Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 29(11): 1853-1863, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-31960911

RESUMO

CADASIL is a vascular protein aggregation disorder caused by cysteine-altering NOTCH3 variants, leading to mid-adult-onset stroke and dementia. Here, we report individuals with a cysteine-altering NOTCH3 variant that induces exon 9 skipping, mimicking therapeutic NOTCH3 cysteine correction. The index came to our attention after a coincidental finding on a commercial screening MRI, revealing white matter hyperintensities. A heterozygous NOTCH3 c.1492G>T, p.Gly498Cys variant, was identified using a gene panel, which was also present in four first- and second-degree relatives. Although some degree of white matter hyperintensities was present on MRI in all family members with the NOTCH3 variant, the CADASIL phenotype was mild, as none had lacunes on MRI and there was no disability or cognitive impairment above the age of 60 years. RT-PCR and Sanger sequencing analysis on patient fibroblast RNA revealed that exon 9 was absent from the majority of NOTCH3 transcripts of the mutant allele, effectively excluding the mutation. NOTCH3 aggregation was assessed in skin biopsies using electron microscopy and immunohistochemistry and did not show granular osmiophilic material and only very mild NOTCH3 staining. For purposes of therapeutic translatability, we show that, in cell models, exon 9 exclusion can be obtained using antisense-mediated exon skipping and CRISPR/Cas9-mediated genome editing. In conclusion, this study provides the first in-human evidence that cysteine corrective NOTCH3 exon skipping is associated with less NOTCH3 aggregation and an attenuated phenotype, justifying further therapeutic development of NOTCH3 cysteine correction for CADASIL.


Assuntos
CADASIL/genética , Cisteína/genética , Agregação Patológica de Proteínas/genética , Receptor Notch3/genética , Substância Branca/metabolismo , Adulto , Idoso , Biópsia , CADASIL/diagnóstico por imagem , CADASIL/metabolismo , CADASIL/fisiopatologia , Sistemas CRISPR-Cas/genética , Éxons/genética , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Agregação Patológica de Proteínas/diagnóstico por imagem , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Índice de Gravidade de Doença , Pele/química , Pele/diagnóstico por imagem , Substância Branca/diagnóstico por imagem , Substância Branca/patologia
2.
Int J Mol Sci ; 22(9)2021 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-33926107

RESUMO

Amyloid ß-peptide (Aß) oligomerization is believed to contribute to the neuronal dysfunction in Alzheimer disease (AD). Despite decades of research, many details of Aß oligomerization in neurons still need to be revealed. Förster resonance energy transfer (FRET) is a simple but effective way to study molecular interactions. Here, we used a confocal microscope with a sensitive Airyscan detector for FRET detection. By live cell FRET imaging, we detected Aß42 oligomerization in primary neurons. The neurons were incubated with fluorescently labeled Aß42 in the cell culture medium for 24 h. Aß42 were internalized and oligomerized in the lysosomes/late endosomes in a concentration-dependent manner. Both the cellular uptake and intracellular oligomerization of Aß42 were significantly higher than for Aß40. These findings provide a better understanding of Aß42 oligomerization in neurons.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/fisiopatologia , Doença de Alzheimer , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide , Animais , Endossomos/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Hipocampo/diagnóstico por imagem , Hipocampo/metabolismo , Humanos , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Células PC12 , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Agregação Patológica de Proteínas/diagnóstico por imagem , Ratos
3.
Neurobiol Dis ; 117: 211-216, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29902557

RESUMO

OBJECTIVE: Our aim was to assess with positron emission tomography (PET) the temporal and spatial inter-relationships between levels of cortical microglial activation and the aggregated amyloid-ß and tau load in mild cognitive impairment (MCI) and early Alzheimer's disease (AD). METHODS: Six clinically probable AD and 20 MCI subjects had inflammation (11C-(R)-PK11195), amyloid (11C-PiB) and tau (18F-flortaucipir) PET, magnetic resonance imaging (MRI) and a neuropsychological assessment. Parametric images of tracer binding were interrogated at a voxel level and by region of interest analyses. RESULTS: 55% of MCI and 83% of AD subjects had a high amyloid-ß load. We have previously reported that clusters of correlated amyloid and inflammation levels are present in cortex. Here we found no correlation between levels of inflammation (11C-(R)-PK11195 BPND) and tau (18F-flortaucipir SUVR) or MMSE scores in high amyloid-ß cases. INTERPRETATION: While correlated levels of amyloid-ß and inflammation can be seen in MCI, we did not detect an association between levels of cortical tau tangles and inflammation in our series of high amyloid-ß cases. High levels of inflammation could be seen in amyloid-ß positive MCI cases where 18F-flortaucipir signals were low suggesting microglial activation precedes tau tangle formation. Inflammation levels were higher in high amyloid-ß MCI than in early AD cases, compatible with it initially playing a protective role.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Agregação Patológica de Proteínas/diagnóstico por imagem , Agregação Patológica de Proteínas/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Disfunção Cognitiva/diagnóstico por imagem , Disfunção Cognitiva/metabolismo , Estudos Transversais , Diagnóstico Precoce , Feminino , Humanos , Inflamação/diagnóstico por imagem , Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade
4.
Anal Chem ; 90(14): 8576-8582, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29902376

RESUMO

To optimize the lipophilicity and improve in vivo pharmacokinetics of near-infrared probes targeted Aß plaques, we designed, synthesized, and evaluated a series of polyethylene glycol modified probes with hydroxyl and methoxyl terminals. The relationships between chemical structure and optical, biological properties were systemically elucidated. The results indicated that a desired Aß probe should keep a balance among molecular rigidity, size, and lipophilicity. Probe 12d displayed improved properties including intense and selective response to Aß1-42 aggregates ( Kd = 7.3 nM, 22-fold fluorescence enhancement and emission maxima at 715 nm upon interaction with Aß1-42 aggregates), sufficient blood-brain barrier penetration (3.04% ID/g), and fast wash out from the brain (brain2 min/brain60 min = 10.1). Clear fluorescence signals retention in transgenic mice than control mice in in vivo near-infrared imaging. Hence, polyethylene glycol modified probes retained favorable optical properties but displayed great improvement of biological properties for Aß detection.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Peptídeos beta-Amiloides/análise , Corantes Fluorescentes/química , Fragmentos de Peptídeos/análise , Polietilenoglicóis/química , Agregação Patológica de Proteínas/diagnóstico por imagem , Animais , Feminino , Corantes Fluorescentes/farmacocinética , Humanos , Raios Infravermelhos , Masculino , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Imagem Óptica/métodos , Polietilenoglicóis/farmacocinética , Agregados Proteicos
5.
Chembiochem ; 19(18): 1944-1948, 2018 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-29953718

RESUMO

Oligomeric amyloid structures are crucial therapeutic targets in Alzheimer's and other amyloid diseases. However, these oligomers are too small to be resolved by standard light microscopy. We have developed a simple and versatile tool to image amyloid structures by using thioflavin T without the need for covalent labeling or immunostaining. The dynamic binding of single dye molecules generates photon bursts that are used for fluorophore localization on a nanometer scale. Thus, photobleaching cannot degrade image quality, allowing for extended observation times. Super-resolution transient amyloid binding microscopy promises to directly image native amyloid by using standard probes and record amyloid dynamics over minutes to days. We imaged amyloid fibrils from multiple polypeptides, oligomeric, and fibrillar structures formed during different stages of amyloid-ß aggregation, as well as the structural remodeling of amyloid-ß fibrils by the compound epi-gallocatechin gallate.


Assuntos
Peptídeos beta-Amiloides/análise , Amiloide/análise , Benzotiazóis/análise , Corantes Fluorescentes/análise , Imagem Óptica/métodos , Agregação Patológica de Proteínas/diagnóstico por imagem , Amiloide/ultraestrutura , Peptídeos beta-Amiloides/ultraestrutura , Desenho de Equipamento , Humanos , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Imagem Óptica/instrumentação , Agregados Proteicos , Agregação Patológica de Proteínas/patologia
7.
J Labelled Comp Radiopharm ; 57(4): 323-31, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24327420

RESUMO

Alzheimer's disease (AD) and related dementias show increasing clinical prevalence, yet our understanding of the etiology and pathobiology of disease-related neurodegeneration remains limited. In this regard, noninvasive imaging with radiotracers for positron emission tomography (PET) presents a unique tool for quantifying spatial and temporal changes in characteristic biological markers of brain disease and for assessing potential drug efficacy. PET radiotracers targeting different protein markers are being developed to address questions pertaining to the molecular and/or genetic heterogeneity of AD and related dementias. For example, radiotracers including [(11) C]-PiB and [(18) F]-AV-45 (Florbetapir) are being used to measure the density of Aß-plaques in AD patients and to interrogate the biological mechanisms of disease initiation and progression. Our focus is on the development of novel PET imaging agents, targeting proteins beyond Aß-plaques, which can be used to investigate the broader mechanism of AD pathogenesis. Here, we present the chemical basis of various radiotracers which show promise in preclinical or clinical studies for use in evaluating the phenotypic or biochemical characteristics of AD. Radiotracers for PET imaging neuroinflammation, metal ion association with Aß-plaques, tau protein, cholinergic and cannabinoid receptors, and enzymes including glycogen-synthase kinase-3ß and monoamine oxidase B amongst others, and their connection to AD are highlighted.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Traçadores Radioativos , Doença de Alzheimer/enzimologia , Doença de Alzheimer/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Humanos , Agregação Patológica de Proteínas/diagnóstico por imagem
8.
STAR Protoc ; 2(1): 100372, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33733241

RESUMO

The accumulation of proteins into insoluble aggregates is a common feature of several neurodegenerative diseases. Aggregated α-synuclein is a major component of Lewy bodies that pathologically define Parkinson's disease (PD). Here, we present methods for the detection of pathogenic conformations of α-synuclein in induced pluripotent stem cell (iPSC) patient-derived neuron models and brain tissue. These methods can be applied to studies of PD pathogenesis and the discovery of novel therapeutics that restore physiological α-synuclein. For complete details on the use and execution of this protocol, please refer to Cuddy et al. (2019) and Zunke et al. (2018).


Assuntos
Agregação Patológica de Proteínas/diagnóstico por imagem , alfa-Sinucleína/análise , alfa-Sinucleína/isolamento & purificação , Cromatografia em Gel/métodos , Imunofluorescência/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/citologia , alfa-Sinucleína/metabolismo
9.
Yakugaku Zasshi ; 141(6): 825-829, 2021.
Artigo em Japonês | MEDLINE | ID: mdl-34078789

RESUMO

The formation of neurotoxic aggregates by amyloid-ß peptide (Aß) is considered to be a key step in the onset of Alzheimer's disease. It is widely accepted that oligomers are more neurotoxic than amyloid fibrils in the aqueous-phase aggregation of Aß. Membrane-mediated amyloidogenesis is also relevant to the pathology, although the relationship between the aggregate size and cytotoxicity has remained elusive. Fluorescence correlation spectroscopy (FCS) is a sensitive method to monitor molecular aggregation processes by measuring diffusion of fluorophore-labeled molecules. Here, we monitored the initial aggregation process of Aß on cell membranes of SH-SY5Y neuroblastoma cells. For example, aggregation of Aß-(1-42) was evaluated by using Aß-(1-42) (5 µM) doped with fluorophore-Aß-(1-42) (10 nM). A membrane-bound Aß component was detected in FCS autocorrelation curve after 1-h incubation of the Aßs with the cells. Following incubation for ~10 h, Aß-(1-42) formed oligomers composed of ~10 Aß molecules. These Aß oligomers formed on membranes did not induce activation of caspase-3, an effector caspase for apoptosis, therefore were not neurotoxic, in contrast to reported Aß oligomers prepared in aqueous phase. Formation of larger Aß fibrils on membranes was found to be critical for Aß neurotoxicity. We also report that trace amounts of pyroglutaminated Aß-(3-42), a minor species of Aß, can enhance initial aggregation process of Aß-(1-42) on the cell membranes. These results indicate the usefulness of FCS detection of small Aß oligomers formed on cell surface, which can act as pathogenic seeds for amyloid fibrils responsible for neurotoxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Membrana Celular/metabolismo , Agregados Proteicos , Agregação Patológica de Proteínas/diagnóstico por imagem , Espectrometria de Fluorescência/métodos , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Linhagem Celular Tumoral , Humanos , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia
10.
Sci Rep ; 11(1): 17977, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504195

RESUMO

Huntington's disease (HD) is caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin (HTT) gene coding for the huntingtin (HTT) protein. The misfolding and consequential aggregation of CAG-expanded mutant HTT (mHTT) underpin HD pathology. Our interest in the life cycle of HTT led us to consider the development of high-affinity small-molecule binders of HTT oligomerized/amyloid-containing species that could serve as either cellular and in vivo imaging tools or potential therapeutic agents. We recently reported the development of PET tracers CHDI-180 and CHDI-626 as suitable for imaging mHTT aggregates, and here we present an in-depth pharmacological investigation of their binding characteristics. We have implemented an array of in vitro and ex vivo radiometric binding assays using recombinant HTT, brain homogenate-derived HTT aggregates, and brain sections from mouse HD models and humans post-mortem to investigate binding affinities and selectivity against other pathological proteins from indications such as Alzheimer's disease and spinocerebellar ataxia 1. Radioligand binding assays and autoradiography studies using brain homogenates and tissue sections from HD mouse models showed that CHDI-180 and CHDI-626 specifically bind mHTT aggregates that accumulate with age and disease progression. Finally, we characterized CHDI-180 and CHDI-626 regarding their off-target selectivity and binding affinity to beta amyloid plaques in brain sections and homogenates from Alzheimer's disease patients.


Assuntos
Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Agregados Proteicos/genética , Agregação Patológica de Proteínas/diagnóstico por imagem , Compostos Radiofarmacêuticos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Autorradiografia/métodos , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Proteína Huntingtina/genética , Doença de Huntington/patologia , Imuno-Histoquímica/métodos , Camundongos , Camundongos Transgênicos , Radioisótopos de Nitrogênio/metabolismo , Traçadores Radioativos , Ensaio Radioligante/métodos , Proteínas Recombinantes/metabolismo
11.
J Med Chem ; 63(15): 8608-8633, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32662649

RESUMO

Mutant huntingtin (mHTT) protein carrying the elongated N-terminal polyglutamine (polyQ) tract misfolds and forms protein aggregates characteristic of Huntington's disease (HD) pathology. A high-affinity ligand specific for mHTT aggregates could serve as a positron emission tomography (PET) imaging biomarker for HD therapeutic development and disease progression. To identify such compounds with binding affinity for polyQ aggregates, we embarked on systematic structural activity studies; lead optimization of aggregate-binding affinity, unbound fractions in brain, permeability, and low efflux culminated in the discovery of compound 1, which exhibited target engagement in autoradiography (ARG) studies in brain slices from HD mouse models and postmortem human HD samples. PET imaging studies with 11C-labeled 1 in both HD mice and WT nonhuman primates (NHPs) demonstrated that the right-hand-side labeled ligand [11C]-1R (CHDI-180R) is a suitable PET tracer for imaging of mHTT aggregates. [11C]-1R is now being advanced to human trials as a first-in-class HD PET radiotracer.


Assuntos
Proteína Huntingtina/análise , Doença de Huntington/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Agregação Patológica de Proteínas/diagnóstico por imagem , Animais , Modelos Animais de Doenças , Cães , Feminino , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Ligantes , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Peptídeos/genética , Agregação Patológica de Proteínas/genética , Compostos Radiofarmacêuticos/análise , Ratos Sprague-Dawley
12.
Sci Rep ; 10(1): 9742, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32546691

RESUMO

Alzheimer's disease (AD) is a progressive disorder of the brain that gradually decreases thinking, memory, and language abilities. The aggregation process of amyloid ß (Aß) is a key step in the expression of its neurocytotoxicity and development of AD because Aß aggregation and accumulation around neuronal cells induces cell death. However, the molecular mechanism underlying the neurocytotoxicity and cell death by Aß aggregation has not been clearly elucidated. In this study, we successfully visualized real-time process of Aß42 aggregation around living cells by applying our established QD imaging method. 3D observations using confocal laser microscopy revealed that Aß42 preferentially started to aggregate at the region where membrane protrusions frequently formed. Furthermore, we found that inhibition of actin polymerization using cytochalasin D reduced aggregation of Aß42 on the cell surface. These results indicate that actin polymerization-dependent cell motility is responsible for the promotion of Aß42 aggregation at the cell periphery. 3D observation also revealed that the aggregates around the cell remained in that location even if cell death occurred, implying that amyloid plaques found in the AD brain grew from the debris of dead cells that accumulated Aß42 aggregates.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Microscopia Confocal/métodos , Agregação Patológica de Proteínas/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Peptídeos beta-Amiloides/fisiologia , Animais , Encéfalo/metabolismo , Imageamento Tridimensional/métodos , Memória/fisiologia , Neurônios/metabolismo , Células PC12 , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Agregação Patológica de Proteínas/fisiopatologia , Ratos
13.
Int J Pharm ; 573: 118814, 2020 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-31759101

RESUMO

Dendrimers are globular structures, presenting an initiator core, repetitive layers starting radially from the core and terminal groups on the surface, resembling tree architecture. These structures have been studied in many biological applications, as drug, DNA, RNA and proteins delivery, as well as imaging and radiocontrast agents. With reference to that, this review focused in providing examples of dendrimers used in nanomedicine. Although most studies emphasize cancer, there are others which reveal action in the neurosystem, reducing either neuroinflammation or protein aggregation. Dendrimers can carry bioactive compounds by covalent bond (dendrimer prodrug), or by ionic interaction or adsortion in the internal space of the nanostructure. Additionally, dendrimers can be associated with other polymers, as PEG (polyethylene glycol), and with targeting structures as aptamers, antibodies, folic acid and carbohydrates. Their products in preclinical/clinical trial and those in the market are also discussed, with a total of six derivatives in clinical trials and seven products available in the market.


Assuntos
Dendrímeros/administração & dosagem , Dendrímeros/química , Nanomedicina/métodos , Nanoestruturas/química , Antineoplásicos/administração & dosagem , Química Farmacêutica , Ensaios Clínicos como Assunto , Meios de Contraste/administração & dosagem , Meios de Contraste/química , Humanos , Nanoestruturas/administração & dosagem , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Doenças do Sistema Nervoso/diagnóstico por imagem , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/imunologia , Polietilenoglicóis/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Agregação Patológica de Proteínas/diagnóstico por imagem , Agregação Patológica de Proteínas/tratamento farmacológico
14.
Nat Commun ; 11(1): 411, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31964863

RESUMO

Alzheimer's disease (AD) is characterized by amyloid plaques and progressive cerebral atrophy. Here, we report FAM222A as a putative brain atrophy susceptibility gene. Our cross-phenotype association analysis of imaging genetics indicates a potential link between FAM222A and AD-related regional brain atrophy. The protein encoded by FAM222A is predominantly expressed in the CNS and is increased in brains of patients with AD and in an AD mouse model. It accumulates within amyloid deposits, physically interacts with amyloid-ß (Aß) via its N-terminal Aß binding domain, and facilitates Aß aggregation. Intracerebroventricular infusion or forced expression of this protein exacerbates neuroinflammation and cognitive dysfunction in an AD mouse model whereas ablation of this protein suppresses the formation of amyloid deposits, neuroinflammation and cognitive deficits in the AD mouse model. Our data support the pathological relevance of protein encoded by FAM222A in AD.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/genética , Disfunção Cognitiva/genética , Proteínas do Tecido Nervoso/genética , Placa Amiloide/genética , Agregação Patológica de Proteínas/genética , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/patologia , Proteínas Amiloidogênicas/metabolismo , Animais , Atrofia/diagnóstico por imagem , Atrofia/genética , Atrofia/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Disfunção Cognitiva/patologia , Conjuntos de Dados como Assunto , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Placa Amiloide/patologia , Polimorfismo de Nucleotídeo Único , Agregação Patológica de Proteínas/diagnóstico por imagem , Agregação Patológica de Proteínas/patologia
15.
J Med Chem ; 62(14): 6694-6704, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31274302

RESUMO

The early noninvasive diagnosis of Alzheimer's disease targeted ß-amyloid (Aß) plaques or Tau tangles is a major challenge because of the coshared ß-sheet structure of the target. In contrast to tailoring probes to specific amyloids, here, we showed that near-infrared (NIR) environment-sensitive probe 18 could fluorescently discriminate Aß and Tau from artificial aggregates to pathological change in the brain tissue. The biological evaluation demonstrated that the substantial fluorescence enhancement, large blueshift in the emission upon interactions with the aggregates, and the high binding affinity significantly contributed to the fluorescent discrimination. A simplified Ooshika-Lippert-Mataga equation provided an effective means of correlating 18 with the static relative permittivity (ε0) of proteins, elucidating the origin of the distinction capabilities, and quantitatively estimating the dielectric properties of proteins. Moreover, 18 possessed high bioavailability, including sufficient blood-brain barrier penetration, in vivo NIR imaging, and ex vivo histology in living mice.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Peptídeos beta-Amiloides/análise , Encéfalo/diagnóstico por imagem , Corantes Fluorescentes/química , Proteínas tau/análise , Amiloide/análise , Animais , Feminino , Masculino , Camundongos Endogâmicos C57BL , Imagem Óptica/métodos , Agregação Patológica de Proteínas/diagnóstico por imagem , Espectroscopia de Luz Próxima ao Infravermelho/métodos
16.
ACS Chem Neurosci ; 10(3): 1240-1250, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30346718

RESUMO

There is now crucial medical importance placed on understanding the role of early stage, subvisible protein aggregation, particularly in neurodegenerative disease. While there are strategies for detecting such aggregates in vitro, there is no approach at present that can detect these toxic species associated with cells and specific subcellular compartments. We have exploited excitation-energy-dependent fluorescence edge-shift of recombinant protein labeled with a molecular beacon, to provide a sensitive read out for the presence of subvisible protein aggregates. To demonstrate the potential utility of the approach, we examine the major peptide associated with the initiation of Alzheimer's disease, amyloid ß-protein (Aß) at a patho-physiologically relevant concentration in mouse cortical neurons. Using our approach, we find preliminary evidence that subvisible Aß aggregates are detected at specific subcellular regions and that neurons drive the formation of specific Aß aggregate conformations. These findings therefore demonstrate the potential of a novel fluorescence-based approach for detecting and imaging protein aggregates in a cellular context, which can be used to sensitively probe the association of early stage toxic protein aggregates within subcellular compartments.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/metabolismo , Neurônios/metabolismo , Agregação Patológica de Proteínas/diagnóstico por imagem , Agregação Patológica de Proteínas/metabolismo , Animais , Células Cultivadas , Difusão Dinâmica da Luz , Camundongos , Microscopia de Fluorescência , Imagem Molecular , Análise Espectral
17.
Acta Neuropathol Commun ; 7(1): 171, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31703739

RESUMO

The detection of amyloid beta deposits and neurofibrillary tangles, both hallmarks of Alzheimer's disease (AD), is key to understanding the mechanisms underlying these pathologies. Luminescent conjugated oligothiophenes (LCOs) enable fluorescence imaging of these protein aggregates. Using LCOs and multiphoton microscopy, individual tangles and amyloid beta deposits were labeled in vivo and imaged longitudinally in a mouse model of tauopathy and cerebral amyloidosis, respectively. Importantly, LCO HS-84, whose emission falls in the green region of the spectrum, allowed for the first time longitudinal imaging of tangle dynamics following a single intravenous injection. In addition, LCO HS-169, whose emission falls in the red region of the spectrum, successfully labeled amyloid beta deposits, allowing multiplexing with other reporters whose emission falls in the green region of the spectrum. In conclusion, this method can provide a new approach for longitudinal in vivo imaging using multiphoton microscopy of AD pathologies as well as other neurodegenerative diseases associated with protein aggregation in mouse models.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Angiopatia Amiloide Cerebral/diagnóstico por imagem , Angiopatia Amiloide Cerebral/patologia , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Emaranhados Neurofibrilares/patologia , Tauopatias/diagnóstico por imagem , Tauopatias/patologia , Animais , Encéfalo/diagnóstico por imagem , Modelos Animais de Doenças , Feminino , Medições Luminescentes , Masculino , Camundongos Transgênicos , Agregação Patológica de Proteínas/diagnóstico por imagem , Proteínas tau/metabolismo
18.
Eur J Med Chem ; 181: 111585, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31404860

RESUMO

Brain amyloid deposits have been identified as the main neuropathological hallmarks of Alzheimer's diseases (AD) and intensive efforts have been devoted to develop aggregation inhibitors preventing the formation of toxic oligomeric Aß for therapeutic. In addition, evidence indicates that the formation and accumulation of ß-amyloid plaques probably precede clinical symptoms by around 20 years and imaging of such plaques would be beneficial for early-stage AD detection. In this study, we investigated phenothiazine-based compounds as novel promising theranostic agents for AD. These multifunctional agents exhibited BBB permeability, low neurotoxicity, good bio-stability as well as strong turn-on fluorescence with a Stokes shift upon binding to Aß aggregates. They had metal-chelating property which could delay Aß aggregation and displayed high binding affinity for ß-amyloid aggregates. Moreover, they have been simultaneously applied to perform in vivo near-infrared fluorescence imaging of ß-amyloid plaques in double transgenic AD mouse model, to prevent self-aggregation of Aß monomer from forming toxic oligomers and to protect human neuroblastoma SH-SY5Y cells against Aß-induced toxicity and oxidative stress.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/análise , Fármacos Neuroprotetores/uso terapêutico , Fenotiazinas/uso terapêutico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacocinética , Imagem Óptica , Fenotiazinas/química , Fenotiazinas/farmacocinética , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/tratamento farmacológico , Agregação Patológica de Proteínas/diagnóstico por imagem , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/prevenção & controle , Nanomedicina Teranóstica
19.
ACS Chem Neurosci ; 9(7): 1757-1767, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-29630333

RESUMO

Using the recently reported cryo-EM structure for the tau fibril [ Fitzpatrick et al. (2017) Nature 547, 185-190 ], which is a potential target concerning Alzheimer's disease, we present the first molecular modeling studies on its interaction with various positron emission tomography (PET) tracers. Experimentally, based on the binding assay studies, at least three different high-affinity binding sites have been reported for tracers in the tau fibril. Herein, through integrated modeling using molecular docking, molecular dynamics, and binding free energy calculations, we provide insight into the binding patterns of various tracers to the tau fibril. We suggest that there are four different high-affinity binding sites available for many of the studied tracers showing varying binding affinity to different binding sites. Thus, PBB3 binds most strongly to site 4, and interestingly, this site is not a preferable site for any other tracers. For THK5351, our data show that it strongly binds to sites 3 and 1, the former one being more preferable. We also find that MK6240 and T807 bind to site 1 specifically. The modeling data also give some insight into whether a tracer bound to a specific site can be replaced by others or not. For example, the displacement of T807 by PBB3 as reported experimentally can also be explained and attributed to the larger binding affinity of the latter compound in all binding sites. The binding free energy results explain very well the small binding affinity of THK523 compared to all the aryl quinoline moieties containing THK tracers. The ability of certain tau tracers, like FDDNP and THK523, to bind to amyloid fibrils has also been investigated. Furthermore, such off-target interaction of tau tracers with amyloid beta fibrils has been validated using a quantum mechanical fragmentation approach.


Assuntos
Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Sítios de Ligação , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Agregação Patológica de Proteínas/diagnóstico por imagem , Agregação Patológica de Proteínas/metabolismo , Multimerização Proteica , Estrutura Secundária de Proteína , Compostos Radiofarmacêuticos/química , Proteínas tau/química
20.
Sci Rep ; 8(1): 6950, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29725045

RESUMO

The deposition of insoluble protein aggregates in the brain is a hallmark of many neurodegenerative diseases. While their exact role in neurodegeneration remains unclear, the presence of these amyloid deposits often precedes clinical symptoms. As a result, recent progress in imaging methods that utilize amyloid-specific small molecule probes have become a promising avenue for antemortem disease diagnosis. Here, we present a series of amino-aryl cyanoacrylate (AACA) fluorophores that show a turn-on fluorescence signal upon binding to amyloids in solution and in tissue. Using a theoretical model for environmental sensitivity of fluorescence together with ab initio computational modeling of the effects of polar environment on electron density distribution and conformational dynamics, we designed, synthesized, and evaluated a set of fluorophores that (1) bind to aggregated forms of Alzheimer's-related ß-amyloid peptides with low micromolar to high nanomolar affinities and (2) have the capability to fluorescently discriminate different amyloids based on differences in amino acid composition within the binding pocket through exploitation of their solvatochromic properties. These studies showcase the rational design of a family of amyloid-binding imaging agents that could be integrated with new optical approaches for the clinical diagnosis of amyloidoses, where accurate identification of the specific neurodegenerative disease could aid in the selection of a proper course for treatment.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Amiloide/análise , Cianoacrilatos/química , Corantes Fluorescentes/química , Imagem Óptica/métodos , Agregação Patológica de Proteínas/diagnóstico por imagem , Acilação , Aminação , Peptídeos beta-Amiloides/análise , Encéfalo/diagnóstico por imagem , Cianoacrilatos/síntese química , Corantes Fluorescentes/síntese química , Humanos , Fragmentos de Peptídeos/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA