Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 17(9): 2127-2140, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-35985329

RESUMO

Mutations in the MAPT gene that encodes tau lead to frontotemporal dementia (FTD) with pathology evident in both cerebral neurons and glia. Human cerebral organoids (hCOs) from individuals harboring pathogenic tau mutations can reveal the earliest downstream effects on molecular pathways within a developmental context, generating interacting neurons and glia. We found that in hCOs carrying the V337M and R406W tau mutations, the cholesterol biosynthesis pathway in astrocytes was the top upregulated gene set compared with isogenic controls by single-cell RNA sequencing (scRNA-seq). The 15 upregulated genes included HMGCR, ACAT2, STARD4, LDLR, and SREBF2. This result was confirmed in a homozygous R406W mutant cell line by immunostaining and sterol measurements. Cholesterol abundance in the brain is tightly regulated by efflux and cholesterol biosynthetic enzyme levels in astrocytes, and dysregulation can cause aberrant phosphorylation of tau. Our findings suggest that cholesterol dyshomeostasis is an early event in the etiology of neurodegeneration caused by tau mutations.


Assuntos
Demência Frontotemporal , Proteínas tau , Colesterol , Demência Frontotemporal/genética , Humanos , Mutação/genética , Organoides/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
2.
Nat Commun ; 13(1): 4403, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35906223

RESUMO

Human brain organoids replicate much of the cellular diversity and developmental anatomy of the human brain. However, the physiology of neuronal circuits within organoids remains under-explored. With high-density CMOS microelectrode arrays and shank electrodes, we captured spontaneous extracellular activity from brain organoids derived from human induced pluripotent stem cells. We inferred functional connectivity from spike timing, revealing a large number of weak connections within a skeleton of significantly fewer strong connections. A benzodiazepine increased the uniformity of firing patterns and decreased the relative fraction of weakly connected edges. Our analysis of the local field potential demonstrate that brain organoids contain neuronal assemblies of sufficient size and functional connectivity to co-activate and generate field potentials from their collective transmembrane currents that phase-lock to spiking activity. These results point to the potential of brain organoids for the study of neuropsychiatric diseases, drug action, and the effects of external stimuli upon neuronal networks.


Assuntos
Células-Tronco Pluripotentes Induzidas , Organoides , Encéfalo/fisiologia , Humanos , Microeletrodos , Neurônios/fisiologia
3.
Sci Rep ; 11(1): 14733, 2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34282275

RESUMO

We developed a method to non-invasively detect synaptic relationships among neurons from in vitro networks. Our method uses microelectrode arrays on which neurons are cultured and from which propagation of extracellular action potentials (eAPs) in single axons are recorded at multiple electrodes. Detecting eAP propagation bypasses ambiguity introduced by spike sorting. Our methods identify short latency spiking relationships between neurons with properties expected of synaptically coupled neurons, namely they were recapitulated by direct stimulation and were sensitive to changing the number of active synaptic sites. Our methods enabled us to assemble a functional subset of neuronal connectivity in our cultures.


Assuntos
Potenciais de Ação/fisiologia , Eletrofisiologia/métodos , Neurônios/fisiologia , Algoritmos , Animais , Animais Recém-Nascidos , Células Cultivadas , Espaço Extracelular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microeletrodos , Neurônios/citologia , Sinapses/fisiologia , Potenciais Sinápticos/fisiologia
4.
Nature ; 580(7803): 381-385, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32296178

RESUMO

The spread of protein aggregates during disease progression is a common theme underlying many neurodegenerative diseases. The microtubule-associated protein tau has a central role in the pathogenesis of several forms of dementia known as tauopathies-including Alzheimer's disease, frontotemporal dementia and chronic traumatic encephalopathy1. Progression of these diseases is characterized by the sequential spread and deposition of protein aggregates in a predictable pattern that correlates with clinical severity2. This observation and complementary experimental studies3,4 have suggested that tau can spread in a prion-like manner, by passing to naive cells in which it templates misfolding and aggregation. However, although the propagation of tau has been extensively studied, the underlying cellular mechanisms remain poorly understood. Here we show that the low-density lipoprotein receptor-related protein 1 (LRP1) controls the endocytosis of tau and its subsequent spread. Knockdown of LRP1 significantly reduced tau uptake in H4 neuroglioma cells and in induced pluripotent stem cell-derived neurons. The interaction between tau and LRP1 is mediated by lysine residues in the microtubule-binding repeat region of tau. Furthermore, downregulation of LRP1 in an in vivo mouse model of tau spread was found to effectively reduce the propagation of tau between neurons. Our results identify LRP1 as a key regulator of tau spread in the brain, and therefore a potential target for the treatment of diseases that involve tau spread and aggregation.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteínas tau/metabolismo , Animais , Linhagem Celular , Endocitose , Feminino , Humanos , Ligantes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Camundongos , Neurônios/metabolismo
5.
Nat Neurosci ; 23(2): 167-171, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31873194

RESUMO

Sex is a key modifier of neurological disease outcomes. Microglia are implicated in neurological diseases and modulated by microRNAs, but it is unknown whether microglial microRNAs have sex-specific influences on disease. We show in mice that microglial microRNA expression differs in males and females and that loss of microRNAs leads to sex-specific changes in the microglial transcriptome and tau pathology. These findings suggest that microglial microRNAs influence tau pathogenesis in a sex-specific manner.


Assuntos
Encéfalo/patologia , MicroRNAs/metabolismo , Microglia/metabolismo , Caracteres Sexuais , Tauopatias/patologia , Animais , Encéfalo/metabolismo , Feminino , Masculino , Camundongos , Microglia/patologia , Tauopatias/metabolismo , Transcriptoma , Proteínas tau/metabolismo
6.
Sci Transl Med ; 11(485)2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30918111

RESUMO

Tau inclusions are a shared feature of many neurodegenerative diseases, among them frontotemporal dementia caused by tau mutations. Treatment approaches for these conditions include targeting posttranslational modifications of tau proteins, maintaining a steady-state amount of tau, and preventing its tendency to aggregate. We discovered a new regulatory pathway for tau degradation that operates through the farnesylated protein, Rhes, a GTPase in the Ras family. Here, we show that treatment with the farnesyltransferase inhibitor lonafarnib reduced Rhes and decreased brain atrophy, tau inclusions, tau sumoylation, and tau ubiquitination in the rTg4510 mouse model of tauopathy. In addition, lonafarnib treatment attenuated behavioral abnormalities in rTg4510 mice and reduced microgliosis in mouse brain. Direct reduction of Rhes in the rTg4510 mouse by siRNA reproduced the results observed with lonafarnib treatment. The mechanism of lonafarnib action mediated by Rhes to reduce tau pathology was shown to operate through activation of lysosomes. We finally showed in mouse brain and in human induced pluripotent stem cell-derived neurons a normal developmental increase in Rhes that was initially suppressed by tau mutations. The known safety of lonafarnib revealed in human clinical trials for cancer suggests that this drug could be repurposed for treating tauopathies.


Assuntos
Farnesiltranstransferase/antagonistas & inibidores , Tauopatias/tratamento farmacológico , Tauopatias/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Piperidinas/farmacologia , Proteólise/efeitos dos fármacos , Piridinas/farmacologia , RNA Interferente Pequeno/genética , Tauopatias/patologia , Pesquisa Translacional Biomédica , Proteínas tau/genética , Proteínas tau/metabolismo
7.
Cell Rep ; 20(5): 1136-1147, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768198

RESUMO

Peripheral nerve regeneration after injury requires a broad program of transcriptional changes. We investigated the basis for the enhanced nerve regenerative capacity of the CAST/Ei mouse strain relative to C57BL/6 mice. RNA sequencing of dorsal root ganglia (DRG) showed a CAST/Ei-specific upregulation of Ascl1 after injury. Ascl1 overexpression in DRG neurons of C57BL/6 mice enhanced their neurite outgrowth. Ascl1 is regulated by miR-7048-3p, which is downregulated in CAST/Ei mice. Inhibition of miR-7048-3p enhances neurite outgrowth. Following injury, CAST/Ei neurons largely retained their mature neuronal profile as determined by single-cell RNA- seq, whereas the C57BL/6 neurons acquired an immature profile. These findings suggest that one facet of the enhanced regenerative phenotype is preservation of neuronal identity in response to injury.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regeneração Nervosa , Neuritos/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Neuritos/patologia , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/patologia
8.
J Cell Biol ; 216(10): 3355-3368, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28768686

RESUMO

During apoptosis, executioner caspase activity has been considered a point of no return. However, recent studies show that cells can survive caspase activation following transient apoptotic stimuli, a process called anastasis. To identify a molecular signature, we performed whole-transcriptome RNA sequencing of untreated, apoptotic, and recovering HeLa cells. We found that anastasis is an active, two-stage program. During the early stage, cells transition from growth-arrested to growing. In the late stage, HeLa cells change from proliferating to migratory. Recovering cells also exhibited prolonged elevation of proangiogenic factors. Strikingly, some early-recovery mRNAs, including Snail, were elevated first during apoptosis, implying that dying cells poise to recover, even while under apoptotic stress. Snail was also required for recovery. This study reveals similarities in the anastasis genes, pathways, and cell behaviors to those activated in wound healing and identifies a repertoire of potential targets for therapeutic manipulation.


Assuntos
Apoptose/genética , Movimento Celular/genética , Proliferação de Células/genética , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Cicatrização/genética , Células HeLa , Humanos
9.
PLoS Biol ; 15(7): e2002183, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28683104

RESUMO

Nonmembrane-bound organelles that behave like liquid droplets are widespread among eukaryotic cells. Their dysregulation appears to be a critical step in several neurodegenerative conditions. Here, we report that tau protein, the primary constituent of Alzheimer neurofibrillary tangles, can form liquid droplets and therefore has the necessary biophysical properties to undergo liquid-liquid phase separation (LLPS) in cells. Consonant with the factors that induce LLPS, tau is an intrinsically disordered protein that complexes with RNA to form droplets. Uniquely, the pool of RNAs to which tau binds in living cells are tRNAs. This phase state of tau is held in an approximately 1:1 charge balance across the protein and the nucleic acid constituents, and can thus be maximal at different RNA:tau mass ratios, depending on the biopolymer constituents involved. This feature is characteristic of complex coacervation. We furthermore show that the LLPS process is directly and sensitively tuned by salt concentration and temperature, implying it is modulated by both electrostatic interactions between the involved protein and nucleic acid constituents, as well as net changes in entropy. Despite the high protein concentration within the complex coacervate phase, tau is locally freely tumbling and capable of diffusing through the droplet interior. In fact, tau in the condensed phase state does not reveal any immediate changes in local protein packing, local conformations and local protein dynamics from that of tau in the dilute solution state. In contrast, the population of aggregation-prone tau as induced by the complexation with heparin is accompanied by large changes in local tau conformations and irreversible aggregation. However, prolonged residency within the droplet state eventually results in the emergence of detectable ß-sheet structures according to thioflavin-T assay. These findings suggest that the droplet state can incubate tau and predispose the protein toward the formation of insoluble fibrils.


Assuntos
Agregação Patológica de Proteínas , RNA/metabolismo , Proteínas tau/metabolismo , Células Cultivadas , Humanos , Temperatura
10.
Development ; 144(10): 1787-1797, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28432214

RESUMO

Here we present a parallel study of mRNA and microRNA expression during oral siphon (OS) regeneration in Ciona robusta, and the derived network of their interactions. In the process of identifying 248 mRNAs and 15 microRNAs as differentially expressed, we also identified 57 novel microRNAs, several of which are among the most highly differentially expressed. Analysis of functional categories identified enriched transcripts related to stress responses and apoptosis at the wound healing stage, signaling pathways including Wnt and TGFß during early regrowth, and negative regulation of extracellular proteases in late stage regeneration. Consistent with the expression results, we found that inhibition of TGFß signaling blocked OS regeneration. A correlation network was subsequently inferred for all predicted microRNA-mRNA target pairs expressed during regeneration. Network-based clustering associated transcripts into 22 non-overlapping groups, the functional analysis of which showed enrichment of stress response, signaling pathway and extracellular protease categories that could be related to specific microRNAs. Predicted targets of the miR-9 cluster suggest a role in regulating differentiation and the proliferative state of neural progenitors through regulation of the cytoskeleton and cell cycle.


Assuntos
Ciona intestinalis/fisiologia , Redes Reguladoras de Genes/fisiologia , MicroRNAs/genética , Boca/fisiologia , RNA Mensageiro/genética , Regeneração/genética , Cicatrização/genética , Animais , Ciona intestinalis/genética , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Boca/citologia
11.
Cell ; 165(2): 410-20, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-27020754

RESUMO

Under defined differentiation conditions, human embryonic stem cells (hESCs) can be directed toward a mesendoderm (ME) or neuroectoderm (NE) fate, the first decision during hESC differentiation. Coupled with lineage-specific G1 lengthening, a divergent ciliation pattern emerged within the first 24 hr of induced lineage specification, and these changes heralded a neuroectoderm decision before any neural precursor markers were expressed. By day 2, increased ciliation in NE precursors induced autophagy that resulted in the inactivation of Nrf2 and thereby relieved transcriptional activation of OCT4 and NANOG. Nrf2 binds directly to upstream regions of these pluripotency genes to promote their expression and repress NE derivation. Nrf2 suppression was sufficient to rescue poorly neurogenic iPSC lines. Only after these events had been initiated did neural precursor markers get expressed at day 4. Thus, we have identified a primary cilium-autophagy-Nrf2 (PAN) control axis coupled to cell-cycle progression that directs hESCs toward NE.


Assuntos
Autofagia , Cílios/metabolismo , Células-Tronco Embrionárias/citologia , Fator 2 Relacionado a NF-E2/metabolismo , Ciclo Celular , Proteínas de Homeodomínio/genética , Humanos , Proteína Homeobox Nanog , Placa Neural/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco Pluripotentes/metabolismo
12.
Hum Mol Genet ; 25(7): 1294-306, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26755828

RESUMO

Williams syndrome (WS) is a neurodevelopmental disorder caused by a genomic deletion of ∼28 genes that results in a cognitive and behavioral profile marked by overall intellectual impairment with relative strength in expressive language and hypersocial behavior. Advancements in protocols for neuron differentiation from induced pluripotent stem cells allowed us to elucidate the molecular circuitry underpinning the ontogeny of WS. In patient-derived stem cells and neurons, we determined the expression profile of the Williams-Beuren syndrome critical region-deleted genes and the genome-wide transcriptional consequences of the hemizygous genomic microdeletion at chromosome 7q11.23. Derived neurons displayed disease-relevant hallmarks and indicated novel aberrant pathways in WS neurons including over-activated Wnt signaling accompanying an incomplete neurogenic commitment. We show that haploinsufficiency of the ATP-dependent chromatin remodeler, BAZ1B, which is deleted in WS, significantly contributes to this differentiation defect. Chromatin-immunoprecipitation (ChIP-seq) revealed BAZ1B target gene functions are enriched for neurogenesis, neuron differentiation and disease-relevant phenotypes. BAZ1B haploinsufficiency caused widespread gene expression changes in neural progenitor cells, and together with BAZ1B ChIP-seq target genes, explained 42% of the transcriptional dysregulation in WS neurons. BAZ1B contributes to regulating the balance between neural precursor self-renewal and differentiation and the differentiation defect caused by BAZ1B haploinsufficiency can be rescued by mitigating over-active Wnt signaling in neural stem cells. Altogether, these results reveal a pivotal role for BAZ1B in neurodevelopment and implicate its haploinsufficiency as a likely contributor to the neurological phenotypes in WS.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Haploinsuficiência , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurogênese , Neurônios/metabolismo , Fatores de Transcrição/genética , Síndrome de Williams/metabolismo , Deleção Cromossômica , Cromossomos Humanos Par 7 , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Transdução de Sinais , Transcrição Gênica , Transcriptoma , Síndrome de Williams/genética , Síndrome de Williams/fisiopatologia
13.
Nature ; 515(7527): 414-8, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25132547

RESUMO

Dysregulated neurodevelopment with altered structural and functional connectivity is believed to underlie many neuropsychiatric disorders, and 'a disease of synapses' is the major hypothesis for the biological basis of schizophrenia. Although this hypothesis has gained indirect support from human post-mortem brain analyses and genetic studies, little is known about the pathophysiology of synapses in patient neurons and how susceptibility genes for mental disorders could lead to synaptic deficits in humans. Genetics of most psychiatric disorders are extremely complex due to multiple susceptibility variants with low penetrance and variable phenotypes. Rare, multiply affected, large families in which a single genetic locus is probably responsible for conferring susceptibility have proven invaluable for the study of complex disorders. Here we generated induced pluripotent stem (iPS) cells from four members of a family in which a frameshift mutation of disrupted in schizophrenia 1 (DISC1) co-segregated with major psychiatric disorders and we further produced different isogenic iPS cell lines via gene editing. We showed that mutant DISC1 causes synaptic vesicle release deficits in iPS-cell-derived forebrain neurons. Mutant DISC1 depletes wild-type DISC1 protein and, furthermore, dysregulates expression of many genes related to synapses and psychiatric disorders in human forebrain neurons. Our study reveals that a psychiatric disorder relevant mutation causes synapse deficits and transcriptional dysregulation in human neurons and our findings provide new insight into the molecular and synaptic etiopathology of psychiatric disorders.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Transtornos Mentais/patologia , Sinapses/patologia , Animais , Diferenciação Celular , Fibroblastos , Glutamina/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Transtornos Mentais/genética , Transtornos Mentais/metabolismo , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Linhagem , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Ligação Proteica , Sinapses/metabolismo , Transcriptoma
14.
Neuron ; 81(6): 1255-1262, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24583023

RESUMO

Major nonprimate-primate differences in cortico-genesis include the dimensions, precursor lineages, and developmental timing of the germinal zones (GZs). microRNAs (miRNAs) of laser-dissected GZ compartments and cortical plate (CP) from embryonic E80 macaque visual cortex were deep sequenced. The CP and the GZ including ventricular zone (VZ) and outer and inner subcompartments of the outer subventricular zone (OSVZ) in area 17 displayed unique miRNA profiles. miRNAs present in primate, but absent in rodent, contributed disproportionately to the differential expression between GZ subregions. Prominent among the validated targets of these miRNAs were cell-cycle and neurogenesis regulators. Coevolution between the emergent miRNAs and their targets suggested that novel miRNAs became integrated into ancient gene circuitry to exert additional control over proliferation. We conclude that multiple cell-cycle regulatory events contribute to the emergence of primate-specific cortical features, including the OSVZ, generated enlarged supragranular layers, largely responsible for the increased primate cortex computational abilities.


Assuntos
Ciclo Celular/genética , Regulação da Expressão Gênica , Macaca/genética , MicroRNAs/genética , Neurogênese/genética , Neurônios/citologia , Córtex Visual/citologia , Animais , Ciclo Celular/fisiologia , Evolução Molecular , Feminino , Neurogênese/fisiologia , Neurônios/metabolismo , Córtex Visual/metabolismo
15.
J Alzheimers Dis ; 39(2): 301-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24150109

RESUMO

We tested the hypothesis that mutant tau proteins that cause neurodegeneration and dementia differentially alter kinesin translocation along microtubules (MTs) relative to normal tau in vitro. We employed complementary in vitro motility assays using purified recombinant kinesin, purified recombinant tau, and purified bovine brain α:ß tubulin to isolate interactions among these components without any contribution by cellular regulatory mechanisms. We found that kinesin translocates slower along MTs assembled by any of three independent tau mutants (4-repeat P301L tau, 4-repeat ΔN296 tau, and 4-repeat R406W tau) relative to its translocation rate along MTs assembled by normal, 4-repeat wild type (WT) tau. Moreover, the R406W mutation exhibited isoform specific effects; while kinesin translocation along 4-repeat R406W tau assembled MTs is slower than along MTs assembled by 4-repeat WT tau, the R406W mutation had no effect in the 3-repeat tau context. These data provide strong support for the notion that aberrant modulation of kinesin translocation is a component of tau-mediated neuronal cell death and dementia. Finally, we showed that assembling MTs with taxol before coating them with mutant tau obscured effects of the mutant tau that were readily apparent using more physiologically relevant MTs assembled with tau alone, raising important issues regarding the use of taxol as an experimental reagent and novel insights into therapeutic mechanisms of taxol action.


Assuntos
Encéfalo/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Mutação , Proteínas tau/genética , Proteínas tau/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Bovinos , Humanos , Microtúbulos/efeitos dos fármacos , Doenças Neurodegenerativas/genética , Paclitaxel/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pontos Quânticos , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacologia
16.
PLoS One ; 8(9): e76409, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086739

RESUMO

Over two dozen mutations in the gene encoding the microtubule associated protein tau cause a variety of neurodegenerative dementias known as tauopathies, including frontotemporal dementia (FTD), PSP, CBD and Pick's disease. The vast majority of these mutations map to the C-terminal region of tau possessing microtubule assembly and microtubule dynamics regulatory activities as well as the ability to promote pathological tau aggregation. Here, we describe a novel and non-conservative tau mutation (G55R) mapping to an alternatively spliced exon encoding part of the N-terminal region of the protein in a patient with the behavioral variant of FTD. Although less well understood than the C-terminal region of tau, the N-terminal region can influence both MT mediated effects as well as tau aggregation. The mutation changes an uncharged glycine to a basic arginine in the midst of a highly conserved and very acidic region. In vitro, 4-repeat G55R tau nucleates microtubule assembly more effectively than wild-type 4-repeat tau; surprisingly, this effect is tau isoform specific and is not observed in a 3-repeat G55R tau versus 3-repeat wild-type tau comparison. In contrast, the G55R mutation has no effect upon the abilities of tau to regulate MT growing and shortening dynamics or to aggregate. Additionally, the mutation has no effect upon kinesin translocation in a microtubule gliding assay. Together, (i) we have identified a novel tau mutation mapping to a mutation deficient region of the protein in a bvFTD patient, and (ii) the G55R mutation affects the ability of tau to nucleate microtubule assembly in vitro in a 4-repeat tau isoform specific manner. This altered capability could markedly affect in vivo microtubule function and neuronal cell biology. We consider G55R to be a candidate mutation for bvFTD since additional criteria required to establish causality are not yet available for assessment.


Assuntos
Demência Frontotemporal/genética , Microtúbulos/metabolismo , Mutação de Sentido Incorreto/genética , Proteínas tau/genética , Proteínas tau/metabolismo , Sequência de Aminoácidos , Feminino , Demência Frontotemporal/patologia , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Polônia , Alinhamento de Sequência , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA