Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Nucl Med ; 60(1): 100-106, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29777003

RESUMO

SUV ratios (SUVRs) are commonly used to quantify tracer uptake in amyloid-ß PET. Here, we explore the impact of target and reference region-of-interest (ROI) selection on SUVR effect sizes using interventional data from the ongoing phase 1b PRIME study (NCT01677572) of aducanumab (BIIB037) in patients with prodromal or mild Alzheimer disease. Methods: The florbetapir PET SUVR was calculated at baseline (screening) and at weeks 26 and 54 for patients randomized to receive placebo and each of 4 aducanumab doses (1, 3, 6, and 10 mg/kg) using the whole cerebellum, cerebellar gray matter, cerebellar white matter, pons, and subcortical white matter as reference regions. In addition to the prespecified composite cortex target ROI, individual cerebral cortical ROIs were assessed as targets. Results: Of the reference regions used, subcortical white matter, cerebellar white matter, and the pons, alone or in combination, generated the largest effect sizes. The use of the anterior cingulate cortex as a target ROI resulted in larger effect sizes than the use of the composite cortex. SUVR calculations were not affected by correction for brain volume changes over time. Conclusion: Dose- and time-dependent reductions in the amyloid PET SUVR were consistently observed with aducanumab only in cortical regions prone to amyloid plaque deposition, regardless of the reference region used. These data support the hypothesis that florbetapir SUVR responses associated with aducanumab treatment are a result of specific dose- and time-dependent reductions in the amyloid burden in patients with Alzheimer disease.


Assuntos
Amiloide/metabolismo , Anticorpos Monoclonais Humanizados/metabolismo , Tomografia por Emissão de Pósitrons/normas , Adulto , Transporte Biológico , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Padrões de Referência
2.
J Med Chem ; 61(4): 1737-1743, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29257860

RESUMO

Changes in expression and dysfunctional signaling of TrkA/B/C receptors and oncogenic Trk fusion proteins are found in neurological diseases and cancers. Here, we describe the development of a first 18F-labeled optimized lead suitable for in vivo imaging of Trk, [18F]TRACK, which is radiosynthesized with ease from a nonactivated aryl precursor concurrently combining largely reduced P-gp liability and improved brain kinetics compared to previous leads while displaying high on-target affinity and human kinome selectivity.


Assuntos
Radioisótopos de Flúor , Tomografia por Emissão de Pósitrons/métodos , Receptor trkA/antagonistas & inibidores , Desenho de Fármacos , Humanos , Neoplasias/diagnóstico por imagem , Doenças Neurodegenerativas/diagnóstico por imagem , Inibidores de Proteínas Quinases
3.
Neuroimage Clin ; 16: 222-233, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28794981

RESUMO

In this work, we propose a diffusion MRI protocol for mining Parkinson's disease diffusion MRI datasets and recover robust disease-specific biomarkers. Using advanced high angular resolution diffusion imaging (HARDI) crossing fiber modeling and tractography robust to partial volume effects, we automatically dissected 50 white matter (WM) fascicles. These fascicles connect deep nuclei (thalamus, putamen, pallidum) to different cortical functional areas (associative, motor, sensorimotor, limbic), basal forebrain and substantia nigra. Then, among these 50 candidate WM fascicles, only the ones that passed a test-retest reproducibility procedure qualified for further tractometry analysis. Leveraging the unique 2-timepoints test-retest Parkinson's Progression Markers Initiative (PPMI) dataset of over 600 subjects, we found statistically significant differences in tract profiles along the subcortico-cortical pathways between Parkinson's disease patients and healthy controls. In particular, significant increases in FA, apparent fiber density, tract-density and generalized FA were detected in some locations of the nigro-subthalamo-putaminal-thalamo-cortical pathway. This connection is one of the major motor circuits balancing the coordination of motor output. Detailed and quantifiable knowledge on WM fascicles in these areas is thus essential to improve the quality and outcome of Deep Brain Stimulation, and to target new WM locations for investigation.


Assuntos
Encéfalo/patologia , Imagem de Difusão por Ressonância Magnética/métodos , Doença de Parkinson/diagnóstico por imagem , Doença de Parkinson/patologia , Substância Branca/patologia , Biomarcadores , Encéfalo/diagnóstico por imagem , Mineração de Dados , Bases de Dados Factuais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Vias Neurais/diagnóstico por imagem , Vias Neurais/patologia , Reprodutibilidade dos Testes , Substância Branca/diagnóstico por imagem
4.
J Med Chem ; 60(16): 6897-6910, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28696690

RESUMO

The proto-oncogenes NTRK1/2/3 encode the tropomyosin receptor kinases TrkA/B/C which play pivotal roles in neurobiology and cancer. We describe herein the discovery of [11C]-(R)-3 ([11C]-(R)-IPMICF16), a first-in-class positron emission tomography (PET) TrkB/C-targeting radiolabeled kinase inhibitor lead. Relying on extensive human kinome vetting, we show that (R)-3 is the most potent and most selective TrkB/C inhibitor characterized to date. It is demonstrated that [11C]-(R)-3 readily crosses the blood-brain barrier (BBB) in rodents and selectively binds to TrkB/C receptors in vivo, as evidenced by entrectinib blocking studies. Substantial TrkB/C-specific binding in human brain tissue is observed in vitro, with specific reduction in the hippocampus of Alzheimer's disease (AD) versus healthy brains. We additionally provide preliminary translational data regarding the brain disposition of [11C]-(R)-3 in primates including first-in-human assessment. These results illustrate for the first time the use of a kinome-wide selective radioactive chemical probe for endogenous kinase PET neuroimaging in human.


Assuntos
Imidazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridazinas/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Receptor trkB/antagonistas & inibidores , Receptor trkC/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Doença de Alzheimer/diagnóstico por imagem , Animais , Benzamidas/farmacologia , Barreira Hematoencefálica/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Radioisótopos de Carbono , Cães , Humanos , Imidazóis/síntese química , Imidazóis/farmacocinética , Indazóis/farmacologia , Macaca mulatta , Células Madin Darby de Rim Canino , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Neuroimagem , Tomografia por Emissão de Pósitrons , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacocinética , Piridazinas/síntese química , Piridazinas/farmacocinética , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Ratos Sprague-Dawley , Receptor trkA/antagonistas & inibidores , Estereoisomerismo , Relação Estrutura-Atividade
5.
J Exp Med ; 212(10): 1529-49, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26347470

RESUMO

Inactivating mutations of the NF-κB essential modulator (NEMO), a key component of NF-κB signaling, cause the genetic disease incontinentia pigmenti (IP). This leads to severe neurological symptoms, but the mechanisms underlying brain involvement were unclear. Here, we show that selectively deleting Nemo or the upstream kinase Tak1 in brain endothelial cells resulted in death of endothelial cells, a rarefaction of brain microvessels, cerebral hypoperfusion, a disrupted blood-brain barrier (BBB), and epileptic seizures. TAK1 and NEMO protected the BBB by activating the transcription factor NF-κB and stabilizing the tight junction protein occludin. They also prevented brain endothelial cell death in a NF-κB-independent manner by reducing oxidative damage. Our data identify crucial functions of inflammatory TAK1-NEMO signaling in protecting the brain endothelium and maintaining normal brain function, thus explaining the neurological symptoms associated with IP.


Assuntos
Encéfalo/irrigação sanguínea , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Circulação Cerebrovascular/genética , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Epilepsia/genética , Feminino , Quinase I-kappa B/metabolismo , Incontinência Pigmentar/metabolismo , Incontinência Pigmentar/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , MAP Quinase Quinase Quinases/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ocludina/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Fator de Transcrição RelA/metabolismo
6.
J Nucl Med ; 56(9): 1351-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26135108

RESUMO

UNLABELLED: Classification of subjects on the basis of amyloid PET scans is increasingly being used in research studies and clinical practice. Although qualitative, visual assessment is currently the gold standard approach, automated classification techniques are inherently more reproducible and efficient. The objective of this work was to develop a statistical approach for the automated classification of subjects with different levels of cognitive impairment into a group with low amyloid levels (AßL) and a group with high amyloid levels (AßH) through the use of amyloid PET data from the Alzheimer Disease Neuroimaging Initiative study. METHODS: In our framework, an iterative, voxelwise, regularized discriminant analysis is combined with a receiver operating characteristic approach that optimizes the selection of a region of interest (ROI) and a cutoff value for the automated classification of subjects into the AßL and AßH groups. The robustness, spatial stability, and generalization of the resulting target ROIs were evaluated by use of the standardized uptake value ratio for (18)F-florbetapir PET images from subjects who served as healthy controls, subjects who had mild cognitive impairment, and subjects who had Alzheimer disease and were participating in the Alzheimer Disease Neuroimaging Initiative study. RESULTS: We determined that several iterations of the discriminant analysis improved the classification of subjects into the AßL and AßH groups. We found that an ROI consisting of the posterior cingulate cortex/precuneus and the medial frontal cortex yielded optimal group separation and showed good stability across different reference regions and cognitive cohorts. A key step in this process was the automated determination of the cutoff value for group separation, which was dependent on the reference region used for the standardized uptake value ratio calculation and which was shown to have a relatively narrow range across subject groups. CONCLUSION: We developed a data-driven approach for the determination of an optimal target ROI and an associated cutoff value for the separation of subjects into the AßL and AßH groups. Future work should include the application of this process to other datasets to facilitate the determination of the translatability of the optimal ROI obtained in this study to other populations. Ideally, the accuracy of our target ROI and cutoff value could be further validated with PET-autopsy data from large-scale studies. It is anticipated that this approach will be extremely useful for the enrichment of study populations in clinical trials involving putative disease-modifying therapeutic agents for Alzheimer disease.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Compostos de Anilina/farmacocinética , Etilenoglicóis/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Idoso , Algoritmos , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Feminino , Humanos , Aumento da Imagem/métodos , Interpretação de Imagem Assistida por Computador/métodos , Masculino , Compostos Radiofarmacêuticos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Índice de Gravidade de Doença , Distribuição Tecidual
7.
Biochem J ; 461(2): 205-12, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24800750

RESUMO

Hyperhomocysteinaemia can contribute to cognitive impairment and brain atrophy. MTRR (methionine synthase reductase) activates methionine synthase, which catalyses homocysteine remethylation to methionine. Severe MTRR deficiency results in homocystinuria with cognitive and motor impairments. An MTRR polymorphism may influence homocysteine levels and reproductive outcomes. The goal of the present study was to determine whether mild hyperhomocysteinaemia affects neurological function in a mouse model with Mtrr deficiency. Mtrr+/+, Mtrr+/gt and Mtrrgt/gt mice (3 months old) were assessed for short-term memory, brain volumes and hippocampal morphology. We also measured DNA methylation, apoptosis, neurogenesis, choline metabolites and expression of ChAT (choline acetyltransferase) and AChE (acetylcholinesterase) in the hippocampus. Mtrrgt/gt mice exhibited short-term memory impairment on two tasks. They had global DNA hypomethylation and decreased choline, betaine and acetylcholine levels. Expression of ChAT and AChE was increased and decreased respectively. At 3 weeks of age, they showed increased neurogenesis. In the cerebellum, mutant mice had DNA hypomethylation, decreased choline and increased expression of ChAT. Our work demonstrates that mild hyperhomocysteinaemia is associated with memory impairment. We propose a mechanism whereby a deficiency in methionine synthesis leads to hypomethylation and compensatory disturbances in choline metabolism in the hippocampus. This disturbance affects the levels of acetylcholine, a critical neurotransmitter in learning and memory.


Assuntos
Cerebelo/metabolismo , Ferredoxina-NADP Redutase/genética , Hipocampo/metabolismo , Homocistinúria/metabolismo , Hiper-Homocisteinemia/metabolismo , Memória de Curto Prazo , Acetilcolinesterase/genética , Acetilcolinesterase/metabolismo , Animais , Apoptose , Betaína/metabolismo , Cerebelo/patologia , Colina/metabolismo , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Metilação de DNA , Modelos Animais de Doenças , Ferredoxina-NADP Redutase/deficiência , Expressão Gênica , Hipocampo/patologia , Homocisteína/metabolismo , Homocistinúria/genética , Homocistinúria/patologia , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/patologia , Masculino , Metionina/metabolismo , Camundongos , Camundongos Knockout , Transmissão Sináptica
8.
Neurobiol Dis ; 54: 59-67, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23454197

RESUMO

Magnetic resonance imaging (MRI) studies have identified aberrant cortical structure in Alzheimer's disease (AD). The association between MRI-derived cortical morphometry measures and ß-amyloid, however, remains poorly understood. In this study, we explored the potential relationship between early alterations in cortical thickness and later stage ß-amyloid deposition, using a novel approach, in a transgenic AD mouse model. We acquired longitudinal anatomical MRI scans from mutant amyloid precursor protein (APP) transgenic mice and age-matched wild-type mice at 1 and 3.5months-of-age, and employed fully-automated image processing methods to derive objective, quantitative measures of cortical thickness on a region-of-interest basis. We also generated 3D quantitative immunohistochemistry (qIHC) volumes of deposited ß-amyloid burden from 18month-old transgenic mice using an automated, production-level process. These studies revealed thinner cortex in most regions in the 1month-old transgenic mice relative to age-matched wild-types, with the exception of the frontal, perirhinal/entorhinal, posterior cingulate, and retrosplenial cortical regions. Between 1 and 3.5months-of-age, the transgenic mice demonstrated stable or increasing cortical thickness, while the wild-type mice showed cortical thinning. Based on data from co-registered 3D MRI and qIHC volumes, we identified an association between abnormal, early, regional cortical thickness change over 2.5months and later ß-amyloid deposition. These observations suggest that the spatio-temporal pattern of early (pre-plaque) alterations in cerebral cortical structure is indicative of regional predisposition to later ß-amyloid pathology in a transgenic AD mouse model.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/patologia , Processamento de Imagem Assistida por Computador/métodos , Doença de Alzheimer/metabolismo , Animais , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos
9.
Neurobiol Aging ; 34(6): 1644-52, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23273599

RESUMO

Magnetic resonance imaging studies have revealed distinct patterns of cortical atrophy and hypoperfusion in patients with Alzheimer's disease. The relationship between these in vivo imaging measures and the corresponding underlying pathophysiological changes, however, remains elusive. Recently, attention has turned to neuroimaging of mouse models of Alzheimer's disease in which imaging-pathological correlations can be readily performed. In this study, anatomical and arterial spin labeling perfusion magnetic resonance imaging scans of amyloid precursor protein transgenic and age-matched wild-type mice were acquired at 3, 12, and 18 months of age. Fully-automated image processing methods were used to derive quantitative measures of cortical thickness and perfusion. These studies revealed increased regional cortical thickness in young transgenic mice relative to age-matched wild-type mice. However, the transgenic mice generally demonstrated a greater rate of cortical thinning over 15 months. Cortical perfusion was significantly reduced in young transgenic mice in comparison with wild-type mice across most brain regions. Previously unreported regional genotype differences and age-related changes in cortical thickness and cerebral perfusion were identified in amyloid precursor protein transgenic and wild-type mice.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/patologia , Animais , Atrofia , Estudos Transversais , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...