Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Mol Neurodegener ; 15(1): 27, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32299471

RESUMO

BACKGROUND: Studies link c-Abl activation with the accumulation of pathogenic α-synuclein (αS) and neurodegeneration in Parkinson's disease (PD). Currently, c-Abl, a tyrosine kinase activated by cellular stress, is thought to promote αS pathology by either directly phosphorylating αS or by causing autophagy deficits. METHODS: αS overexpressing transgenic (Tg) mice were used in this study. A53T Tg mice that express high levels of human mutant A53TαS under the control of prion protein promoter. Two different approaches were used in this study. Natural aging and seeding model of synucleinopathy. In seeding model, intracortical/intrastriatal (IC/IS) stereotaxic injection of toxic lysates was done using tissue lysates from end-stage symptomatic mice. In this study, nilotinib and pifithrin-α was used as a c-Abl and p53 inhibitor, respectively. Both Tg and non-transgenic (nTg) mice from each group were subjected to nilotinib (10 mg/kg) or vehicle (DMSO) treatment. Frozen brain tissues from PD and control human cases were analyzed. In vitro cells study was implied for c-Abl/p53 genetic manipulation to uncover signal transduction. RESULTS: Herein, we show that the pathologic effects of c-Abl in PD also involve activation of p53, as c-Abl activation in a transgenic mouse model of α-synucleinopathy (TgA53T) and human PD cases are associated with the increased p53 activation. Significantly, active p53 in TgA53T neurons accumulates in the cytosol, which may lead to inhibition of autophagy. Thus, we hypothesized that c-Abl-dependent p53 activation contributes to autophagy impairment in α-synucleinopathy. In support of the hypothesis, we show that c-Abl activation is sufficient to inhibit autophagy in p53-dependent manner. Moreover, inhibition of either c-Abl, using nilotinib, or p53, using pifithrin-α, was sufficient to increase autophagic flux in neuronal cells by inducing phosphorylation of AMP-activated kinase (AMPK), ULK1 activation, and down-regulation of mTORC1 signaling. Finally, we show that pharmacological attenuation of c-Abl activity by nilotinib treatment in the TgA53T mouse model reduces activation of p53, stimulates autophagy, decreases accumulation αS pathology, and delays disease onset. CONCLUSION: Collectively, our data show that c-Abl activation by α-synucleinopathy causes p53 dependent autophagy deficits and both c-Abl and p53 represent therapeutic target for PD.


Assuntos
Autofagia/fisiologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Humanos , Camundongos , Camundongos Transgênicos , Sinucleinopatias/metabolismo , Sinucleinopatias/patologia , alfa-Sinucleína/metabolismo
2.
PLoS Med ; 17(1): e1003012, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31978055

RESUMO

BACKGROUND: There is growing evidence that Alzheimer disease (AD) is a pervasive metabolic disorder with dysregulation in multiple biochemical pathways underlying its pathogenesis. Understanding how perturbations in metabolism are related to AD is critical to identifying novel targets for disease-modifying therapies. In this study, we test whether AD pathogenesis is associated with dysregulation in brain transmethylation and polyamine pathways. METHODS AND FINDINGS: We first performed targeted and quantitative metabolomics assays using capillary electrophoresis-mass spectrometry (CE-MS) on brain samples from three groups in the Baltimore Longitudinal Study of Aging (BLSA) (AD: n = 17; Asymptomatic AD [ASY]: n = 13; Control [CN]: n = 13) (overall 37.2% female; mean age at death 86.118 ± 9.842 years) in regions both vulnerable and resistant to AD pathology. Using linear mixed-effects models within two primary brain regions (inferior temporal gyrus [ITG] and middle frontal gyrus [MFG]), we tested associations between brain tissue concentrations of 26 metabolites and the following primary outcomes: group differences, Consortium to Establish a Registry for Alzheimer's Disease (CERAD) (neuritic plaque burden), and Braak (neurofibrillary pathology) scores. We found significant alterations in concentrations of metabolites in AD relative to CN samples, as well as associations with severity of both CERAD and Braak, mainly in the ITG. These metabolites represented biochemical reactions in the (1) methionine cycle (choline: lower in AD, p = 0.003; S-adenosyl methionine: higher in AD, p = 0.005); (2) transsulfuration and glutathione synthesis (cysteine: higher in AD, p < 0.001; reduced glutathione [GSH]: higher in AD, p < 0.001); (3) polyamine synthesis/catabolism (spermidine: higher in AD, p = 0.004); (4) urea cycle (N-acetyl glutamate: lower in AD, p < 0.001); (5) glutamate-aspartate metabolism (N-acetyl aspartate: lower in AD, p = 0.002); and (6) neurotransmitter metabolism (gamma-amino-butyric acid: lower in AD, p < 0.001). Utilizing three Gene Expression Omnibus (GEO) datasets, we then examined mRNA expression levels of 71 genes encoding enzymes regulating key reactions within these pathways in the entorhinal cortex (ERC; AD: n = 25; CN: n = 52) and hippocampus (AD: n = 29; CN: n = 56). Complementing our metabolomics results, our transcriptomics analyses also revealed significant alterations in gene expression levels of key enzymatic regulators of biochemical reactions linked to transmethylation and polyamine metabolism. Our study has limitations: our metabolomics assays measured only a small proportion of all metabolites participating in the pathways we examined. Our study is also cross-sectional, limiting our ability to directly test how AD progression may impact changes in metabolite concentrations or differential-gene expression. Additionally, the relatively small number of brain tissue samples may have limited our power to detect alterations in all pathway-specific metabolites and their genetic regulators. CONCLUSIONS: In this study, we observed broad dysregulation of transmethylation and polyamine synthesis/catabolism, including abnormalities in neurotransmitter signaling, urea cycle, aspartate-glutamate metabolism, and glutathione synthesis. Our results implicate alterations in cellular methylation potential and increased flux in the transmethylation pathways, increased demand on antioxidant defense mechanisms, perturbations in intermediate metabolism in the urea cycle and aspartate-glutamate pathways disrupting mitochondrial bioenergetics, increased polyamine biosynthesis and breakdown, as well as abnormalities in neurotransmitter metabolism that are related to AD.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Redes e Vias Metabólicas/fisiologia , Metaboloma/fisiologia , Poliaminas/metabolismo , Transcriptoma/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Encéfalo/patologia , Feminino , Humanos , Estudos Longitudinais , Masculino , Metilação
3.
J Exp Med ; 216(6): 1411-1430, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31036561

RESUMO

Persistent microglia-mediated neuroinflammation is a major pathophysiological contributor to the progression of Parkinson's disease (PD), but the cell-signaling mechanisms governing chronic neuroinflammation are not well understood. Here, we show that Fyn kinase, in conjunction with the class B scavenger receptor CD36, regulates the microglial uptake of aggregated human α-synuclein (αSyn), which is the major component of PD-associated Lewy bodies. αSyn can effectively mediate LPS-independent priming and activation of the microglial NLRP3 inflammasome. Fyn kinase regulates both of these processes; it mediates PKCδ-dependent NF-κB-p65 nuclear translocation, leading to inflammasome priming, and facilitates αSyn import into microglia, contributing to the generation of mitochondrial reactive oxygen species and consequently to inflammasome activation. In vivo experiments using A53T and viral-αSyn overexpression mouse models as well as human PD neuropathological results further confirm the role of Fyn in NLRP3 inflammasome activation. Collectively, our study identifies a novel Fyn-mediated signaling mechanism that amplifies neuroinflammation in PD.


Assuntos
Inflamassomos/metabolismo , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Dobramento de Proteína , Proteínas Proto-Oncogênicas c-fyn/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Animais , Antígenos CD36/metabolismo , Dependovirus/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Gliose/metabolismo , Gliose/patologia , Humanos , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Agregados Proteicos , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas c-fyn/deficiência , Espécies Reativas de Oxigênio/metabolismo
4.
Neurobiol Aging ; 75: 223.e1-223.e10, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30448004

RESUMO

The role of genetic variability in dementia with Lewy bodies (DLB) is now indisputable; however, data regarding copy number variation (CNV) in this disease has been lacking. Here, we used whole-genome genotyping of 1454 DLB cases and 1525 controls to assess copy number variability. We used 2 algorithms to confidently detect CNVs, performed a case-control association analysis, screened for candidate CNVs previously associated with DLB-related diseases, and performed a candidate gene approach to fully explore the data. We identified 5 CNV regions with a significant genome-wide association to DLB; 2 of these were only present in cases and absent from publicly available databases: one of the regions overlapped LAPTM4B, a known lysosomal protein, whereas the other overlapped the NME1 locus and SPAG9. We also identified DLB cases presenting rare CNVs in genes previously associated with DLB or related neurodegenerative diseases, such as SNCA, APP, and MAPT. To our knowledge, this is the first study reporting genome-wide CNVs in a large DLB cohort. These results provide preliminary evidence for the contribution of CNVs in DLB risk.


Assuntos
Variações do Número de Cópias de DNA/genética , Predisposição Genética para Doença/genética , Doença por Corpos de Lewy/genética , Proteínas Oncogênicas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Idoso de 80 Anos ou mais , Feminino , Genoma , Estudo de Associação Genômica Ampla , Humanos , Masculino , Proteínas de Membrana/genética , Polimorfismo de Nucleotídeo Único/genética
5.
Science ; 362(6414)2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30385548

RESUMO

The pathologic accumulation and aggregation of α-synuclein (α-syn) underlies Parkinson's disease (PD). The molecular mechanisms by which pathologic α-syn causes neurodegeneration in PD are not known. Here, we found that pathologic α-syn activates poly(adenosine 5'-diphosphate-ribose) (PAR) polymerase-1 (PARP-1), and PAR generation accelerates the formation of pathologic α-syn, resulting in cell death via parthanatos. PARP inhibitors or genetic deletion of PARP-1 prevented pathologic α-syn toxicity. In a feed-forward loop, PAR converted pathologic α-syn to a more toxic strain. PAR levels were increased in the cerebrospinal fluid and brains of patients with PD, suggesting that PARP activation plays a role in PD pathogenesis. Thus, strategies aimed at inhibiting PARP-1 activation could hold promise as a disease-modifying therapy to prevent the loss of dopamine neurons in PD.


Assuntos
Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , alfa-Sinucleína/metabolismo , Animais , Benzimidazóis/farmacologia , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Ativação Enzimática , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/genética , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/genética
6.
Am J Pathol ; 188(3): 739-756, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29248459

RESUMO

Despite increasing appreciation that oligomeric amyloid-ß (Aß) may contribute to cognitive decline of Alzheimer disease, defining the most critical forms has been thwarted by the changeable nature of these aggregates and the varying methods used for detection. Herein, using a broad approach, we quantified Aß oligomers during the evolution of cognitive deficits in an aggressive model of Aß amyloidosis. Amyloid precursor protein/tetracycline transactivator mice underwent behavioral testing at 3, 6, 9, and 12 months of age to evaluate spatial learning and memory, followed by histologic assessment of amyloid burden and biochemical characterization of oligomeric Aß species. Transgenic mice displayed progressive impairments in acquisition and immediate recall of the trained platform location. Biochemical analysis of cortical extracts from behaviorally tested mice revealed distinct age-dependent patterns of accumulation in multiple oligomeric species. Dot blot analysis demonstrated that nonfibrillar Aß oligomers were highly soluble and extracted into a fraction enriched for extracellular proteins, whereas prefibrillar species required high-detergent conditions to retrieve, consistent with membrane localization. Low-detergent extracts tested by 82E1 enzyme-linked immunosorbent assay confirmed the presence of bona fide Aß oligomers, whereas immunoprecipitation-Western blotting using high-detergent extracts revealed a variety of SDS-stable low-n species. These findings show that different Aß oligomers vary in solubility, consistent with distinct localization, and identify nonfibrillar Aß oligomer-positive aggregates as tracking most closely with cognitive decline in this model.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Aprendizagem Espacial/fisiologia , Memória Espacial/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Atividade Motora/fisiologia
7.
Elife ; 62017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28440221

RESUMO

Memory loss in Alzheimer's disease (AD) is attributed to pervasive weakening and loss of synapses. Here, we present findings supporting a special role for excitatory synapses connecting pyramidal neurons of the hippocampus and cortex with fast-spiking parvalbumin (PV) interneurons that control network excitability and rhythmicity. Excitatory synapses on PV interneurons are dependent on the AMPA receptor subunit GluA4, which is regulated by presynaptic expression of the synaptogenic immediate early gene NPTX2 by pyramidal neurons. In a mouse model of AD amyloidosis, Nptx2-/- results in reduced GluA4 expression, disrupted rhythmicity, and increased pyramidal neuron excitability. Postmortem human AD cortex shows profound reductions of NPTX2 and coordinate reductions of GluA4. NPTX2 in human CSF is reduced in subjects with AD and shows robust correlations with cognitive performance and hippocampal volume. These findings implicate failure of adaptive control of pyramidal neuron-PV circuits as a pathophysiological mechanism contributing to cognitive failure in AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Proteína C-Reativa/análise , Disfunção Cognitiva/fisiopatologia , Proteínas do Tecido Nervoso/análise , Doença de Alzheimer/patologia , Animais , Proteína C-Reativa/líquido cefalorraquidiano , Córtex Cerebral/patologia , Modelos Animais de Doenças , Hipocampo/patologia , Humanos , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/líquido cefalorraquidiano
8.
Oncotarget ; 8(63): 106721-106739, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29290984

RESUMO

RNF146 is an E3 ubiquitin ligase that specifically recognizes and polyubiquitinates poly (ADP-ribose) (PAR)-conjugated substrates for proteasomal degradation. RNF146 has been shown to be neuroprotective against PAR polymerase-1 (PARP1)-induced cell death during stroke. Here we report that RNF146 expression and RNF146 inducers can prevent cell death elicited by Parkinson's disease (PD)-associated and PARP1-activating stimuli. In SH-SY5Y cells, RNF146 expression conferred resistance to toxic stimuli that lead to PARP1 activation. High-throughput screen using a luciferase construct harboring the RNF146 promoter identified liquiritigenin as an RNF146 inducer. We found that RNF146 expression by liquiritigenin was mediated by estrogen receptor activation and contributed to cytoprotective effect of liquiritigenin. Finally, RNF146 expression by liquiritigenin in mouse brains provided dopaminergic neuroprotection in a 6-hydroxydopamine PD mouse model. Given the presence of PARP1 activity and RNF146 deficits in PD, it could be a potential therapeutic strategy to restore RNF146 expression by natural compounds or estrogen receptor activation.

10.
Aging Cell ; 16(2): 281-292, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27995769

RESUMO

Multiple loss-of-function mutations in TRIAD3 (a.k.a. RNF216) have recently been identified in patients suffering from Gordon Holmes syndrome (GHS), characterized by cognitive decline, dementia, and movement disorders. TRIAD3A is an E3 ubiquitin ligase that recognizes and facilitates the ubiquitination of its target for degradation by the ubiquitin-proteasome system (UPS). Here, we demonstrate that two of these missense substitutions in TRIAD3 (R660C and R694C) could not regulate the degradation of their neuronal target, activity-regulated cytoskeletal-associated protein (Arc/Arg 3.1), whose expression is critical for synaptic plasticity and memory. The synaptic deficits due to the loss of endogenous TRIAD3A could not be rescued by TRIAD3A harboring GHS-associated missense mutations. Moreover, we demonstrate that the loss of endogenous TRIAD3A in the mouse hippocampal CA1 region led to deficits in spatial learning and memory. Finally, we show that these missense mutations abolished the interaction of TRIAD3A with Arc, disrupting Arc ubiquitination, and consequently Arc degradation. Our current findings of Arc misregulation by TRIAD3A variants suggest that loss-of-function mutations in TRIAD3A may contribute to dementia observed in patients with GHS driven by dysfunctional UPS components, leading to cognitive impairments through the synaptic protein Arc.


Assuntos
Ataxia Cerebelar/genética , Disfunção Cognitiva/patologia , Proteínas do Citoesqueleto/metabolismo , Hormônio Liberador de Gonadotropina/deficiência , Hipogonadismo/genética , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Sinapses/patologia , Ubiquitina-Proteína Ligases/genética , Animais , Região CA1 Hipocampal/patologia , Clatrina/metabolismo , Disfunção Cognitiva/metabolismo , Endocitose , Técnicas de Silenciamento de Genes , Hormônio Liberador de Gonadotropina/genética , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto/genética , Ligação Proteica , Proteólise , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Memória Espacial , Sinapses/metabolismo , Transmissão Sináptica , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
11.
Hum Mol Genet ; 25(16): 3454-3466, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27378699

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG expansion in the gene-encoding Huntingtin (HTT). Transcriptome dysregulation is a major feature of HD pathogenesis, as revealed by a large body of work on gene expression profiling of tissues from human HD patients and mouse models. These studies were primarily focused on transcriptional changes affecting steady-state overall gene expression levels using microarray based approaches. A major missing component, however, has been the study of transcriptome changes at the post-transcriptional level, such as alternative splicing. Alternative splicing is a critical mechanism for expanding regulatory and functional diversity from a limited number of genes, and is particularly complex in the mammalian brain. Here we carried out a deep RNA-seq analysis of the BA4 (Brodmann area 4) motor cortex from seven human HD brains and seven controls to systematically discover aberrant alternative splicing events and characterize potential associated splicing factors in HD. We identified 593 differential alternative splicing events between HD and control brains. Using two expanded panels with a total of 108 BA4 tissues from patients and controls, we identified four splicing factors exhibiting significantly altered expression levels in HD patient brains. Moreover, follow-up molecular analyses of one splicing factor PTBP1 revealed its impact on disease-associated splicing patterns in HD. Collectively, our data provide genomic evidence for widespread splicing dysregulation in HD brains, and suggest the role of aberrant alternative splicing in the pathogenesis of HD.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/genética , Doença de Huntington/genética , Córtex Motor/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Transcriptoma/genética , Adulto , Idoso , Processamento Alternativo/genética , Animais , Autopsia , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Ribonucleoproteínas Nucleares Heterogêneas/biossíntese , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Proteína Huntingtina/genética , Doença de Huntington/fisiopatologia , Masculino , Camundongos , Pessoa de Meia-Idade , Córtex Motor/patologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/biossíntese
13.
J Clin Invest ; 126(8): 2970-88, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27348587

RESUMO

Aggregation of α-synuclein contributes to the formation of Lewy bodies and neurites, the pathologic hallmarks of Parkinson disease (PD) and α-synucleinopathies. Although a number of human mutations have been identified in familial PD, the mechanisms that promote α-synuclein accumulation and toxicity are poorly understood. Here, we report that hyperactivity of the nonreceptor tyrosine kinase c-Abl critically regulates α-synuclein-induced neuropathology. In mice expressing a human α-synucleinopathy-associated mutation (hA53Tα-syn mice), deletion of the gene encoding c-Abl reduced α-synuclein aggregation, neuropathology, and neurobehavioral deficits. Conversely, overexpression of constitutively active c-Abl in hA53Tα-syn mice accelerated α-synuclein aggregation, neuropathology, and neurobehavioral deficits. Moreover, c-Abl activation led to an age-dependent increase in phosphotyrosine 39 α-synuclein. In human postmortem samples, there was an accumulation of phosphotyrosine 39 α-synuclein in brain tissues and Lewy bodies of PD patients compared with age-matched controls. Furthermore, in vitro studies show that c-Abl phosphorylation of α-synuclein at tyrosine 39 enhances α-synuclein aggregation. Taken together, this work establishes a critical role for c-Abl in α-synuclein-induced neurodegeneration and demonstrates that selective inhibition of c-Abl may be neuroprotective. This study further indicates that phosphotyrosine 39 α-synuclein is a potential disease indicator for PD and related α-synucleinopathies.


Assuntos
Encéfalo/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , alfa-Sinucleína/metabolismo , Idoso , Animais , Modelos Animais de Doenças , Feminino , Deleção de Genes , Células HEK293 , Humanos , Corpos de Lewy/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Neuritos/metabolismo , Neuroproteção , Doença de Parkinson/genética , Fosforilação , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas c-abl/genética , alfa-Sinucleína/genética
14.
Neuron ; 88(4): 667-77, 2015 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-26590344

RESUMO

Huntington disease (HD) is caused by a CAG ⋅ CTG expansion in the huntingtin (HTT) gene. While most research has focused on the HTT polyGln-expansion protein, we demonstrate that four additional, novel, homopolymeric expansion proteins (polyAla, polySer, polyLeu, and polyCys) accumulate in HD human brains. These sense and antisense repeat-associated non-ATG (RAN) translation proteins accumulate most abundantly in brain regions with neuronal loss, microglial activation and apoptosis, including caudate/putamen, white matter, and, in juvenile-onset cases, also the cerebellum. RAN protein accumulation and aggregation are length dependent, and individual RAN proteins are toxic to neural cells independent of RNA effects. These data suggest RAN proteins contribute to HD and that therapeutic strategies targeting both sense and antisense genes may be required for efficacy in HD patients. This is the first demonstration that RAN proteins are expressed across an expansion located in an open reading frame and suggests RAN translation may also contribute to other polyglutamine diseases.


Assuntos
Encéfalo/metabolismo , Doença de Huntington/genética , Proteínas do Tecido Nervoso/genética , RNA Antissenso/genética , Expansão das Repetições de Trinucleotídeos/genética , Adulto , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Cerebelo/metabolismo , Criança , Feminino , Lobo Frontal/metabolismo , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Masculino , Pessoa de Meia-Idade , Neostriado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Biossíntese de Proteínas , RNA Antissenso/metabolismo , Adulto Jovem
15.
Science ; 349(6248): 650-5, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26250685

RESUMO

Cytoplasmic aggregation of TDP-43, accompanied by its nuclear clearance, is a key common pathological hallmark of amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD). However, a limited understanding of this RNA-binding protein (RBP) impedes the clarification of pathogenic mechanisms underlying TDP-43 proteinopathy. In contrast to RBPs that regulate splicing of conserved exons, we found that TDP-43 repressed the splicing of nonconserved cryptic exons, maintaining intron integrity. When TDP-43 was depleted from mouse embryonic stem cells, these cryptic exons were spliced into messenger RNAs, often disrupting their translation and promoting nonsense-mediated decay. Moreover, enforced repression of cryptic exons prevented cell death in TDP-43-deficient cells. Furthermore, repression of cryptic exons was impaired in ALS-FTD cases, suggesting that this splicing defect could potentially underlie TDP-43 proteinopathy.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteínas de Ligação a DNA/fisiologia , Éxons/genética , Demência Frontotemporal/genética , Splicing de RNA , Animais , Proteínas Relacionadas à Autofagia , Sequência de Bases , Células Cultivadas , Cisteína Endopeptidases/genética , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias , Técnicas de Inativação de Genes , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Estabilidade de RNA , RNA Mensageiro/metabolismo , Análise de Sequência de DNA
16.
PLoS One ; 10(6): e0129618, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26086915

RESUMO

In Alzheimer's disease (AD), one of the early responses to Aß amyloidosis is recruitment of microglia to areas of new plaque. Microglial receptors such as cannabinoid receptor 2 (CB2) might be a suitable target for development of PET radiotracers that could serve as imaging biomarkers of Aß-induced neuroinflammation. Mouse models of amyloidosis (J20APPswe/ind and APPswe/PS1ΔE9) were used to investigate the cellular distribution of CB2 receptors. Specificity of CB2 antibody (H60) was confirmed using J20APPswe/ind mice lacking CB2 receptors. APPswe/PS1ΔE9 mice were used in small animal PET with a CB2-targeting radiotracer, [11C]A836339. These studies revealed increased binding of [11C]A836339 in amyloid-bearing mice. Specificity of the PET signal was confirmed in a blockade study with a specific CB2 antagonist, AM630. Confocal microscopy revealed that CB2-receptor immunoreactivity was associated with astroglial (GFAP) and, predominantly, microglial (CD68) markers. CB2 receptors were observed, in particular, in microglial processes forming engulfment synapses with Aß plaques. In contrast to glial cells, neuron (NeuN)-derived CB2 signal was equal between amyloid-bearing and control mice. The pattern of neuronal CB2 staining in amyloid-bearing mice was similar to that in human cases of AD. The data collected in this study indicate that Aß amyloidosis without concomitant tau pathology is sufficient to activate CB2 receptors that are suitable as an imaging biomarker of neuroinflammation. The main source of enhanced CB2 PET binding in amyloid-bearing mice is increased CB2 immunoreactivity in activated microglia. The presence of CB2 immunoreactivity in neurons does not likely contribute to the enhanced CB2 PET signal in amyloid-bearing mice due to a lack of significant neuronal loss in this model. However, significant loss of neurons as seen at late stages of AD might decrease the CB2 PET signal due to loss of neuronally-derived CB2. Thus this study in mouse models of AD indicates that a CB2-specific radiotracer can be used as a biomarker of neuroinflammation in the early preclinical stages of AD, when no significant neuronal loss has yet developed.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/análise , Amiloidose/patologia , Inflamação/patologia , Neurônios/patologia , Receptor CB2 de Canabinoide/análise , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/imunologia , Precursor de Proteína beta-Amiloide/imunologia , Amiloidose/diagnóstico por imagem , Amiloidose/imunologia , Animais , Biomarcadores/análise , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Inflamação/diagnóstico por imagem , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/diagnóstico por imagem , Microglia/imunologia , Microglia/patologia , Neurônios/diagnóstico por imagem , Neurônios/imunologia , Tomografia por Emissão de Pósitrons , Receptor CB2 de Canabinoide/imunologia
17.
Neurobiol Dis ; 73: 229-43, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25461191

RESUMO

Mutations in the ATP13A2 (PARK9) gene cause early-onset, autosomal recessive Parkinson's disease (PD) and Kufor-Rakeb syndrome. ATP13A2 mRNA is spliced into three distinct isoforms encoding a P5-type ATPase involved in regulating heavy metal transport across vesicular membranes. Here, we demonstrate that three ATP13A2 mRNA isoforms are expressed in the normal human brain and are modestly increased in the cingulate cortex of PD cases. ATP13A2 can mediate protection toward a number of stressors in mammalian cells and can protect against α-synuclein-induced toxicity in cellular and invertebrate models of PD. Using a primary cortical neuronal model combined with lentiviral-mediated gene transfer, we demonstrate that human ATP13A2 isoforms 1 and 2 display selective neuroprotective effects toward toxicity induced by manganese and hydrogen peroxide exposure through an ATPase-independent mechanism. The familial PD mutations, F182L and G504R, abolish the neuroprotective effects of ATP13A2 consistent with a loss-of-function mechanism. We further demonstrate that the AAV-mediated overexpression of human ATP13A2 is not sufficient to attenuate dopaminergic neurodegeneration, neuropathology, and striatal dopamine and motoric deficits induced by human α-synuclein expression in a rat model of PD. Intriguingly, the delivery of an ATPase-deficient form of ATP13A2 (D513N) to the substantia nigra is sufficient to induce dopaminergic neuronal degeneration and motor deficits in rats, potentially suggesting a dominant-negative mechanism of action. Collectively, our data demonstrate a distinct lack of ATP13A2-mediated protection against α-synuclein-induced neurotoxicity in the rat nigrostriatal dopaminergic pathway, and limited neuroprotective capacity overall, and raise doubts about the potential of ATP13A2 as a therapeutic target for PD.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Peróxido de Hidrogênio/toxicidade , Manganês/toxicidade , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Bancos de Tecidos , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/genética , Gravidez , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/farmacologia , Ratos , Ratos Sprague-Dawley
18.
Cell Rep ; 7(5): 1401-1409, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-24857657

RESUMO

The primarily neuronal RNA-binding protein HuD is implicated in learning and memory. Here, we report the identification of several HuD target transcripts linked to Alzheimer's disease (AD) pathogenesis. HuD interacted with the 3' UTRs of APP mRNA (encoding amyloid precursor protein) and BACE1 mRNA (encoding ß-site APP-cleaving enzyme 1) and increased the half-lives of these mRNAs. HuD also associated with and stabilized the long noncoding (lnc)RNA BACE1AS, which partly complements BACE1 mRNA and enhances BACE1 expression. Consistent with HuD promoting production of APP and APP-cleaving enzyme, the levels of APP, BACE1, BACE1AS, and Aß were higher in the brain of HuD-overexpressing mice. Importantly, cortex (superior temporal gyrus) from patients with AD displayed significantly higher levels of HuD and, accordingly, elevated APP, BACE1, BACE1AS, and Aß than did cortical tissue from healthy age-matched individuals. We propose that HuD jointly promotes the production of APP and the cleavage of its amyloidogenic fragment, Aß.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas ELAV/metabolismo , RNA Longo não Codificante/metabolismo , Regiões 3' não Traduzidas , Idoso , Idoso de 80 Anos ou mais , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Estudos de Casos e Controles , Linhagem Celular Tumoral , Córtex Cerebral/metabolismo , Proteínas ELAV/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Estabilidade de RNA , RNA Longo não Codificante/genética
19.
Dement. neuropsychol ; 7(1): 83-87, jan.-mar. 2013. tab
Artigo em Inglês | LILACS | ID: lil-670739

RESUMO

OBJECTIVE: To describe characteristics of hippocampal sclerosis dementia. METHODS: Convenience sample of Hippocampal sclerosis dementia (HSD) recruited from the Johns Hopkins University Brain Resource Center. Twenty-four cases with post-mortem pathological diagnosis of hippocampal sclerosis dementia were reviewed for clinical characterization. RESULTS: The cases showed atrophy and neuronal loss localized to the hippocampus, amygdala and entorrhinal cortex. The majority (79.2%) had amnesia at illness onset, and many (54.2%) showed abnormal conduct and psychiatric disorder. Nearly 42% presented with an amnesic state, and 37.5% presented with amnesia plus abnormal conduct and psychiatric disorder. All eventually developed a behavioral or psychiatric disorder. Disorientation, executive dysfunction, aphasia, agnosia and apraxia were uncommon at onset. Alzheimer disease (AD) was the initial clinical diagnosis in 89% and the final clinical diagnosis in 75%. Diagnosis of frontotemporal dementia (FTD) was uncommon (seen in 8%). CONCLUSION: HSD shows pathological characteristics of FTD and clinical features that mimic AD and overlap with FTD. The findings, placed in the context of earlier work, support the proposition that HSD belongs to the FTD family, where it may be identified as an amnesic variant.


OBJETIVO: descrever as características da demência com esclerose hipocampal. MÉTODOS: Uma amostra de conveniência de HSD foi recrutada no Johns Hopkins University Brain Resource Center. Vinte e quatro casos com diagnóstico patológico pós-morte de demência com esclerose hipocampal foram revisados para caracterização clínica, utilizando variáveis obtidas de prontuários médicos. OBJETIVO: descrever as características da demência com esclerose hipocampal.MÉTODOS: Uma amostra de conveniência de HSD foi recrutada no Johns Hopkins University Brain Resource Center. Vinte e quatro casos com diagnóstico patológico pós-morte de demência com esclerose hipocampal foram revisados para caracterização clínica, utilizando variáveis obtidas de prontuários médicos. RESULTADOS: A maioria dos pacientes (79,2%) tinha amnésia no início, e a maioria (54,2%) apresentava comportamento anormal e transtorno psiquiátrico. Aproximadamente 42% apresentavam um estado amnésico e 37,5% amnésia mais conduta anormal e transtorno psiquiátrico. Todos acabaram por desenvolver um distúrbio comportamental ou psiquiátrico. Desorientação, disfunção executiva, afasia, agnosia e apraxia foram incomuns no início. A doença de Alzheimer (AD) foi o diagnóstico clínico inicial em 89% e o diagnóstico clínico final em 75%. A maior parte dos casos mostrou atrofia e perda neuronal localizada no hipocampo, amígadale cortex entorrinal. CONCLUSÃO: HSD apresenta características patológicas de DFT e as características clínicas que mimetizam AD. Os resultados, colocadas no contexto do trabalho anterior, suporta a ideia de que HSD pertence à família FTD, em que pode ser identificada como uma variante amnésica. RESULTADOS: A maioria dos pacientes (79,2%) tinha amnésia no início, e a maioria (54,2%) apresentava comportamento anormal e transtorno psiquiátrico. Aproximadamente 42% apresentavam um estado amnésico e 37,5% amnésia mais conduta anormal e transtorno psiquiátrico. Todos acabaram por desenvolver um distúrbio comportamental ou psiquiátrico. Desorientação, disfunção executiva, afasia, agnosia e apraxia foram incomuns no início. A doença de Alzheimer (AD) foi o diagnóstico clínico inicial em 89% e o diagnóstico clínico final em 75%. A maior parte dos casos mostrou atrofia e perda neuronal localizada no hipocampo, amígadale cortex entorrinal. CONCLUSÃO: HSD apresenta características patológicas de DFT e as características clínicas que mimetizam AD. Os resultados, colocadas no contexto do trabalho anterior, suporta a ideia de que HSD pertence à família FTD, em que pode ser identificada como uma variante amnésica.


Assuntos
Humanos , Esclerose , Demência Frontotemporal , Neuropsiquiatria , Testes de Estado Mental e Demência
20.
Mov Disord ; 27(11): 1379-86, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22975850

RESUMO

Huntington's disease (HD) is characterized clinically by chorea, motor impairment, psychiatric manifestations, and dementia. Atrophy of the striatum is the neuropathological hallmark of HD, and previous studies have suggested that striatal atrophy correlates more closely with motor impairment than with chorea. Motor impairment, as measured by motor impairment score, correlates with functional disability in HD patients, but chorea does not. In this study, we investigated the relation between neuronal loss and these motor features. We conducted neuropathological and stereologic assessments of neurons in putamen and subthalamic nuclei in HD patients and age-matched controls. In putamen, we estimated the total number and volume of medium spiny neurons labeled with dopamine- and cAMP-regulated phosphoprotein 32 kDa (DARPP-32). In subthalamic nuclei, we estimated the total number of neurons on hematoxylin & eosin/luxol fast blue stains. In putamen of HD, immunohistochemistry showed DARPP-32 neuronal atrophy with extensive disruption of neurites and neuropil; stereologic studies found significant decreases in both the number and size of DARPP-32 neurons; we also detected a significant reduction of overall putamen volume in HD patients, compared to controls. In subthalamic nuclei, there was a mild, but significant, neuronal loss in the HD group. The loss of neurons in putamen and subthalamic nuclei as well as putaminal atrophy were significantly correlated with severity of motor impairment, but not with chorea. Our findings suggest that neuronal loss and atrophy in striatum and neuronal loss in subthalamic nuclei contribute specifically to the motor impairment of HD, but not to chorea.


Assuntos
Corpo Estriado/patologia , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Atividade Motora/fisiologia , Neurônios/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Autopsia , Estudos de Casos e Controles , Morte Celular , Tamanho Celular , Avaliação da Deficiência , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Neurônios/fisiologia , Escalas de Graduação Psiquiátrica , Estatística como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA