Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 27(15): 2614-2627, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29741614

RESUMO

Loss of excitatory amino acid transporters (EAATs) has been implicated in a number of human diseases including spinocerebellar ataxias, Alzhiemer's disease and motor neuron disease. EAAT4 and GLAST/EAAT1 are the two predominant EAATs responsible for maintaining low extracellular glutamate levels and preventing neurotoxicity in the cerebellum, the brain region essential for motor control. Here using genetically modified mice we identify new critical roles for EAAT4 and GLAST/EAAT1 as modulators of Purkinje cell (PC) spontaneous firing patterns. We show high EAAT4 levels, by limiting mGluR1 signalling, are essential in constraining inherently heterogeneous firing of zebrin-positive PCs. Moreover mGluR1 antagonists were found to restore regular spontaneous PC activity and motor behaviour in EAAT4 knockout mice. In contrast, GLAST/EAAT1 expression is required to sustain normal spontaneous simple spike activity in low EAAT4 expressing (zebrin-negative) PCs by restricting NMDA receptor activation. Blockade of NMDA receptor activity restores spontaneous activity in zebrin-negative PCs of GLAST knockout mice and furthermore alleviates motor deficits. In addition both transporters have differential effects on PC survival, with zebrin-negative PCs more vulnerable to loss of GLAST/EAAT1 and zebrin-positive PCs more vulnerable to loss of EAAT4. These findings reveal that glutamate transporter dysfunction through elevated extracellular glutamate and the aberrant activation of extrasynaptic receptors can disrupt cerebellar output by altering spontaneous PC firing. This expands our understanding of disease mechanisms in cerebellar ataxias and establishes EAATs as targets for restoring homeostasis in a variety of neurological diseases where altered cerebellar output is now thought to play a key role in pathogenesis.


Assuntos
Cerebelo/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Transportador 4 de Aminoácido Excitatório/genética , Células de Purkinje/fisiologia , Animais , Ataxia/genética , Sobrevivência Celular/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 4 de Aminoácido Excitatório/metabolismo , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Células de Purkinje/citologia , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
2.
Brain ; 142(1): 80-92, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30544257

RESUMO

Alterations of the N-methyl-d-aspartate receptor (NMDAR) subunit GluN2A, encoded by GRIN2A, have been associated with a spectrum of neurodevelopmental disorders with prominent speech-related features, and epilepsy. We performed a comprehensive assessment of phenotypes with a standardized questionnaire in 92 previously unreported individuals with GRIN2A-related disorders. Applying the criteria of the American College of Medical Genetics and Genomics to all published variants yielded 156 additional cases with pathogenic or likely pathogenic variants in GRIN2A, resulting in a total of 248 individuals. The phenotypic spectrum ranged from normal or near-normal development with mild epilepsy and speech delay/apraxia to severe developmental and epileptic encephalopathy, often within the epilepsy-aphasia spectrum. We found that pathogenic missense variants in transmembrane and linker domains (misTMD+Linker) were associated with severe developmental phenotypes, whereas missense variants within amino terminal or ligand-binding domains (misATD+LBD) and null variants led to less severe developmental phenotypes, which we confirmed in a discovery (P = 10-6) as well as validation cohort (P = 0.0003). Other phenotypes such as MRI abnormalities and epilepsy types were also significantly different between the two groups. Notably, this was paralleled by electrophysiology data, where misTMD+Linker predominantly led to NMDAR gain-of-function, while misATD+LBD exclusively caused NMDAR loss-of-function. With respect to null variants, we show that Grin2a+/- cortical rat neurons also had reduced NMDAR function and there was no evidence of previously postulated compensatory overexpression of GluN2B. We demonstrate that null variants and misATD+LBD of GRIN2A do not only share the same clinical spectrum (i.e. milder phenotypes), but also result in similar electrophysiological consequences (loss-of-function) opposing those of misTMD+Linker (severe phenotypes; predominantly gain-of-function). This new pathomechanistic model may ultimately help in predicting phenotype severity as well as eligibility for potential precision medicine approaches in GRIN2A-related disorders.


Assuntos
Epilepsia/genética , Transtornos do Neurodesenvolvimento/genética , Receptores de N-Metil-D-Aspartato/genética , Adolescente , Adulto , Idoso , Animais , Células Cultivadas , Córtex Cerebelar/metabolismo , Criança , Pré-Escolar , Epilepsia/fisiopatologia , Feminino , Genótipo , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Mutação , Transtornos do Neurodesenvolvimento/fisiopatologia , Fenótipo , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia , Adulto Jovem
3.
J Physiol ; 597(6): 1691-1704, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30604514

RESUMO

KEY POINTS: NMDA receptors are neurotransmitter-gated ion channels that are critically involved in brain cell communication Variations in genes encoding NMDA receptor subunits have been found in a range of neurodevelopmental disorders. We investigated a de novo genetic variant found in patients with epileptic encephalopathy that changes a residue located in the ion channel pore of the GluN2A NMDA receptor subunit. We found that this variant (GluN2AN615K ) impairs physiologically important receptor properties: it markedly reduces Mg2+ blockade and channel conductance, even for receptors in which one GluN2AN615K is co-assembled with one wild-type GluN2A subunit. Our findings are consistent with the GluN2AN615K mutation being the primary cause of the severe neurodevelopmental disorder in carriers. ABSTRACT: NMDA receptors are ionotropic calcium-permeable glutamate receptors with a voltage-dependence mediated by blockade by Mg2+ . Their activation is important in signal transduction, as well as synapse formation and maintenance. Two unrelated individuals with epileptic encephalopathy carry a de novo variant in the gene encoding the GluN2A NMDA receptor subunit: a N615K missense variant in the M2 pore helix (GRIN2AC1845A ). We hypothesized that this variant underlies the neurodevelopmental disorders in carriers and explored its functional consequences by electrophysiological analysis in heterologous systems. We focused on GluN2AN615K co-expressed with wild-type GluN2 subunits in physiologically relevant triheteromeric NMDA receptors containing two GluN1 and two distinct GluN2 subunits, whereas previous studies have investigated the impact of the variant in diheteromeric NMDA receptors with two GluN1 and two identical GluN2 subunits. We found that GluN2AN615K -containing triheteromers showed markedly reduced Mg2+ blockade, with a value intermediate between GluN2AN615K diheteromers and wild-type NMDA receptors. Single-channel conductance was reduced by four-fold in GluN2AN615K diheteromers, again with an intermediate value in GluN2AN615K -containing triheteromers. Glutamate deactivation rates were unaffected. Furthermore, we expressed GluN2AN615K in cultured primary mouse cortical neurons, observing a decrease in Mg2+ blockade and reduction in current density, confirming that the variant continues to have significant functional impact in neuronal systems. Our results demonstrate that the GluN2AN615K variant has substantial effects on NMDA receptor properties fundamental to the roles of the receptor in synaptic plasticity, even when expressed alongside wild-type subunits. This work strengthens the evidence indicating that the GluN2AN615K variant underlies the disabling neurodevelopmental phenotype in carriers.


Assuntos
Potenciais de Ação , Epilepsia/genética , Mutação de Sentido Incorreto , Receptores de N-Metil-D-Aspartato/genética , Animais , Células Cultivadas , Feminino , Células HEK293 , Humanos , Magnésio/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , Multimerização Proteica , Receptores de N-Metil-D-Aspartato/metabolismo
4.
BMC Genomics ; 19(1): 318, 2018 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-29720086

RESUMO

BACKGROUND: CRISPR/Cas9 enables the targeting of genes in zygotes; however, efficient approaches to create loxP-flanked (floxed) alleles remain elusive. RESULTS: Here, we show that the electroporation of Cas9, two gRNAs, and long single-stranded DNA (lssDNA) into zygotes, termed CLICK (CRISPR with lssDNA inducing conditional knockout alleles), enables the quick generation of floxed alleles in mice and rats. CONCLUSIONS: The high efficiency of CLICK provides homozygous knock-ins in oocytes carrying tissue-specific Cre, which allows the one-step generation of conditional knockouts in founder (F0) mice.


Assuntos
Engenharia Genética/métodos , Alelos , Animais , Sequência de Bases , Sistemas CRISPR-Cas/genética , Injeções , Camundongos , Camundongos Knockout , Zigoto/metabolismo
5.
Hum Mol Genet ; 23(14): 3875-82, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24603075

RESUMO

Beta III spectrin is present throughout the elaborate dendritic tree of cerebellar Purkinje cells and is required for normal neuronal morphology and cell survival. Spinocerebellar ataxia type 5 (SCA5) and spectrin associated autosomal recessive cerebellar ataxia type 1 are human neurodegenerative diseases involving progressive gait ataxia and cerebellar atrophy. Both disorders appear to result from loss of ß-III spectrin function. Further elucidation of ß-III spectrin function is therefore needed to understand disease mechanisms and identify potential therapeutic options. Here, we report that ß-III spectrin is essential for the recruitment and maintenance of ankyrin R at the plasma membrane of Purkinje cell dendrites. Two SCA5-associated mutations of ß-III spectrin both reduce ankyrin R levels at the cell membrane. Moreover, a wild-type ß-III spectrin/ankyrin-R complex increases sodium channel levels and activity in cell culture, whereas mutant ß-III spectrin complexes fail to enhance sodium currents. This suggests impaired ability to form stable complexes between the adaptor protein ankyrin R and its interacting partners in the Purkinje cell dendritic tree is a key mechanism by which mutant forms of ß-III spectrin cause ataxia, initially by Purkinje cell dysfunction and exacerbated by subsequent cell death.


Assuntos
Anquirinas/metabolismo , Células de Purkinje/metabolismo , Canais de Sódio/fisiologia , Espectrina/genética , Espectrina/metabolismo , Ataxias Espinocerebelares/genética , Animais , Membrana Celular/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Mutação , Estabilidade Proteica , Células de Purkinje/ultraestrutura , Ratos , Ratos Sprague-Dawley
6.
Lancet ; 385 Suppl 1: S65, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26312887

RESUMO

UNLABELLED: Background GRIN2A encodes the GluN2A subunit of the NMDA receptor (NMDAR), an ionotropic glutamate receptor that has important roles in synaptogenesis and synaptic plasticity. Some individuals with early onset epilepsies and intellectual disability carry heterozygous missense mutations in this gene, including a de-novo mutation in the receptor pore region (GluN2A(N615K)). We hypothesised that this mutation underlies the carrier's brain disorder and sought to explore its functional consequences. METHODS: We made two-electrode voltage clamp recordings from Xenopus laevis oocytes expressing GluN1/GluN2A(N615K) (N615K) NMDARs and compared them with wild-type (WT) NMDARs to assess the mutation's effect on potency of inhibition by Mg(2+) and other channel blockers. We then used whole-cell patch-clamping to evaluate NMDAR-mediated currents in mouse primary cortical pyramidal neurons transfected with either GluN2A(WT) or GluN2A(N651K) subunits. Means were compared by use of independent two-tailed t tests. FINDINGS: In oocytes, Mg(2+) (1 mM) block at -60 mV was significantly decreased (N615K [n=13], mean 5% [SE 8] vs WT [n=15], 89 [4]; p<0·0001). Furthermore, in N615K (n=17) and WT (n=17) oocytes, block by 10 µM memantine was also reduced (mean 26% [6] vs 75 [7], p<0·0001) as was block by 100 µM amantadine (18% [4] vs 44 [12], p<0·0001). Block by ketamine (N615K, n=14; WT, n=14) was not significantly affected, whereas block by dextromethorphan was increased (N615K [n=9], 56% [8] vs WT [n=8], 44 [6]; p=0·003). In N615K (n=10) and WT (n=10) neurons we observed a significant decrease in Mg(2+) sensitivity (49% [18] vs 95 [5], p<0·0001) and a significant decrease in current density (42 pA/pF [19] vs 61 [20], p=0·044). INTERPRETATION: This study suggests that the disease-associated mutation GluN2A(N615K) has substantial effects on NMDAR inhibition by both endogenous and exogenous channel blockers, and on NMDA current density. It is plausible that these changes underlie the carrier's phenotype. FUNDING: Wellcome Trust via an Edinburgh Clinical Academic Training PhD Fellowship.

7.
Brain ; 138(Pt 7): 1817-32, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25981959

RESUMO

Cerebral palsy is a sporadic disorder with multiple likely aetiologies, but frequently considered to be caused by birth asphyxia. Genetic investigations are rarely performed in patients with cerebral palsy and there is little proven evidence of genetic causes. As part of a large project investigating children with ataxia, we identified four patients in our cohort with a diagnosis of ataxic cerebral palsy. They were investigated using either targeted next generation sequencing or trio-based exome sequencing and were found to have mutations in three different genes, KCNC3, ITPR1 and SPTBN2. All the mutations were de novo and associated with increased paternal age. The mutations were shown to be pathogenic using a combination of bioinformatics analysis and in vitro model systems. This work is the first to report that the ataxic subtype of cerebral palsy can be caused by de novo dominant point mutations, which explains the sporadic nature of these cases. We conclude that at least some subtypes of cerebral palsy may be caused by de novo genetic mutations and patients with a clinical diagnosis of cerebral palsy should be genetically investigated before causation is ascribed to perinatal asphyxia or other aetiologies.


Assuntos
Ataxia/genética , Paralisia Cerebral/genética , Doenças Genéticas Inatas/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Mutação Puntual , Canais de Potássio Shaw/genética , Espectrina/genética , Sequência de Bases , Criança , Pré-Escolar , Análise Mutacional de DNA , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Recém-Nascido , Masculino , Técnicas de Patch-Clamp , Análise de Sequência de DNA
8.
Brain Commun ; 6(3): fcae184, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38846532

RESUMO

Amyotrophic lateral sclerosis is an age-dependent cell type-selective degenerative disease. Genetic studies indicate that amyotrophic lateral sclerosis is part of a spectrum of disorders, ranging from spinal muscular atrophy to frontotemporal dementia that share common pathological mechanisms. Amyotrophic lateral sclerosis Type 8 is a familial disease caused by mis-sense mutations in VAPB. VAPB is localized to the cytoplasmic surface of the endoplasmic reticulum, where it serves as a docking point for cytoplasmic proteins and mediates inter-organelle interactions with the endoplasmic reticulum membrane. A gene knock-in model of amyotrophic lateral sclerosis Type 8 based on the VapBP56S mutation and VapB gene deletion has been generated in rats. These animals display a range of age-dependent phenotypes distinct from those previously reported in mouse models of amyotrophic lateral sclerosis Type 8. A loss of motor neurones in VapBP56S/+ and VapBP56S/P56S animals is indicated by a reduction in the number of large choline acetyl transferase-staining cells in the spinal cord. VapB-/- animals exhibit a relative increase in cytoplasmic TDP-43 levels compared with the nucleus, but no large protein aggregates. Concomitant with these spinal cord pathologies VapBP56S/+ , VapBP56S/P56S and VapB-/- animals exhibit age-dependent changes in paw placement and exerted pressures when traversing a CatWalk apparatus, consistent with a somatosensory dysfunction. Extramotor dysfunction is reported in half the cases of motor neurone disease, and this is the first indication of an associated sensory dysfunction in a rodent model of amyotrophic lateral sclerosis. Different rodent models may offer complementary experimental platforms with which to understand the human disease.

9.
J Neurochem ; 124(1): 26-35, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23083128

RESUMO

The Class IIa histone deacetylases (HDAC)4 and HDAC5 play a role in neuronal survival and behavioral adaptation in the CNS. Phosphorylation at 2/3 N-terminal sites promote their nuclear export. We investigated whether non-canonical signaling routes to Class IIa HDAC export exist because of their association with the co-repressor Silencing Mediator Of Retinoic And Thyroid Hormone Receptors (SMRT). We found that, while HDAC5 and HDAC4 mutants lacking their N-terminal phosphorylation sites (HDAC4(MUT), HDAC5(MUT)) are constitutively nuclear, co-expression with SMRT renders them exportable by signals that trigger SMRT export, such as synaptic activity, HDAC inhibition, and Brain Derived Neurotrophic Factor (BDNF) signaling. We found that SMRT's repression domain 3 (RD3) is critical for co-shuttling of HDAC5(MUT), consistent with the role for this domain in Class IIa HDAC association. In the context of BDNF signaling, we found that HDAC5(WT), which was more cytoplasmic than HDAC5(MUT), accumulated in the nucleus after BDNF treatment. However, co-expression of SMRT blocked BDNF-induced HDAC5(WT) import in a RD3-dependent manner. In effect, SMRT-mediated HDAC5(WT) export was opposing the BDNF-induced HDAC5 nuclear accumulation observed in SMRT's absence. Thus, SMRT's presence may render Class IIa HDACs exportable by a wider range of signals than those which simply promote direct phosphorylation.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Histona Desacetilases/metabolismo , Correpressor 2 de Receptor Nuclear/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Proteínas de Fluorescência Verde/genética , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/genética , Ácidos Hidroxâmicos/farmacologia , Mutação/genética , Neurônios , Correpressor 2 de Receptor Nuclear/genética , Fosforilação/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transfecção
10.
Blood ; 116(12): 2183-91, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20538801

RESUMO

Endothelial cells are reported to contain several distinct populations of regulated secretory organelles, including Weibel-Palade bodies (WPBs), the tissue plasminogen activator (tPA) organelle, and the type-2 chemokine-containing organelle. We show that the tPA and type-2 organelles in human endothelial cells represent a single compartment primarily responsible for unstimulated secretion of tPA or, in cells exposed to interleukin-1ß (IL-1ß), the cytokines IL-8, IL-6, monocyte chemoattractant protein-1 (MCP-1), and growth-regulated oncogene-α (GRO-α). This compartment was distinct from WPBs in that it lacked detectable von Willebrand factor, P-selectin, Rab27a, or CD63 immunoreactivity, displayed no time-dependent decrease in intragranule pH, underwent detectable unstimulated exocytosis, and was very poorly responsive to Ca(2+)-elevating secretagogues. WPBs could also contain tPA, and in IL-1ß-treated cells, IL-8, IL-6, MCP-1, and GRO-α, and were the primary source for histamine or ionomycin-stimulated secretion of these molecules. However, analysis of the storage efficiency of cytokines and tPA revealed that all were very poorly stored compared with von Willebrand factor. The nonmammalian, nonsecretory protein EGFP, when expressed in the secretory pathway, also entered WPBs and had a storage efficiency similar to tPA and the cytokines tested. Based on these data, we proposed a revised model for storage and secretion of cytokines and tPA.


Assuntos
Citocinas/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Ativador de Plasminogênio Tecidual/metabolismo , Compartimento Celular , Células Cultivadas , Humanos , Modelos Biológicos , Corpos de Weibel-Palade/metabolismo
11.
J Neurochem ; 117(2): 286-94, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21275991

RESUMO

VAMP/synaptobrevin associated proteins A and B (VAPA and VAPB), are type IV membrane proteins enriched on ER and Golgi membranes. Both VAPA and B interact with cytoplasmic lipid transport proteins and cytoskeletal elements to maintain the structure and composition of ER and Golgi membranes. Truncated forms of both proteins are present in some tissues but the functional significance of this is not clear. In rodents processing of VAPA occurs in most tissues, however, truncated forms of VAPB have only been reported in brain tissue. It is demonstrated here that the extent of VAPB processing in rat increases during postnatal development and that it is restricted to neurons. The C-terminal polypeptide generated by this cleavage reaction remains associated with cell membranes, but its subcellular distribution is distinct from the full-length protein. A mutant form of VAPB is associated with a familial form of neurodegenerative disease, amyotrophic lateral sclerosis type 8. The mutant protein, VAPB(P56S) , is resistant to truncation in primary neuronal cultures, although remains sensitive to some form of proteolysis when over-expressed in HEK293 cells. These data suggest that neuronal cells have a particular requirement for VAPB proteolysis and that reduced levels of processed polypeptides may contribute to the neurodegeneration associated with amyotrophic lateral sclerosis type 8.


Assuntos
Proteínas de Membrana/genética , Mutação/genética , Neurônios/fisiologia , Prolina/genética , Serina/genética , Substituição de Aminoácidos , Animais , Animais Recém-Nascidos , Transporte Biológico/genética , Morte Celular , Córtex Cerebral/citologia , Embrião de Mamíferos , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Microscopia Eletrônica de Transmissão/métodos , Peso Molecular , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , RNA Mensageiro , Ratos , Frações Subcelulares/metabolismo , Sinaptofisina/genética , Sinaptofisina/metabolismo , Transfecção/métodos , Resposta a Proteínas não Dobradas/genética
12.
Brain Commun ; 3(3): fcab152, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34396110

RESUMO

Primary hippocampal cell cultures are routinely used as an experimentally accessible model platform for the hippocampus and brain tissue in general. Containing multiple cell types including neurons, astrocytes and microglia in a state that can be readily analysed optically, biochemically and electrophysiologically, such cultures have been used in many in vitro studies. To what extent the in vivo environment is recapitulated in primary cultures is an on-going question. Here, we compare the transcriptomic profiles of primary hippocampal cell cultures and intact hippocampal tissue. In addition, by comparing profiles from wild type and the PrP 101LL transgenic model of prion disease, we also demonstrate that gene conservation is predominantly conserved across genetically altered lines.

13.
Cells ; 10(12)2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34943911

RESUMO

Synapses are particularly susceptible to the effects of advancing age, and mitochondria have long been implicated as organelles contributing to this compartmental vulnerability. Despite this, the mitochondrial molecular cascades promoting age-dependent synaptic demise remain to be elucidated. Here, we sought to examine how the synaptic mitochondrial proteome (including strongly mitochondrial associated proteins) was dynamically and temporally regulated throughout ageing to determine whether alterations in the expression of individual candidates can influence synaptic stability/morphology. Proteomic profiling of wild-type mouse cortical synaptic and non-synaptic mitochondria across the lifespan revealed significant age-dependent heterogeneity between mitochondrial subpopulations, with aged organelles exhibiting unique protein expression profiles. Recapitulation of aged synaptic mitochondrial protein expression at the Drosophila neuromuscular junction has the propensity to perturb the synaptic architecture, demonstrating that temporal regulation of the mitochondrial proteome may directly modulate the stability of the synapse in vivo.


Assuntos
Envelhecimento/genética , Proteínas Mitocondriais/genética , Distrofias Musculares/genética , Proteoma/genética , Sinapses/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Drosophila/genética , Drosophila/fisiologia , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Mitocôndrias/genética , Distrofias Musculares/patologia , Junção Neuromuscular/genética , Junção Neuromuscular/patologia , Neurônios/metabolismo
14.
Biomolecules ; 11(10)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34680132

RESUMO

Live imaging of neuromuscular junctions (NMJs) in situ has been constrained by the suitability of ligands for inert vital staining of motor nerve terminals. Here, we constructed several truncated derivatives of the tetanus toxin C-fragment (TetC) fused with Emerald Fluorescent Protein (emGFP). Four constructs, namely full length emGFP-TetC (emGFP-865:TetC) or truncations comprising amino acids 1066-1315 (emGFP-1066:TetC), 1093-1315 (emGFP-1093:TetC) and 1109-1315 (emGFP-1109:TetC), produced selective, high-contrast staining of motor nerve terminals in rodent or human muscle explants. Isometric tension and intracellular recordings of endplate potentials from mouse muscles indicated that neither full-length nor truncated emGFP-TetC constructs significantly impaired NMJ function or transmission. Motor nerve terminals stained with emGFP-TetC constructs were readily visualised in situ or in isolated preparations using fibre-optic confocal endomicroscopy (CEM). emGFP-TetC derivatives and CEM also visualised regenerated NMJs. Dual-waveband CEM imaging of preparations co-stained with fluorescent emGFP-TetC constructs and Alexa647-α-bungarotoxin resolved innervated from denervated NMJs in axotomized WldS mouse muscle and degenerating NMJs in transgenic SOD1G93A mouse muscle. Our findings highlight the region of the TetC fragment required for selective binding and visualisation of motor nerve terminals and show that fluorescent derivatives of TetC are suitable for in situ morphological and physiological characterisation of healthy, injured and diseased NMJs.


Assuntos
Microscopia Confocal , Junção Neuromuscular/diagnóstico por imagem , Toxina Tetânica/toxicidade , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Sítios de Ligação , Fluorescência , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Tecido Nervoso/efeitos dos fármacos , Tecido Nervoso/metabolismo , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/patologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos
15.
Hum Mol Genet ; 17(11): 1517-26, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18263603

RESUMO

A mis-sense point mutation in the human VAPB gene is associated with a familial form of motor neuron disease that has been classified as Amyotrophic Lateral Sclerosis type VIII. Affected individuals suffer from a spinal muscular atrophy (SMA), amyotrophic lateral sclerosis (ALS) or an atypical slowly progressing form of ALS. Mammals have two homologous VAP genes, vapA and vapB. VAPA and VAPB share 76% similar or identical amino acid residues; both are COOH-terminally anchored membrane proteins enriched on the endoplasmic reticulum. Several functions have been ascribed to VAP proteins including membrane trafficking, cytoskeleton association and membrane docking interactions for cytoplasmic factors. It is shown here that VAPA and VAPB are expressed in tissues throughout the body but at different levels, and that they are present in overlapping but distinct regions of the endoplasmic reticulum. The disease-associated mutation in VAPB, VAPB(P56S), lies within a highly conserved N-terminal region of the protein that shares extensive structural homology with the major sperm protein (MSP) from nematodes. The MSP domain of VAPA and VAPB is found to interact with the ER-localized transcription factor ATF6. Over expression of VAPB or VAPB(P56S) attenuates the activity of ATF6-regulated transcription and the mutant protein VAPB(P56S) appears to be a more potent inhibitor of ATF6 activity. These data indicate that VAP proteins interact directly with components of ER homeostatic and stress signalling systems and may therefore be parts of a previously unidentified regulatory pathway. The mis-function of such regulatory systems may contribute to the pathological mechanisms of degenerative motor neuron disease.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Fator 6 Ativador da Transcrição/antagonistas & inibidores , Fator 6 Ativador da Transcrição/genética , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Estrutura Terciária de Proteína/genética , Ratos , Distribuição Tecidual , Transcrição Gênica , Proteínas de Transporte Vesicular/genética
16.
Biochem Soc Trans ; 38(2): 443-4, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20298199

RESUMO

Synaptopathy is an increasingly popular term used to define key features of neurodegenerative and psychiatric disease. It implies that disruptions in synaptic structure and function are potentially the major determinant of such brain diseases. The Synaptopathies: Dysfunction of Synaptic Function Biochemical Society Focused Meeting brought together several invited speakers, supplemented with short communications from young scientists, who addressed this possibility. The talks spanned the full gamut of approaches that brought molecular, cellular, systems and whole-animal experimentation together to address how fundamental synaptic biology was increasingly informing on dysfunction in disease. The disease and models thereof discussed included Alzheimer's disease, prions, Huntington's disease, Parkinson's disease, schizophrenia and autism. The audience were asked to reflect on whether synaptopathy, although attractive and conceptually useful, provided a significant explanation as the cause of these major diseases. The breadth of the meeting reinforced the complexity of these brain diseases, supported the significance of synaptic dysfunction in disease, but left open the issue as to whether the prime cause of these disorders could be resolved as simple synaptic dysfunction. Thus, despite revealing a value of synaptopathy, further investigation will be required to reveal its balance in the cause and effect in each of the major brain diseases.


Assuntos
Congressos como Assunto , Transtornos Mentais/etiologia , Doenças Neurodegenerativas/etiologia , Transmissão Sináptica/fisiologia , Animais , Humanos , Transtornos Mentais/fisiopatologia , Doenças Neurodegenerativas/fisiopatologia , Sinapses/genética , Sinapses/metabolismo , Sinapses/patologia , Sinapses/fisiologia
17.
Blood ; 111(11): 5282-90, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18252862

RESUMO

Exocytosis of specialized endothelial cell secretory organelles, Weibel-Palade bodies (WPBs), is thought to play an important role in regulating hemostasis and intravascular inflammation. The major WPB core proteins are Von Willebrand factor (VWF) and its propolypeptide (Proregion), constituting more than 95% of the content. Although the composition of the WPBs can be fine-tuned to include cytokines and chemokines (eg, interleukin-8 [IL-8] and eotaxin-3), it is generally assumed that WPB exocytosis is inextricably associated with secretion of VWF. Here we show that WPBs can undergo a form of exocytosis during which VWF and Proregion are retained while smaller molecules, such as IL-8, are released. Imaging individual WPBs containing fluorescent cargo molecules revealed that during weak stimulation approximately 25% of fusion events result in a failure to release VWF or Proregion. The WPB membrane protein P-selectin was also retained; however, the membrane tetraspannin CD63 was released. Accumulation or exclusion of extracellular fluorescent dextran molecules ranging from 3 kDa to 2 mDa show that these events arise due to the formation of a fusion pore approximately 12 nm in diameter. The pore behaves as a molecular filter, allowing selective release of WPB core and membrane proteins. WPB exocytosis is not inextricably associated with secretion of VWF.


Assuntos
Células Endoteliais/metabolismo , Exocitose/fisiologia , Corpos de Weibel-Palade/metabolismo , Proteínas de Fluorescência Verde , Humanos , Immunoblotting , Imuno-Histoquímica , Interleucina-8/metabolismo , Microscopia Confocal , Estresse Mecânico , Fator de von Willebrand/metabolismo
18.
Pharmacol Res Perspect ; 7(4): e00495, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31249692

RESUMO

N-methyl-D-aspartate (NMDA) receptors are glutamate receptors with key roles in synaptic plasticity, due in part to their Mg2+ mediated voltage-dependence. A large number of genetic variants affecting NMDA receptor subunits have been found in people with a range of neurodevelopmental disorders, including GluN2AN615K (GRIN2AC1845A) in two unrelated individuals with severe epileptic encephalopathy. This missense variant substitutes a lysine in place of an asparagine known to be important for blockade by Mg2+ and other small molecule channel blockers. We therefore measured the impact of GluN2AN615K on a range of NMDA receptor channel blockers using two-electrode voltage clamp recordings made in Xenopus oocytes. We found that GluN2AN615K resulted in block by Mg2+ 1 mmol/L being greatly reduced (89% vs 8%), block by memantine 10 µmol/L (76% vs 27%) and amantadine 100 µmol/L (45% vs 17%) being substantially reduced, block by ketamine 10 µmol/L being modestly reduced (79% vs 73%) and block by dextromethorphan 10 µmol/L being enhanced (45% vs 55%). Coapplying Mg2+ with memantine or amantadine did not reduce the GluN2AN615K block seen with either small molecule. In addition, we measured single-channel conductance of GluN2AN615K-containing NMDA receptors in outside-out patches pulled from Xenopus oocytes, finding a 4-fold reduction in conductance (58 vs 15 pS). In conclusion, the GluN2AN615K variant is associated with substantial changes to important physiological and pharmacological properties of the NMDA receptor. Our findings are consistent with GluN2AN615K having a disease-causing role, and inform potential therapeutic strategies.


Assuntos
Substituição de Aminoácidos , Oócitos/citologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Amantadina/farmacologia , Animais , Dextrometorfano/farmacologia , Feminino , Humanos , Ketamina/farmacologia , Magnésio/farmacologia , Memantina/farmacologia , Mutação de Sentido Incorreto , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Receptores de N-Metil-D-Aspartato/genética , Xenopus
19.
Cells ; 8(6)2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31212843

RESUMO

Putative oogonial stem cells (OSCs) have been isolated by fluorescence-activated cell sorting (FACS) from adult human ovarian tissue using an antibody against DEAD-box helicase 4 (DDX4). DDX4 has been reported to be germ cell specific within the gonads and localised intracellularly. White et al. (2012) hypothesised that the C-terminus of DDX4 is localised on the surface of putative OSCs but is internalised during the process of oogenesis. This hypothesis is controversial since it is assumed that RNA helicases function intracellularly with no extracellular expression. To determine whether the C-terminus of DDX4 could be expressed on the cell surface, we generated a novel expression construct to express full-length DDX4 as a DsRed2 fusion protein with unique C- and N-terminal epitope tags. DDX4 and the C-terminal myc tag were detected at the cell surface by immunocytochemistry and FACS of non-permeabilised human embryonic kidney HEK 293T cells transfected with the DDX4 construct. DDX4 mRNA expression was detected in the DDX4-positive sorted cells by RT-PCR. This study clearly demonstrates that the C-terminus of DDX4 can be expressed on the cell surface despite its lack of a conventional membrane-targeting or secretory sequence. These results validate the use of antibody-based FACS to isolate DDX4-positive putative OSCs.


Assuntos
RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/metabolismo , Espaço Extracelular/metabolismo , Citometria de Fluxo/métodos , Imuno-Histoquímica/métodos , Anticorpos/farmacologia , Especificidade de Anticorpos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Epitopos/metabolismo , Feminino , Células HEK293 , Humanos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Ovário/metabolismo , Transporte Proteico/efeitos dos fármacos , Reprodutibilidade dos Testes
20.
Cell Rep ; 27(4): 1018-1026.e4, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31018120

RESUMO

Normal mammalian brain aging is characterized by the selective loss of discrete populations of dendritic spines and synapses, particularly affecting neuroanatomical regions such as the hippocampus. Although previous investigations have quantified this morphologically, the molecular pathways orchestrating preferential synaptic vulnerability remain to be elucidated. Using quantitative proteomics and healthy rhesus macaque and human patient brain regional tissues, we have comprehensively profiled the temporal expression of the synaptic proteome throughout the adult lifespan in differentially vulnerable brain regions. Comparative profiling of hippocampal (age vulnerable) and occipital cortex (age resistant) synapses revealed discrete and dynamic alterations in the synaptic proteome, which appear unequivocally conserved between species. The generation of these unique and important datasets will aid in delineating the molecular mechanisms underpinning primate brain aging, in addition to deciphering the regulatory biochemical cascades governing neurodegenerative disease pathogenesis.


Assuntos
Envelhecimento , Proteoma , Sinapses/metabolismo , Animais , Hipocampo/metabolismo , Humanos , Macaca mulatta , Lobo Occipital/metabolismo , Proteômica , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA