Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Acta Neuropathol ; 146(1): 97-119, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37120788

RESUMO

Heterozygous mutations in the granulin (GRN) gene, resulting in the haploinsufficiency of the progranulin (PGRN) protein, is a leading cause of frontotemporal lobar degeneration (FTLD). Complete loss of the PGRN protein causes neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disorder. Polymorphisms in the GRN gene have also been associated with several other neurodegenerative diseases, including Alzheimer's disease (AD), and Parkinson's disease (PD). PGRN deficiency has been shown to cause myelination defects previously, but how PGRN regulates myelination is unknown. Here, we report that PGRN deficiency leads to a sex-dependent myelination defect with male mice showing more severe demyelination in response to cuprizone treatment. This is accompanied by exacerbated microglial proliferation and activation in the male PGRN-deficient mice. Interestingly, both male and female PGRN-deficient mice show sustained microglial activation after cuprizone removal and a defect in remyelination. Specific ablation of PGRN in microglia results in similar sex-dependent phenotypes, confirming a microglial function of PGRN. Lipid droplets accumulate in microglia specifically in male PGRN-deficient mice. RNA-seq analysis and mitochondrial function assays reveal key differences in oxidative phosphorylation in male versus female microglia under PGRN deficiency. A significant decrease in myelination and accumulation of myelin debris and lipid droplets in microglia were found in the corpus callosum regions of FTLD patients with GRN mutations. Taken together, our data support that PGRN deficiency leads to sex-dependent alterations in microglia with subsequent myelination defects.


Assuntos
Doenças Desmielinizantes , Demência Frontotemporal , Degeneração Lobar Frontotemporal , Animais , Feminino , Masculino , Camundongos , Cuprizona/metabolismo , Demência Frontotemporal/metabolismo , Degeneração Lobar Frontotemporal/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lisossomos/metabolismo , Microglia/metabolismo , Progranulinas/genética
2.
Neuropathol Appl Neurobiol ; 48(2): e12778, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34823271

RESUMO

AIMS: Accumulating evidence suggests that patients with frontotemporal lobar degeneration (FTLD) can have pathologic accumulation of multiple proteins, including tau and TDP-43. This study aimed to determine the frequency and characteristics of concurrent tau pathology in FTLD with TDP-43 pathology (FTLD-TDP). METHODS: The study included 146 autopsy-confirmed cases of FTLD-TDP and 55 cases of FTLD-TDP with motor neuron disease (FTLD-MND). Sections from the basal forebrain were screened for tau pathology with phosphorylated-tau immunohistochemistry. For cases with tau pathology on the screening section, additional brain sections were studied to establish a diagnosis. Genetic analysis of C9orf72, GRN and MAPT was performed on select cases. RESULTS: We found 72 cases (36%) with primary age-related tauopathy (PART), 85 (42%) with ageing-related tau astrogliopathy (ARTAG), 45 (22%) with argyrophilic grain disease (AGD) and 2 cases (1%) with corticobasal degeneration (CBD). Patients with ARTAG or AGD were significantly older than those without these comorbidities. One of the patients with FTLD-TDP and CBD had C9orf72 mutation and relatively mild tau pathology, consistent with incidental CBD. CONCLUSION: The coexistence of TDP-43 and tau pathologies was relatively common, particularly PART and ARTAG. Although rare, patients with FTLD can have multiple neurodegenerative proteinopathies. The absence of TDP-43-positive astrocytic plaques may suggest that CBD and FTLD-TDP were independent disease processes in the two patients with both tau and TDP-43 pathologies. It remains to be determined if mixed cases represent a unique disease process or two concurrent disease processes in an individual.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/complicações , Neurônios/metabolismo , Tauopatias/complicações , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Encéfalo/patologia , Feminino , Degeneração Lobar Frontotemporal/metabolismo , Degeneração Lobar Frontotemporal/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/patologia , Tauopatias/metabolismo , Tauopatias/patologia
3.
Mov Disord ; 37(9): 1943-1948, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35810449

RESUMO

BACKGROUND: Loss-of-function mutations in GRN are a cause of familial frontotemporal dementia, and common variants within the gene have been associated with an increased risk of developing Alzheimer's disease and Parkinson's disease. Although TDP-43-positive inclusions are characteristic of GRN-related neurodegeneration, Lewy body copathology has also been observed in many GRN mutation carriers. OBJECTIVE: The objective of this study was to assess a Lewy body dementia (LBD) case-control cohort for pathogenic variants in GRN and to test whether there is an enrichment of damaging mutations among patients with LBD. METHODS: We analyzed whole-genome sequencing data generated for 2591 European-ancestry LBD cases and 4032 neurologically healthy control subjects to identify disease-causing mutations in GRN. RESULTS: We identified six heterozygous exonic GRN mutations in seven study participants (cases: n = 6; control subjects: n = 1). Each variant was predicted to be pathogenic or likely pathogenic. We found significant enrichment of GRN loss-of-function mutations in patients with LBD compared with control subjects (Optimized Sequence Kernel Association Test P = 0.0162). Immunohistochemistry in three definite LBD cases demonstrated Lewy body pathology and TDP-43-positive neuronal inclusions. CONCLUSIONS: Our findings suggest that deleterious GRN mutations are a rare cause of familial LBD. © 2022 International Parkinson Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Assuntos
Doença por Corpos de Lewy , Progranulinas , Proteínas de Ligação a DNA/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Doença por Corpos de Lewy/genética , Mutação/genética , Progranulinas/genética
4.
Neurol Sci ; 43(1): 319-326, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33999292

RESUMO

The MEF2C gene encodes a transcription factor known to play a crucial role in molecular pathways affecting neuronal development. MEF2C mutations were described as a genetic cause of developmental disease (MRD20), and several reports sustain its involvement in dementia-related conditions, such as Alzheimer's disease and amyotrophic lateral sclerosis. These pathologies and frontotemporal degeneration (FTLD) are thought to share common physiopathological pathways. In this exploratory study, we searched for alterations in the DNA sequence of exons and boundaries, including 5'- and 3'-untranslated regions (5'UTR, 3'UTR), of MEF2C gene in 11 patients with clinical phenotypes related with MRD20 or FTLD. We identified a heterozygous deletion of 13 nucleotides in the 5'UTR region of a 69 years old FTLD patient. This alteration was absent in 200 healthy controls, suggesting a contribution to this patient's disease phenotype. In silico analysis of the mutated sequence indicated changes in mRNA secondary structure and stability, thus potentially affecting MEF2C protein levels. Furthermore, in vitro functional analysis of this mutation revealed that the presence of this deletion abolished the transcriptional activity of the gene in human embryonic cells and rat brain neurons, probably by modifying MEF2C expression. Altogether, our results provide evidence for the involvement of MEF2C in FTLD manifesting with seizures.


Assuntos
Degeneração Lobar Frontotemporal , Fatores de Transcrição MEF2 , Idoso , Degeneração Lobar Frontotemporal/genética , Humanos , Fatores de Transcrição MEF2/genética , Mutação
5.
Neurobiol Dis ; 154: 105360, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33812000

RESUMO

Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are fatal neurodegenerative disorders that are thought to exist on a clinical and pathological spectrum. FTD and ALS are linked by shared genetic causes (e.g. C9orf72 hexanucleotide repeat expansions) and neuropathology, such as inclusions of ubiquitinated, misfolded proteins (e.g. TAR DNA-binding protein 43; TDP-43) in the CNS. Furthermore, some genes that cause FTD or ALS when mutated encode proteins that localize to the lysosome or modulate endosome-lysosome function, including lysosomal fusion, cargo trafficking, lysosomal acidification, autophagy, or TFEB activity. In this review, we summarize evidence that lysosomal dysfunction, caused by genetic mutations (e.g. C9orf72, GRN, MAPT, TMEM106B) or toxic-gain of function (e.g. aggregation of TDP-43 or tau), is an important pathogenic disease mechanism in FTD and ALS. Further studies into the normal function of many of these proteins are required and will help uncover the mechanisms that cause lysosomal dysfunction in FTD and ALS. Mutations or polymorphisms in genes that encode proteins important for endosome-lysosome function also occur in other age-dependent neurodegenerative diseases, including Alzheimer's (e.g. APOE, PSEN1, APP) and Parkinson's (e.g. GBA, LRRK2, ATP13A2) disease. A more complete understanding of the common and unique features of lysosome dysfunction across the spectrum of neurodegeneration will help guide the development of therapies for these devastating diseases.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Demência Frontotemporal/metabolismo , Demência Frontotemporal/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Esclerose Lateral Amiotrófica/genética , Animais , Autofagia/fisiologia , Demência Frontotemporal/genética , Humanos , Lisossomos/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia
6.
Neuropathol Appl Neurobiol ; 47(7): 1033-1049, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33971027

RESUMO

AIM: The heterogeneity in the distribution and morphological features of TAR DNA-binding protein-43 (TDP-43) pathology in the brains of frontotemporal lobar degeneration (FTLD-TDP) patients and their different clinical manifestations suggest that distinct pathological TDP-43 strains could play a role in this heterogeneity between different FTLD-TDP subtypes (A-E). Our aim was to evaluate the existence of distinct TDP-43 strains in the brains of different FTLD-TDP subtypes and characterise their specific seeding properties in vitro and in vivo. METHODS AND RESULTS: We used an inducible stable cell line expressing a mutant cytoplasmic TDP-43 (iGFP-NLSm) to evaluate the seeding properties of distinct pathological TDP-43 strains. Brain-derived TDP-43 protein extracts from FTLD-TDP types A (n = 6) and B (n = 3) cases induced the formation of round/spherical phosphorylated TDP-43 aggregates that morphologically differed from the linear and wavy wisps and bigger heterogeneous filamentous (skein-like) aggregates induced by type E (n = 3) cases. These morphological differences correlated with distinct biochemical banding patterns of sarkosyl-insoluble TDP-43 protein recovered from the transduced cells. Moreover, brain-derived TDP-43 extracts from type E cases showed higher susceptibility to PK digestion of full-length TDP-43 and the most abundant C-terminal fragments that characterise type E extracts. Finally, we showed that intracerebral injections of different TDP-43 strains induced a distinctive morphological and subcellular distribution of TDP-43 pathology and different spreading patterns in the brains of CamKIIa-hTDP-43NLSm Tg mice. CONCLUSIONS: We show the existence of distinct TDP-43 strains in the brain of different FTLD-TDP subtypes with distinctive seeding and spreading properties in the brains of experimental animal models.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação a DNA/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Demência Frontotemporal/metabolismo , Demência Frontotemporal/patologia , Degeneração Lobar Frontotemporal/patologia , Humanos , Camundongos Transgênicos
7.
J Neurosci Res ; 98(2): 262-283, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31549445

RESUMO

The finding of an effective cure or treatment for neurodegenerative diseases is one of the biggest challenges for this century. Although these diseases show different clinical manifestations, the presence of toxic protein aggregates in the brain of patients is a common feature to all of them, suggesting a loss of protein homeostasis. Aging, the primary risk factor for the majority of neurodegenerative disorders, is linked to the impairment of degradative compartments such as lysosomes and autophagosomes. Besides, many genetic factors for Alzheimer's disease, Parkinson's disease, or frontotemporal dementia, as examples of frequent neurodegenerative diseases, are causative of endo-lysosomal and autophagosomal dysfunctions. There is scientific evidence suggesting that neurons can counteract the accumulation of undegraded cellular material by the secretion of extracellular vesicles (EVs), which are vesicles with a size ranging from 50 to 100 nm generated in a type of endosomal compartment named multivesicular body. EVs play a crucial role in removing cellular waste, promoting protein aggregation, and spreading toxic protein aggregates in the brain of patients. In this review, the interplay between the impairment of degradative compartments, the secretion of EVs, and their pathological/beneficial role in neurodegeneration is described.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Vesículas Extracelulares/metabolismo , Degeneração Neural/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteostase/fisiologia , Envelhecimento/patologia , Animais , Encéfalo/patologia , Vesículas Extracelulares/patologia , Humanos , Degeneração Neural/patologia , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia
8.
J Neuropsychiatry Clin Neurosci ; 32(3): 274-279, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31687868

RESUMO

OBJECTIVE: The experience of embarrassment signals violations in social norms, and impairment in this social emotion may underlie much of the social dysfunction in behavioral variant frontotemporal dementia (bvFTD). The authors investigated whether impaired self-awareness of embarrassment may distinguish patients with bvFTD early in the course of disease from healthy control subjects (HCs). METHODS: Self-reported embarrassment was examined among 18 patients with early bvFTD and 23 HCs by using the 36-item Embarrassability Scale, which includes items of situations eliciting embarrassment for oneself ("self-embarrassment") and embarrassment for others ("vicarious embarrassment"). The two study groups were also compared with the Social Norms Questionnaire (SNQ). The analyses included correlations of SNQ results (total score, violations or "break" errors, and overendorsement of social rules or "overadhere" errors) with Embarrassability Scale scores. RESULTS: Patients with bvFTD did not differ from HCs on total or self-embarrassment scores but did have significantly higher vicarious embarrassment scores. Unlike in the HC group, reports of vicarious embarrassment did not differ from reports of self-embarrassment among patients in the bvFTD group. The Embarrassability Score further correlated with overadherence to norms on the SNQ. CONCLUSIONS: In the presence of social dysfunction and emotional blunting, these findings suggest that patients with bvFTD rely on their own perspective for a rule-based application of social norms in reporting vicarious embarrassment. The assessment of reports of embarrassment for others may indicate an early and previously unrecognized clinical measure for detecting bvFTD.


Assuntos
Constrangimento , Demência Frontotemporal/fisiopatologia , Autoimagem , Normas Sociais , Percepção Social , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Autorrelato , Índice de Gravidade de Doença
9.
Psychogeriatrics ; 19(3): 282-285, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30393946

RESUMO

Adult-onset leucoencephalopathy with axonal spheroids and pigmented glia (ALSP), also known as hereditary diffuse leucoencephalopathy with spheroids (HDLS), is a progressive neurocognitive disorder that predominantly affects the cerebral white matter, mainly the frontal subcortical areas and the corpus callosum. Patients with ALSP are clinically characterized by a gradual onset of cognitive and behavioural dysfunction and personality changes, followed by motor impairments such as gait disturbance and bradykinesia. Given the disease-related degenerative changes of the frontal white matter, it is no wonder that patients with ALSP present with behavioural symptoms and non-fluent aphasia, which are found in patients with frontotemporal lobar degeneration. However, behavioural symptoms and non-fluent aphasia in a patient with ALSP have rarely reported in detail. Here, we describe a patient with ALSP who initially presented with remarkable behavioural signs and non-fluent primary progressive aphasia, which resembled symptoms of frontotemporal lobar degeneration. The present case suggests that ALSP should be included in the differential diagnosis for frontotemporal lobar degeneration.


Assuntos
Degeneração Lobar Frontotemporal/patologia , Leucoencefalopatias/patologia , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Substância Branca/diagnóstico por imagem , Adulto , Humanos , Leucoencefalopatias/genética , Imageamento por Ressonância Magnética , Masculino , Neuroglia , Tecnécio
10.
Acta Neuropathol ; 136(1): 1-17, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29744576

RESUMO

Progranulin (PGRN), encoded by the GRN gene in humans, is a secreted growth factor implicated in a multitude of processes ranging from regulation of inflammation to wound healing and tumorigenesis. The clinical importance of PGRN became especially evident in 2006, when heterozygous mutations in the GRN gene, resulting in haploinsufficiency, were found to be one of the main causes of frontotemporal lobar degeneration (FTLD). FTLD is a clinically heterogenous disease that results in the progressive atrophy of the frontal and temporal lobes of the brain. Despite significant research, the exact function of PGRN and its mechanistic relationship to FTLD remain unclear. However, growing evidence suggests a role for PGRN in the lysosome-most striking being that homozygous GRN mutation leads to neuronal ceroid lipofuscinosis, a lysosomal storage disease. Since this discovery, several links between PGRN and the lysosome have been established, including the existence of two independent lysosomal trafficking pathways, intralysosomal processing of PGRN into discrete functional peptides, and direct and indirect regulation of lysosomal hydrolases. Here, we summarize the cellular functions of PGRN, its roles in the nervous system, and its link to multiple neurodegenerative diseases, with a particular focus dedicated to recent lysosome-related mechanistic developments.


Assuntos
Lisossomos/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/terapia , Progranulinas/metabolismo , Animais , Humanos , Modelos Biológicos , Mutação/genética , Progranulinas/genética , Transporte Proteico
11.
J Biol Chem ; 291(29): 14996-5007, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226551

RESUMO

Cytoplasmic protein aggregates are one of the pathological hallmarks of neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Several RNA-binding proteins have been identified as components of inclusion bodies. Developmentally regulated RNA-binding protein 1 (Drb1)/RNA-binding motif protein 45 is an RNA-binding protein that was recently described as a component in ALS- and FTLD-related inclusion bodies. However, the molecular mechanism underlying cytoplasmic Drb1 aggregation remains unclear. Here, using an in vitro cellular model, we demonstrated that Drb1 co-localizes with cytoplasmic aggregates mediated by TAR DNA-binding protein 43, a major component of ALS and FTLD-related inclusion bodies. We also defined the domains involved in the subcellular localization of Drb1 to clarify the role of Drb1 in the formation of cytoplasmic aggregates in ALS and FTLD. Drb1 predominantly localized in the nucleus via a classical nuclear localization signal in its carboxyl terminus and is a shuttling protein between the nucleus and cytoplasm. Furthermore, we identify a double leucine motif serving as a nuclear export signal. The Drb1 mutant, presenting mutations in both nuclear localization signal and nuclear export signal, is prone to aggregate in the cytoplasm. The mutant Drb1-induced cytoplasmic aggregates not only recruit TAR DNA-binding protein 43 but also decrease the mitochondrial membrane potential. Taken together, these results indicate that perturbation of Drb1 nuclear-cytoplasmic trafficking induces toxic cytoplasmic aggregates, suggesting that mislocalization of Drb1 is involved in the cause of cytotoxicity in neuronal cells.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/genética , Degeneração Lobar Frontotemporal/metabolismo , Células HeLa , Humanos , Corpos de Inclusão/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/genética , Sinais de Exportação Nuclear/genética , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Agregação Patológica de Proteínas/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
12.
J Biol Chem ; 291(11): 5473-5483, 2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26769969

RESUMO

Intracellular aggregates of phosphorylated TDP-43 are a major component of ubiquitin-positive inclusions in the brains of patients with frontotemporal lobar degeneration and ALS and are considered a pathological hallmark. Here, to gain insight into the mechanism of intracellular TDP-43 accumulation, we examined the relationship between phosphorylation and aggregation of TDP-43. We found that expression of a hyperactive form of casein kinase 1 δ (CK1δ1-317, a C-terminally truncated form) promotes mislocalization and cytoplasmic accumulation of phosphorylated TDP-43 (ubiquitin- and p62-positive) in cultured neuroblastoma SH-SY5Y cells. Insoluble phosphorylated TDP-43 prepared from cells co-expressing TDP-43 and CK1δ1-317 functioned as seeds for TDP-43 aggregation in cultured cells, indicating that CK1δ1-317-induced aggregated TDP-43 has prion-like properties. A striking toxicity and alterations of TDP-43 were also observed in yeast expressing TDP-43 and CK1δ1-317. Therefore, abnormal activation of CK1δ causes phosphorylation of TDP-43, leading to the formation of cytoplasmic TDP-43 aggregates, which, in turn, may trigger neurodegeneration.


Assuntos
Caseína Quinase Idelta/metabolismo , Proteínas de Ligação a DNA/metabolismo , Agregados Proteicos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/análise , Ativação Enzimática , Humanos , Dados de Sequência Molecular , Fosforilação , Príons/análise , Príons/metabolismo , Agregação Patológica de Proteínas/metabolismo , Transporte Proteico
13.
Mol Genet Metab ; 117(2): 53-65, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26422115

RESUMO

Sixty years after its discovery, the lysosome is no longer considered as cell's waste bin but as an organelle playing a central role in cell metabolism. Besides its well known association with lysosomal storage disorders (mostly rare and life-threatening diseases), recent data have shown that the lysosome is also a player in some of the most common conditions of our time; and, perhaps even most important, it is not only a target for orphan drugs (rare disease therapeutic approaches) but also a putative target to treat patients suffering from common complex diseases worldwide. Here we review the striking associations linking rare lysosomal storage disorders such as the well-known Gaucher disease, or even the recently discovered, extremely rare Neuronal Ceroid Lipofuscinosis-11 and some of the most frequent, multifaceted and complex disorders of modern society such as cancer, Parkinson's disease and frontotemporal lobar degeneration.


Assuntos
Doenças por Armazenamento dos Lisossomos/complicações , Animais , Degeneração Lobar Frontotemporal/etiologia , Degeneração Lobar Frontotemporal/genética , Degeneração Lobar Frontotemporal/metabolismo , Humanos , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Doenças por Armazenamento dos Lisossomos/genética , Lisossomos/metabolismo , Mutação , Neoplasias/etiologia , Neoplasias/genética , Neoplasias/metabolismo , Doença de Parkinson/etiologia , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doenças Raras/complicações , Doenças Raras/tratamento farmacológico , Doenças Raras/genética
14.
Acta Neuropathol ; 131(4): 587-604, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26895297

RESUMO

Deposition of the nuclear DNA/RNA-binding protein Fused in sarcoma (FUS) in cytosolic inclusions is a common hallmark of some cases of frontotemporal lobar degeneration (FTLD-FUS) and amyotrophic lateral sclerosis (ALS-FUS). Whether both diseases also share common pathological mechanisms is currently unclear. Based on our previous finding that FUS deposits are hypomethylated in FTLD-FUS but not in ALS-FUS, we have now investigated whether genetic or pharmacological inactivation of Protein arginine methyltransferase 1 (PRMT1) activity results in unmethylated FUS or in alternatively methylated forms of FUS. To do so, we generated FUS-specific monoclonal antibodies that specifically recognize unmethylated arginine (UMA), monomethylated arginine (MMA) or asymmetrically dimethylated arginine (ADMA). Loss of PRMT1 indeed not only results in an increase of UMA FUS and a decrease of ADMA FUS, but also in a significant increase of MMA FUS. Compared to ADMA FUS, UMA and MMA FUS exhibit much higher binding affinities to Transportin-1, the nuclear import receptor of FUS, as measured by pull-down assays and isothermal titration calorimetry. Moreover, we show that MMA FUS occurs exclusively in FTLD-FUS, but not in ALS-FUS. Our findings therefore provide additional evidence that FTLD-FUS and ALS-FUS are caused by distinct disease mechanisms although both share FUS deposits as a common denominator.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Degeneração Lobar Frontotemporal/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , beta Carioferinas/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Anticorpos/farmacologia , Arginina/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Células-Tronco Embrionárias , Inibidores Enzimáticos/farmacologia , Feminino , Degeneração Lobar Frontotemporal/genética , Humanos , Corpos de Inclusão/efeitos dos fármacos , Corpos de Inclusão/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Proteína-Arginina N-Metiltransferases/genética , Proteína FUS de Ligação a RNA/imunologia , Ratos , beta Carioferinas/imunologia
15.
J Neurosci ; 34(23): 7802-13, 2014 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-24899704

RESUMO

Abnormal cytoplasmic accumulation of Fused in Sarcoma (FUS) in neurons defines subtypes of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). FUS is a member of the FET protein family that includes Ewing's sarcoma (EWS) and TATA-binding protein-associated factor 2N (TAF15). FET proteins are predominantly localized to the nucleus, where they bind RNA and DNA to modulate transcription, mRNA splicing, and DNA repair. In ALS cases with FUS inclusions (ALS-FUS), mutations in the FUS gene cause disease, whereas FTLD cases with FUS inclusions (FTLD-FUS) do not harbor FUS mutations. Notably, in FTLD-FUS, all FET proteins accumulate with their nuclear import receptor Transportin 1 (TRN1), in contrast ALS-FUS inclusions are exclusively positive for FUS. In the present study, we show that induction of DNA damage replicates several pathologic hallmarks of FTLD-FUS in immortalized human cells and primary human neurons and astrocytes. Treatment with the antibiotic calicheamicin γ1, which causes DNA double-strand breaks, leads to the cytoplasmic accumulation of FUS, TAF15, EWS, and TRN1. Moreover, cytoplasmic translocation of FUS is mediated by phosphorylation of its N terminus by the DNA-dependent protein kinase. Finally, we observed elevated levels of phospho-H2AX in FTLD-FUS brains, indicating that DNA damage occurs in patients. Together, our data reveal a novel regulatory mechanism for FUS localization in cells and suggest that DNA damage may contribute to the accumulation of FET proteins observed in human FTLD-FUS cases, but not in ALS-FUS.


Assuntos
Citoplasma/metabolismo , Dano ao DNA/fisiologia , Proteína Quinase Ativada por DNA/metabolismo , Degeneração Lobar Frontotemporal/patologia , Proteína FUS de Ligação a RNA/metabolismo , Aminoglicosídeos/farmacologia , Antibióticos Antineoplásicos/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Células Cultivadas , Citoplasma/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Enedi-Inos/farmacologia , Degeneração Lobar Frontotemporal/metabolismo , Humanos , Imunoprecipitação , Mutagênicos/farmacologia , Mutação/genética , Neurônios , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Proteína EWS de Ligação a RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo
16.
J Biol Chem ; 289(12): 8264-76, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24497641

RESUMO

Pathological alteration of TDP-43 (TAR DNA-binding protein-43), a protein involved in various RNA-mediated processes, is a hallmark feature of the neurodegenerative diseases amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Fragments of TDP-43, composed of the second RNA recognition motif (RRM2) and the disordered C terminus, have been observed in cytoplasmic inclusions in sporadic amyotrophic lateral sclerosis cases, suggesting that conformational changes involving RRM2 together with the disordered C terminus play a role in aggregation and toxicity. The biophysical data collected by CD and fluorescence spectroscopies reveal a three-state equilibrium unfolding model for RRM2, with a partially folded intermediate state that is not observed in RRM1. Strikingly, a portion of RRM2 beginning at position 208, which mimics a cleavage site observed in patient tissues, increases the population of this intermediate state. Mutually stabilizing interactions between the domains in the tethered RRM1 and RRM2 construct reduce the population of the intermediate state and enhance DNA/RNA binding. Despite the high sequence homology of the two domains, a network of large hydrophobic residues in RRM2 provides a possible explanation for the increased stability of RRM2 compared with RRM1. The cluster analysis suggests that the intermediate state may play a functional role by enhancing access to the nuclear export signal contained within its sequence. The intermediate state may also serve as a molecular hazard linking productive folding and function with pathological misfolding and aggregation that may contribute to disease.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Dobramento de Proteína , RNA/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Esclerose Lateral Amiotrófica/genética , Proteínas de Ligação a DNA/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência , Termodinâmica
17.
Acta Neuropathol ; 130(5): 643-60, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26197969

RESUMO

Accumulation of phosphorylated cytoplasmic TDP-43 inclusions accompanied by loss of normal nuclear TDP-43 in neurons and glia of the brain and spinal cord are the molecular hallmarks of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD-TDP). However, the role of cytoplasmic TDP-43 in the pathogenesis of these neurodegenerative TDP-43 proteinopathies remains unclear, due in part to a lack of valid mouse models. We therefore generated new mice with doxycycline (Dox)-suppressible expression of human TDP-43 (hTDP-43) harboring a defective nuclear localization signal (∆NLS) under the control of the neurofilament heavy chain promoter. Expression of hTDP-43∆NLS in these 'regulatable NLS' (rNLS) mice resulted in the accumulation of insoluble, phosphorylated cytoplasmic TDP-43 in brain and spinal cord, loss of endogenous nuclear mouse TDP-43 (mTDP-43), brain atrophy, muscle denervation, dramatic motor neuron loss, and progressive motor impairments leading to death. Notably, suppression of hTDP-43∆NLS expression by return of Dox to rNLS mice after disease onset caused a dramatic decrease in phosphorylated TDP-43 pathology, an increase in nuclear mTDP-43 to control levels, and the prevention of further motor neuron loss. rNLS mice back on Dox also showed a significant increase in muscle innervation, a rescue of motor impairments, and a dramatic extension of lifespan. Thus, the rNLS mice are new TDP-43 mouse models that delineate the timeline of pathology development, muscle denervation and neuron loss in ALS/FTLD-TDP. Importantly, even after neurodegeneration and onset of motor dysfunction, removal of cytoplasmic TDP-43 and the concomitant return of nuclear TDP-43 led to neuron preservation, muscle re-innervation and functional recovery.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/fisiopatologia , Recuperação de Função Fisiológica/fisiologia , Esclerose Lateral Amiotrófica/patologia , Animais , Atrofia , Encéfalo/metabolismo , Encéfalo/patologia , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Citoplasma/patologia , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Doxiciclina , Feminino , Degeneração Lobar Frontotemporal/patologia , Humanos , Masculino , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Transtornos dos Movimentos/patologia , Transtornos dos Movimentos/fisiopatologia , Músculo Esquelético/inervação , Distribuição Aleatória , Medula Espinal/metabolismo , Medula Espinal/patologia
18.
Brain Behav Immun ; 49: 182-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26021560

RESUMO

Mutations in progranulin gene (GRN) are one of the major causes of autosomal dominant Frontotemporal Lobar Degeneration (FTLD). Progranulin displays anti-inflammatory properties and is likely a ligand of Tumor Necrosis Factor (TNF) receptor 2, expressed on microglia. A few cytokines and chemokines are altered in cerebrospinal fluid (CSF) from patients with sporadic FTLD, whereas no information is available in familial cases. We evaluated, through BioPlex, levels of 27 inflammatory molecules, including cytokines, chemokines, and related receptors, in CSF and matched serum, from FTLD patients carrying GRN mutations as compared with sporadic FTLD with no GRN mutations and controls. Mean±SD Monocyte Chemoattractant Protein-1 (MCP-1) levels were significantly increased in CSF from sporadic FTLD patients as compared with controls (334.27±151.5 versus 159.7±49pg/ml; P⩽0.05). In GRN mutation carriers versus controls, CSF levels of MCP-1 were unchanged, whereas Interferon-γ-inducible protein-10 (IP-10) levels were increased (809.17±240.0 versus 436.61±202.5pg/ml; P=0.012). In the same group, TNFα and Interleukin (IL)-15 levels were decreased (3.18±1.41 versus 35.68±30.5pg/ml; P=0.013 and 9.34±5.54 versus 19.15±10.03pg/ml; P=0.023, respectively). Conversely, Regulated upon Activation, Normal T-cell Expressed, and Secreted (RANTES) levels were decreased in patients, with or without mutations, as compared with controls (4.63±3.30 and 2.58±20 versus 87.57±70pg/ml, respectively; P<0.05). Moreover, IP-10, IL-15 and RANTES CSF levels were not influenced by age, whereas MCP-1 levels increased with age (ρ=0.48; P=0.007). In conclusion, inflammatory de-regulation was observed in both sporadic FTLD and GRN carriers compared to controls, with a specific inflammatory profile for the latter group.


Assuntos
Demência Frontotemporal/líquido cefalorraquidiano , Demência Frontotemporal/genética , Mediadores da Inflamação/líquido cefalorraquidiano , Inflamação/líquido cefalorraquidiano , Inflamação/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Idoso , Quimiocina CCL2/sangue , Quimiocina CCL2/líquido cefalorraquidiano , Quimiocina CXCL10/sangue , Quimiocina CXCL10/líquido cefalorraquidiano , Feminino , Demência Frontotemporal/complicações , Humanos , Inflamação/complicações , Mediadores da Inflamação/sangue , Interleucina-15/sangue , Interleucina-15/líquido cefalorraquidiano , Masculino , Pessoa de Meia-Idade , Mutação , Progranulinas , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/líquido cefalorraquidiano
19.
J Neurol ; 271(8): 5549-5555, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38909120

RESUMO

BACKGROUND: Good accuracy for the clinical diagnosis of frontotemporal lobar degeneration (FTLD) by specialists in an early onset dementia clinic has been reported. OBJECTIVE: To assess the diagnostic accuracy of FTLD in an entire population, without restrictions related to patient age or diagnosing physician. METHODS: Volumes of the "Annual of the Pathological Autopsy Cases in Japan," with reports of 130,105 autopsies throughout Japan from 2007 to 2016, were descriptively analyzed. RESULTS: There were 219 patients with clinical and/or pathological diagnoses of FTLD. The sensitivity and specificity were 24.5% and 76.9%, respectively. Age at death for pathologically confirmed patients was 76.3 ± 11.6 years (mean ± standard deviation). Overlooked patients died significantly older than patients with an accurate clinical diagnosis. CONCLUSIONS: Clinical diagnoses of FTLD had low sensitivity. Furthermore, the age at death of pathologically confirmed patients suggests that FTLD affects a wide age range and is not restricted to presenile individuals.


Assuntos
Autopsia , Degeneração Lobar Frontotemporal , Humanos , Japão/epidemiologia , Degeneração Lobar Frontotemporal/patologia , Degeneração Lobar Frontotemporal/mortalidade , Idoso , Masculino , Feminino , Idoso de 80 Anos ou mais , Pessoa de Meia-Idade , Sensibilidade e Especificidade
20.
Mol Neurodegener ; 19(1): 46, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38862967

RESUMO

RNA binding proteins have emerged as central players in the mechanisms of many neurodegenerative diseases. In particular, a proteinopathy of fused in sarcoma (FUS) is present in some instances of familial Amyotrophic lateral sclerosis (ALS) and about 10% of sporadic Frontotemporal lobar degeneration (FTLD). Here we establish that focal injection of sonicated human FUS fibrils into brains of mice in which ALS-linked mutant or wild-type human FUS replaces endogenous mouse FUS is sufficient to induce focal cytoplasmic mislocalization and aggregation of mutant and wild-type FUS which with time spreads to distal regions of the brain. Human FUS fibril-induced FUS aggregation in the mouse brain of humanized FUS mice is accelerated by an ALS-causing FUS mutant relative to wild-type human FUS. Injection of sonicated human FUS fibrils does not induce FUS aggregation and subsequent spreading after injection into naïve mouse brains containing only mouse FUS, indicating a species barrier to human FUS aggregation and its prion-like spread. Fibril-induced human FUS aggregates recapitulate pathological features of FTLD including increased detergent insolubility of FUS and TAF15 and amyloid-like, cytoplasmic deposits of FUS that accumulate ubiquitin and p62, but not TDP-43. Finally, injection of sonicated FUS fibrils is shown to exacerbate age-dependent cognitive and behavioral deficits from mutant human FUS expression. Thus, focal seeded aggregation of FUS and further propagation through prion-like spread elicits FUS-proteinopathy and FTLD-like disease progression.


Assuntos
Progressão da Doença , Demência Frontotemporal , Camundongos Transgênicos , Proteína FUS de Ligação a RNA , Animais , Humanos , Camundongos , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Demência Frontotemporal/patologia , Demência Frontotemporal/metabolismo , Demência Frontotemporal/genética , Agregação Patológica de Proteínas/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteína FUS de Ligação a RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA