channels and is closely linked to chloride permeation. We also show that the current-voltage relationships of glycine-evoked currents in primary hippocampal neurons are inwardly rectifying upon desensitization. Thus, the alanine residue Ala(254) determines voltage-dependent rectification upon receptor desensitization and reveals a physio-molecular signature of homomeric glycine receptor channels, which provides unprecedented opportunities for the identification of these channels at the single cell level.


Assuntos
Fenômenos Eletrofisiológicos , Multimerização Proteica/fisiologia , Receptores de Glicina/metabolismo , Processamento Alternativo/fisiologia , Substituição de Aminoácidos , Animais , Humanos , Masculino , Mutação de Sentido Incorreto , Ratos , Ratos Wistar , Receptores de Glicina/genética
2.
Cereb Cortex ; 26(5): 2325-2340, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26908632

RESUMO

Parvalbumin-positive (PV) basket cells provide perisomatic inhibition in the cortex and hippocampus and control generation of memory-related network activity patterns, such as sharp wave ripples (SPW-R). Deterioration of this class of fast-spiking interneurons has been observed in neuropsychiatric disorders and evidence from animal models suggests their involvement in the acquisition and extinction of fear memories. Here, we used mice with neuron type-targeted expression of the presynaptic gain-of-function glycine receptor RNA variant GlyR α3L(185L)to genetically enhance the network activity of PV interneurons. These mice showed reduced extinction of contextual fear memory but normal auditory cued fear memory. They furthermore displayed increase of SPW-R activity in area CA3 and CA1 and facilitated propagation of this particular network activity pattern, as determined in ventral hippocampal slice preparations. Individual freezing levels during extinction and SPW-R propagation were correlated across genotypes. The same was true for parvalbumin immunoreactivity in the ventral hippocampus, which was generally augmented in the GlyR mutant mice and correlated with individual freezing levels. Together, these results identify PV interneurons as critical cellular substrate of fear memory persistence and associated SPW-R activity in the hippocampus. Our findings may be relevant for the identification and characterization of physiological correlates for posttraumatic stress and anxiety disorders.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Neurônios GABAérgicos/fisiologia , Interneurônios/fisiologia , Memória/fisiologia , Parvalbuminas/metabolismo , Animais , Aprendizagem da Esquiva/fisiologia , Condicionamento Clássico , Feminino , Hipocampo/fisiologia , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Transgênicos
3.
J Cell Sci ; 127(Pt 17): 3687-98, 2014 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-24994934

RESUMO

The neuronal function of Cys-loop neurotransmitter receptors is established; however, their role in non-neuronal cells is poorly defined. As brain tumors are enriched in the neurotransmitter glycine, we studied the expression and function of glycine receptors (GlyRs) in glioma cells. Human brain tumor biopsies selectively expressed the GlyR α1 and α3 subunits, which have nuclear localization signals (NLSs). The mouse glioma cell line GL261 expressed GlyR α1, and knockdown of GlyR α1 protein expression impaired the self-renewal capacity and tumorigenicity of GL261 glioma cells, as shown by a neurosphere assay and GL261 cell inoculation in vivo, respectively. We furthermore showed that the pronounced tumorigenic effect of GlyR α1 relies on a new intracellular signaling function that depends on the NLS region in the large cytosolic loop and impacts on GL261 glioma cell gene regulation. Stable expression of GlyR α1 and α3 loops rescued the self-renewal capacity of GlyR α1 knockdown cells, which demonstrates their functional equivalence. The new intracellular signaling function identified here goes beyond the well-established role of GlyRs as neuronal ligand-gated ion channels and defines NLS-containing GlyRs as new potential targets for brain tumor therapies.


Assuntos
Citoplasma/metabolismo , Glioma/metabolismo , Receptores de Glicina/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Glioma/patologia , Glicina/metabolismo , Humanos , Camundongos , Receptores de Glicina/genética
4.
Proc Natl Acad Sci U S A ; 110(37): 15073-8, 2013 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-23980149

RESUMO

GABA(B) receptors (GABA(B)Rs) mediate slow inhibitory effects on neuronal excitability and synaptic transmission in the brain. However, the GABA(B)R agonist baclofen can also promote excitability and seizure generation in human patients and animals models. Here we show that baclofen has concentration-dependent effects on the hippocampal network in a mouse model of mesial temporal lobe epilepsy. Application of baclofen at a high dose (10 mg/kg i.p.) reduced the power of γ oscillations and the frequency of pathological discharges in the Cornu Ammonis area 3 (CA3) area of freely moving epileptic mice. Unexpectedly, at a lower dose (1 mg/kg), baclofen markedly increased γ activity accompanied by a higher incidence of pathological discharges. Intracellular recordings from CA3 pyramidal cells in vitro further revealed that, although at a high concentration (10 µM), baclofen invariably resulted in hyperpolarization, at low concentrations (0.5 µM), the drug had divergent effects, producing depolarization and an increase in firing frequency in epileptic but not control mice. These excitatory effects were mediated by the selective muting of inhibitory cholecystokinin-positive basket cells (CCK(+) BCs), through enhanced inhibition of GABA release via presynaptic GABA(B)Rs. We conclude that cell type-specific up-regulation of GABA(B)R-mediated autoinhibition in CCK(+) BCs promotes aberrant high frequency oscillations and hyperexcitability in hippocampal networks of chronic epileptic mice.


Assuntos
Autorreceptores/fisiologia , Epilepsia do Lobo Temporal/fisiopatologia , Receptores de GABA-B/fisiologia , Animais , Baclofeno/administração & dosagem , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/patologia , Região CA3 Hipocampal/fisiopatologia , Colecistocinina/metabolismo , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos , Epilepsia do Lobo Temporal/patologia , Agonistas de Aminoácidos Excitatórios/administração & dosagem , Agonistas dos Receptores de GABA-B/administração & dosagem , Humanos , Ácido Caínico/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/patologia , Rede Nervosa/fisiopatologia
5.
J Biol Chem ; 289(39): 26973-26988, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25124043

RESUMO

The basic helix-loop-helix transcription factor hASH1, encoded by the ASCL1 gene, plays an important role in neurogenesis and tumor development. Recent findings indicate that local oxygen tension is a critical determinant for the progression of neuroblastomas. Here we investigated the molecular mechanisms underlying the oxygen-dependent expression of hASH1 in neuroblastoma cells. Exposure of human neuroblastoma-derived Kelly cells to 1% O2 significantly decreased ASCL1 mRNA and hASH1 protein levels. Using reporter gene assays, we show that the response of hASH1 to hypoxia is mediated mainly by post-transcriptional inhibition via the ASCL1 mRNA 5'- and 3'-UTRs, whereas additional inhibition of the ASCL1 promoter was observed under prolonged hypoxia. By RNA pulldown experiments followed by MALDI/TOF-MS analysis, we identified heterogeneous nuclear ribonucleoprotein (hnRNP)-A2/B1 and hnRNP-R as interactors binding directly to the ASCL1 mRNA 5'- and 3'-UTRs and influencing its expression. We further demonstrate that hnRNP-A2/B1 is a key positive regulator of ASCL1, findings that were also confirmed by analysis of a large compilation of gene expression data. Our data suggest that a prominent down-regulation of hnRNP-A2/B1 during hypoxia is associated with the post-transcriptional suppression of hASH1 synthesis. This novel post-transcriptional mechanism for regulating hASH1 levels will have important implications in neural cell fate development and disease.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/biossíntese , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular/genética , Linhagem Celular Tumoral , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Humanos , Proteínas de Neoplasias/genética , Neuroblastoma/genética , Regiões Promotoras Genéticas , Coelhos , Ratos Wistar
6.
Eur J Neurosci ; 37(4): 544-54, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23205938

RESUMO

GABAergic transmission is essential to brain function, and a large repertoire of GABA type A receptor (GABA(A) R) subunits is at a neuron's disposition to serve this function. The glycine receptor (GlyR)-associated protein gephyrin has been shown to be essential for the clustering of a subset of GABA(A) R. Despite recent progress in the field of gephyrin-dependent mechanisms of postsynaptic GABA(A) R stabilisation, the role of gephyrin in synaptic GABA(A) R localisation has remained a complex matter with many open questions. Here, we analysed comparatively the interaction of purified rat gephyrin and mouse brain gephyrin with the large cytoplasmic loops of GABA(A) R α1, α2, ß2 and ß3 subunits. Binding affinities were determined using surface plasmon resonance spectroscopy, and showed an ~ 20-fold lower affinity of the ß2 loop to gephyrin as compared to the GlyR ß loop-gephyrin interaction. We also probed in vivo binding in primary cortical neurons by the well-established use of chimaeras of GlyR α1 that harbour respective gephyrin-binding motifs derived from the different GABA(A) R subunits. These studies identify a novel gephyrin-binding motif in GABA(A) R ß2 and ß3 large cytoplasmic loops.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Western Blotting , Encéfalo/metabolismo , Células HEK293 , Humanos , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratos , Ratos Wistar , Receptores de GABA-A/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ressonância de Plasmônio de Superfície , Transfecção
8.
Front Mol Neurosci ; 10: 439, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29375302

RESUMO

C-to-U RNA editing of glycine receptors (GlyR) can play an important role in disease progression of temporal lobe epilepsy (TLE) as it may contribute in a neuron type-specific way to neuropsychiatric symptoms of the disease. It is therefore necessary to develop tools that allow identification of neuron types that express RNA-edited GlyR protein. In this study, we identify NH4 as agonist of C-to-U RNA edited GlyRs. Furthermore, we generated a new molecular C-to-U RNA editing sensor tool that detects Apobec-1- dependent RNA editing in HEPG2 cells and rat primary hippocampal neurons. Using this sensor combined with NH4 application, we were able to identify C-to-U RNA editing-competent neurons and expression of C-to-U RNA-edited GlyR protein in neurons. Bioinformatic analysis of 1,000 Genome Project Phase 3 allele frequencies coding for human Apobec-1 80M and 80I variants showed differences between populations, and the results revealed a preference of the 80I variant to generate RNA-edited GlyR protein. Finally, we established a new PCR-based restriction fragment length polymorphism (RFLP) approach to profile mRNA expression with regard to the genetic APOBEC1 dimorphism of patients with intractable temporal lobe epilepsy (iTLE) and found that the patients fall into two groups. Patients with expression of the Apobec-1 80I variant mostly suffered from simple or complex partial seizures, whereas patients with 80M expression exhibited secondarily generalized seizure activity. Thus, our method allows the characterization of Apobec-1 80M and 80l variants in the brain and provides a new way to epidemiologically and semiologically classify iTLE according to the two different APOBEC1 alleles. Together, these results demonstrate Apobec-1-dependent expression of RNA-edited GlyR protein in neurons and identify the APOBEC1 80I/M-coding alleles as new genetic risk factors for iTLE patients.

9.
J Clin Invest ; 127(12): 4365-4378, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29106383

RESUMO

Molybdenum cofactor deficiency (MoCD) is an autosomal recessive inborn error of metabolism characterized by neurodegeneration and death in early childhood. The rapid and progressive neurodegeneration in MoCD presents a major clinical challenge and may relate to the poor understanding of the molecular mechanisms involved. Recently, we reported that treating patients with cyclic pyranopterin monophosphate (cPMP) is a successful therapy for a subset of infants with MoCD and prevents irreversible brain damage. Here, we studied S-sulfocysteine (SSC), a structural analog of glutamate that accumulates in the plasma and urine of patients with MoCD, and demonstrated that it acts as an N-methyl D-aspartate receptor (NMDA-R) agonist, leading to calcium influx and downstream cell signaling events and neurotoxicity. SSC treatment activated the protease calpain, and calpain-dependent degradation of the inhibitory synaptic protein gephyrin subsequently exacerbated SSC-mediated excitotoxicity and promoted loss of GABAergic synapses. Pharmacological blockade of NMDA-R, calcium influx, or calpain activity abolished SSC and glutamate neurotoxicity in primary murine neurons. Finally, the NMDA-R antagonist memantine was protective against the manifestation of symptoms in a tungstate-induced MoCD mouse model. These findings demonstrate that SSC drives excitotoxic neurodegeneration in MoCD and introduce NMDA-R antagonists as potential therapeutics for this fatal disease.


Assuntos
Sinalização do Cálcio , Cisteína/análogos & derivados , Neurônios GABAérgicos/metabolismo , Erros Inatos do Metabolismo dos Metais/metabolismo , Doenças Neurodegenerativas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Cisteína/metabolismo , Modelos Animais de Doenças , Neurônios GABAérgicos/patologia , Células HEK293 , Humanos , Memantina/farmacologia , Erros Inatos do Metabolismo dos Metais/tratamento farmacológico , Erros Inatos do Metabolismo dos Metais/patologia , Camundongos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Compostos Organofosforados/farmacologia , Pterinas/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Sinapses/metabolismo , Sinapses/patologia , Compostos de Tungstênio/toxicidade
10.
PLoS One ; 10(5): e0125413, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25955356

RESUMO

Establishment of phylogenetic relationships remains a challenging task because it is based on computational analysis of genomic hot spots that display species-specific sequence variations. Here, we identify a species-specific thymine-to-guanine sequence variation in the Glrb gene which gives rise to species-specific splice donor sites in the Glrb genes of mouse and bushbaby. The resulting splice insert in the receptor for the inhibitory neurotransmitter glycine (GlyR) conveys synaptic receptor clustering and specific association with a particular synaptic plasticity-related splice variant of the postsynaptic scaffold protein gephyrin. This study identifies a new genomic hot spot which contributes to phylogenetic diversification of protein function and advances our understanding of phylogenetic relationships.


Assuntos
Evolução Biológica , Proteínas de Transporte/genética , Galago/genética , Genoma , Proteínas de Membrana/genética , Camundongos/genética , Receptores de Glicina/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Éxons , Galago/classificação , Galago/metabolismo , Expressão Gênica , Variação Genética , Íntrons , Proteínas de Membrana/metabolismo , Camundongos/classificação , Camundongos/metabolismo , Dados de Sequência Molecular , Plasticidade Neuronal , Neurônios/metabolismo , Neurônios/ultraestrutura , Filogenia , Receptores de Glicina/metabolismo , Especificidade da Espécie , Medula Espinal/citologia , Medula Espinal/metabolismo , Sinapses/metabolismo , Sinapses/ultraestrutura
11.
Front Cell Neurosci ; 8: 164, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24987332

RESUMO

Synaptic communication requires constant adjustments of pre- and postsynaptic efficacies. In addition to synaptic long term plasticity, the presynaptic machinery underlies homeostatic regulations which prevent out of range transmitter release. In this minireview we will discuss the relevance of selected presynaptic mechanisms to epilepsy including voltage- and ligand-gated ion channels as well as cannabinoid and adenosine receptor signaling.

12.
J Clin Invest ; 124(2): 696-711, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24430185

RESUMO

The mechanisms that regulate the strength of synaptic transmission and intrinsic neuronal excitability are well characterized; however, the mechanisms that promote disease-causing neural network dysfunction are poorly defined. We generated mice with targeted neuron type-specific expression of a gain-of-function variant of the neurotransmitter receptor for glycine (GlyR) that is found in hippocampectomies from patients with temporal lobe epilepsy. In this mouse model, targeted expression of gain-of-function GlyR in terminals of glutamatergic cells or in parvalbumin-positive interneurons persistently altered neural network excitability. The increased network excitability associated with gain-of-function GlyR expression in glutamatergic neurons resulted in recurrent epileptiform discharge, which provoked cognitive dysfunction and memory deficits without affecting bidirectional synaptic plasticity. In contrast, decreased network excitability due to gain-of-function GlyR expression in parvalbumin-positive interneurons resulted in an anxiety phenotype, but did not affect cognitive performance or discriminative associative memory. Our animal model unveils neuron type-specific effects on cognition, formation of discriminative associative memory, and emotional behavior in vivo. Furthermore, our data identify a presynaptic disease-causing molecular mechanism that impairs homeostatic regulation of neural network excitability and triggers neuropsychiatric symptoms.


Assuntos
Transtornos Cognitivos/fisiopatologia , Memória , Rede Nervosa , Animais , Ansiedade/metabolismo , Encéfalo/metabolismo , Citoplasma/metabolismo , Genótipo , Glutamina/química , Glutationa Transferase/metabolismo , Glicina/química , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Hipocampo/metabolismo , Homeostase , Humanos , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Plasticidade Neuronal/fisiologia , Oscilometria , Parvalbuminas/química , Fenótipo , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Transmissão Sináptica
13.
Front Mol Neurosci ; 4: 1, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21441980

RESUMO

Human achaete-scute homolog-1 (hASH1), encoded by the human ASCL1 gene, belongs to the family of basic helix-loop-helix transcription factors. hASH1 and its mammalian homolog Mash1 are expressed in the central and peripheral nervous system during development, and promote early neuronal differentiation. Furthermore, hASH1 is involved in the specification of neuronal subtype identities. Misexpression of the transcription factor is correlated with a variety of tumors, including lung cancer and neuroendocrine tumors. To gain insights into the molecular mechanisms of hASH1 regulation, we screened for conditions causing changes in hASH1 gene expression rate. We found that treatment of human neuroblastoma-derived Kelly cells with phorbol 12-myristate 13-acetate (PMA) resulted in a fast, strong and long-lasting suppression of hASH1 synthesis. Reporter gene assays with constructs, in which the luciferase activity was controlled either by the ASCL1 promoter or by the hASH1 mRNA untranslated regions (UTRs), revealed a mainly UTR-dependent mechanism. The hASH1 promoter activity was decreased only after 48 h of PMA administration. Our data indicate that different mechanisms acting consecutively at the transcriptional and post-transcriptional level are responsible for hASH1 suppression after PMA treatment. We provide evidence that short term inhibition of hASH1 synthesis is attributed to hASH1 mRNA destabilization, which seems to depend mainly on protein kinase C activity. Under prolonged conditions (48 h), hASH1 suppression is mediated by decreased promoter activity and inhibition of mRNA translation.

14.
J Gen Virol ; 90(Pt 9): 2221-33, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19439557

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous pathogen with a predilection for dendritic cells (DCs). Latently infected myeloid progenitor cells develop into actively infected DCs with impaired functionality, allowing dissemination and transfer of virus throughout the body. However, the viral genes expressed in DCs and their effect on the cellular transcriptome are currently unknown. We investigated human DCs infected with HCMV by using SuperSAGE, allowing us to analyse the transcriptomes of both host and pathogen simultaneously. A small number of viral transcripts were expressed strongly and rapidly post-infection. However, only two were of the immediate-early class, including one with an unknown function. The viral genes expressed reflected the cellular milieu, with the majority having a known or suspected immune-evasion function. Several viral genes identified lack a known function and may fulfil specialized roles within DCs. The cellular response to infection included a strong interferon response, induction of cytokine and anti-apoptotic genes and alterations in genes involved in antigen presentation. We demonstrated the validity of our approach by showing that novel changes first seen in the transcriptome were reflected in the phenotype of HCMV-infected DCs. Delineation of the transcriptional changes underlying the phenotype of HCMV-infected DCs allows a better understanding of how a herpesvirus infects DCs and pinpoints linkages between phenotype and specific viral genes.


Assuntos
Infecções por Citomegalovirus/genética , Citomegalovirus/genética , Células Dendríticas/virologia , Interações Hospedeiro-Patógeno , Células Cultivadas , Citomegalovirus/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Células Dendríticas/imunologia , Regulação Viral da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Virais/genética , Proteínas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa