Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
NMR Biomed ; 37(2): e5048, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37798964

RESUMEN

Paravascular cerebrospinal fluid (pCSF) surrounding the cerebral arteries within the glymphatic system is pulsatile and moves in synchrony with the pressure waves of the vessel wall. Whether such pulsatile pCSF can infer pulse wave propagation-a property tightly related to arterial stiffness-is unknown and has never been explored. Our recently developed imaging technique, dynamic diffusion-weighted imaging (dynDWI), captures the pulsatile pCSF dynamics in vivo and can explore this question. In this work, we evaluated the time shifts between pCSF waves and finger pulse waves, where pCSF waves were measured by dynDWI and finger pulse waves were measured by the scanner's built-in finger pulse oximeter. We hypothesized that the time shifts reflect brain-finger pulse wave travel time and are sensitive to arterial stiffness. We applied the framework to 36 participants aged 18-82 years to study the age effect of travel time, as well as its associations with cognitive function within the older participants (N = 15, age > 60 years). Our results revealed a strong and consistent correlation between pCSF pulse and finger pulse (mean CorrCoeff = 0.66), supporting arterial pulsation as a major driver for pCSF dynamics. The time delay between pCSF and finger pulses (TimeDelay) was significantly lower (i.e., faster pulse propagation) with advanced age (Pearson's r = -0.44, p = 0.007). Shorter TimeDelay was further associated with worse cognitive function in the older participants. Overall, our study demonstrated pCSF as a viable pathway for measuring intracranial pulses and encouraged future studies to investigate its relevance with cerebrovascular functions.


Asunto(s)
Rigidez Vascular , Humanos , Hidrodinámica , Arterias/diagnóstico por imagen
2.
Alzheimers Dement ; 20(2): 1406-1420, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38015980

RESUMEN

INTRODUCTION: Social connectedness is associated with slower cognitive decline among older adults. Recent research suggests that distinct aspects of social networks may have differential effects on cognitive resilience, but few studies analyze brain structure. METHODS: This study includes 117 cognitively impaired and 59 unimpaired older adults. The effects of social network characteristics (bridging/bonding) on brain regions of interests were analyzed using linear regressions and voxel-wise multiple linear regressions of gray matter density. RESULTS: Increased social bridging was associated with greater bilateral amygdala volume and insular thickness, and left frontal lobe thickness, putamen, and thalamic volumes. Increased social bonding was associated with greater bilateral medial orbitofrontal and caudal anterior cingulate thickness, as well as right frontal lobe thickness, putamen, and amygdala volumes. DISCUSSION: The associations between social connectedness and brain structure vary depending on the types of social enrichment accessible through social networks, suggesting that psychosocial interventions could mitigate neurodegeneration. HIGHLIGHTS: Distinct forms of social capital are uniquely linked to gray matter density (GMD). Bridging is associated with preserved GMD in limbic system structures. Bonding is associated with preserved GMD in frontal lobe regions. Bridging is associated with increased brain reserve in sensory processing regions. Bonding is associated with increased brain reserve in regions of stress modulation.


Asunto(s)
Encéfalo , Imagen por Resonancia Magnética , Humanos , Anciano , Encéfalo/diagnóstico por imagen , Sustancia Gris/diagnóstico por imagen , Corteza Cerebral , Red Social
3.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33971669

RESUMEN

A large number of genetic variations have been identified to be associated with Alzheimer's disease (AD) and related quantitative traits. However, majority of existing studies focused on single types of omics data, lacking the power of generating a community including multi-omic markers and their functional connections. Because of this, the immense value of multi-omics data on AD has attracted much attention. Leveraging genomic, transcriptomic and proteomic data, and their backbone network through functional relations, we proposed a modularity-constrained logistic regression model to mine the association between disease status and a group of functionally connected multi-omic features, i.e. single-nucleotide polymorphisms (SNPs), genes and proteins. This new model was applied to the real data collected from the frontal cortex tissue in the Religious Orders Study and Memory and Aging Project cohort. Compared with other state-of-art methods, it provided overall the best prediction performance during cross-validation. This new method helped identify a group of densely connected SNPs, genes and proteins predictive of AD status. These SNPs are mostly expression quantitative trait loci in the frontal region. Brain-wide gene expression profile of these genes and proteins were highly correlated with the brain activation map of 'vision', a brain function partly controlled by frontal cortex. These genes and proteins were also found to be associated with the amyloid deposition, cortical volume and average thickness of frontal regions. Taken together, these results suggested a potential pathway underlying the development of AD from SNPs to gene expression, protein expression and ultimately brain functional and structural changes.


Asunto(s)
Enfermedad de Alzheimer/genética , Bases de Datos de Ácidos Nucleicos , Genómica , Polimorfismo de Nucleótido Simple , Transcriptoma , Enfermedad de Alzheimer/metabolismo , Estudio de Asociación del Genoma Completo , Humanos
4.
Acta Neuropathol ; 146(2): 211-226, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37351604

RESUMEN

Two siblings with deletion mutation ∆K281 in MAPT developed frontotemporal dementia. At autopsy, numerous inclusions of hyperphosphorylated 3R Tau were present in neurons and glial cells of neocortex and some subcortical regions, including hippocampus, caudate/putamen and globus pallidus. The inclusions were argyrophilic with Bodian silver, but not with Gallyas-Braak silver. They were not labelled by an antibody specific for tau phosphorylated at S262 and/or S356. The inclusions were stained by luminescent conjugated oligothiophene HS-84, but not by bTVBT4. Electron cryo-microscopy revealed that the core of tau filaments was made of residues K254-F378 of 3R Tau and was indistinguishable from that of Pick's disease. We conclude that MAPT mutation ∆K281 causes Pick's disease.


Asunto(s)
Demencia Frontotemporal , Enfermedad de Pick , Humanos , Enfermedad de Pick/genética , Plata , Proteínas tau/genética , Proteínas tau/química , Demencia Frontotemporal/genética , Neuronas , Mutación/genética
5.
Brain ; 145(6): 2149-2160, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35411392

RESUMEN

Hippocampal subfields exhibit differential vulnerabilities to Alzheimer's disease-associated pathology including abnormal accumulation of amyloid-ß deposition and neurofibrillary tangles. These pathological processes extensively impact on the structural and functional interconnectivities of the subfields and may explain the association between hippocampal dysfunction and cognitive deficits. In this study, we investigated the degree of alterations in the microstructure of hippocampal subfields across the clinical continuum of Alzheimer's disease. We applied a grey matter-specific multi-compartment diffusion model (Cortical-Neurite orientation dispersion and density imaging) to understand the differential effects of Alzheimer's disease pathology on the hippocampal subfield microstructure. A total of 119 participants were included in this cross-sectional study. Participants were stratified into three categories, cognitively normal (n = 47), mild cognitive impairment (n = 52), and Alzheimer's disease (n = 19). Diffusion MRI, plasma biomarkers and neuropsychological test scores were used to determine the association between the microstructural integrity and Alzheimer's disease-associated molecular indicators and cognition. For Alzheimer's disease-related plasma biomarkers, we studied amyloid-ß, total tau and neurofilament light; for Alzheimer's disease-related neuropsychological tests, we included the Trail Making Test, Rey Auditory Verbal Learning Test, Digit Span and Montreal Cognitive Assessment. Comparisons between cognitively normal subjects and those with mild cognitive impairment showed significant microstructural alterations in the hippocampal cornu ammonis (CA) 4 and dentate gyrus region, whereas CA 1-3 was the most sensitive region for the later stages in the Alzheimer's disease clinical continuum. Among imaging metrics for microstructures, the volume fraction of isotropic diffusion for interstitial free water demonstrated the largest effect size in between-group comparisons. Regarding the plasma biomarkers, neurofilament light appeared to be the most sensitive biomarker for associations with microstructural imaging findings in CA4-dentate gyrus. CA 1-3 was the subfield which had stronger correlations between cognitive performance and microstructural metrics. Particularly, poor performance on the Rey Auditory Verbal Learning Test and Montreal Cognitive Assessment was associated with decreased intracellular volume fraction. Overall, our findings support the value of tissue-specific microstructural imaging for providing pathologically relevant information manifesting in the plasma biomarkers and neuropsychological outcomes across various stages of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Biomarcadores , Disfunción Cognitiva/patología , Estudios Transversales , Hipocampo/diagnóstico por imagen , Hipocampo/patología , Humanos , Imagen por Resonancia Magnética
6.
Alzheimers Dement ; 19(6): 2655-2665, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37037592

RESUMEN

Individuals with more complex jobs experience better cognitive function in old age and a lower risk of dementia, yet complexity has multiple dimensions. Drawing on the Social Networks in Alzheimer Disease study, we examine the association between occupational complexity and cognition in a sample of older adults (N = 355). A standard deviation (SD) increase in complex work with people is associated with a 9% to 12% reduction in the probability of mild cognitive impairment or dementia, a 0.14-0.19 SD increase in episodic memory, and a 0.18-0.25 SD increase in brain reserve, defined as the gap (residual) between global cognitive function and magnetic resonance imaging (MRI) indicators of brain atrophy. In contrast, complexity with data or things is rarely associated with cognitive outcomes. We discuss the clinical and methodological implications of these findings, including the need to complement data-centered activities (e.g., Sudoku puzzles) with person-centered interventions that increase social complexity.


Asunto(s)
Envejecimiento Cognitivo , Reserva Cognitiva , Demencia , Memoria Episódica , Carga de Trabajo , Anciano , Femenino , Humanos , Masculino , Atrofia , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Disfunción Cognitiva/epidemiología , Disfunción Cognitiva/prevención & control , Demencia/epidemiología , Demencia/prevención & control , Satisfacción en el Trabajo , Factores Sociales , Interacción Social , Carga de Trabajo/psicología
7.
Alzheimers Dement ; 19(12): 5690-5699, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37409680

RESUMEN

BACKGROUND: Identifying genetic patterns that contribute to Alzheimer's disease (AD) is important not only for pre-symptomatic risk assessment but also for building personalized therapeutic strategies. METHODS: We implemented a novel simulative deep learning model to chromosome 19 genetic data from the Alzheimer's Disease Neuroimaging Initiative and the Imaging and Genetic Biomarkers of Alzheimer's Disease datasets. The model quantified the contribution of each single nucleotide polymorphism (SNP) and their epistatic impact on the likelihood of AD using the occlusion method. The top 35 AD-risk SNPs in chromosome 19 were identified, and their ability to predict the rate of AD progression was analyzed. RESULTS: Rs561311966 (APOC1) and rs2229918 (ERCC1/CD3EAP) were recognized as the most powerful factors influencing AD risk. The top 35 chromosome 19 AD-risk SNPs were significant predictors of AD progression. DISCUSSION: The model successfully estimated the contribution of AD-risk SNPs that account for AD progression at the individual level. This can help in building preventive precision medicine.


Asunto(s)
Enfermedad de Alzheimer , Aprendizaje Profundo , Humanos , Enfermedad de Alzheimer/genética , Polimorfismo de Nucleótido Simple/genética , Cromosomas Humanos Par 19 , Neuroimagen/métodos , Progresión de la Enfermedad , Imagen por Resonancia Magnética/métodos
8.
Mol Psychiatry ; 26(1): 309-321, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-30361487

RESUMEN

Deposition of amyloid plaques in the brain is one of the two main pathological hallmarks of Alzheimer's disease (AD). Amyloid positron emission tomography (PET) is a neuroimaging tool that selectively detects in vivo amyloid deposition in the brain and is a reliable endophenotype for AD that complements cerebrospinal fluid biomarkers with regional information. We measured in vivo amyloid deposition in the brains of ~1000 subjects from three collaborative AD centers and ADNI using 11C-labeled Pittsburgh Compound-B (PiB)-PET imaging followed by meta-analysis of genome-wide association studies, first to our knowledge for PiB-PET, to identify novel genetic loci for this endophenotype. The APOE region showed the most significant association where several SNPs surpassed the genome-wide significant threshold, with APOE*4 being most significant (P-meta = 9.09E-30; ß = 0.18). Interestingly, after conditioning on APOE*4, 14 SNPs remained significant at P < 0.05 in the APOE region that were not in linkage disequilibrium with APOE*4. Outside the APOE region, the meta-analysis revealed 15 non-APOE loci with P < 1E-05 on nine chromosomes, with two most significant SNPs on chromosomes 8 (P-meta = 4.87E-07) and 3 (P-meta = 9.69E-07). Functional analyses of these SNPs indicate their potential relevance with AD pathogenesis. Top 15 non-APOE SNPs along with APOE*4 explained 25-35% of the amyloid variance in different datasets, of which 14-17% was explained by APOE*4 alone. In conclusion, we have identified novel signals in APOE and non-APOE regions that affect amyloid deposition in the brain. Our data also highlights the presence of yet to be discovered variants that may be responsible for the unexplained genetic variance of amyloid deposition.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Compuestos de Anilina/análisis , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Estudio de Asociación del Genoma Completo , Tomografía de Emisión de Positrones , Tiazoles/análisis , Enfermedad de Alzheimer/genética , Apolipoproteína E4/genética , Endofenotipos , Femenino , Humanos , Masculino , Polimorfismo de Nucleótido Simple/genética
9.
Neuroimage ; 226: 117560, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33189932

RESUMEN

Progressive accumulation of tau neurofibrillary tangles in the brain is a defining pathologic feature of Alzheimer's disease (AD). Tau pathology exhibits a predictable spatiotemporal spreading pattern, but the underlying mechanisms of this spread are poorly understood. Although AD is conventionally considered a disease of the gray matter, it is also associated with pronounced and progressive deterioration of the white matter (WM). A link between abnormal tau and WM degeneration is suggested by findings from both animal and postmortem studies, but few studies demonstrated their interplay in vivo. Recent advances in diffusion magnetic resonance imaging and the availability of tau positron emission tomography (PET) have made it possible to evaluate the association of tau and WM degeneration (tau-WM) in vivo. In this study, we explored the spatial pattern of tau-WM associations across the whole brain to evaluate the hypothesis that tau deposition is associated with WM microstructural alterations not only in isolated tracts, but in continuous structural connections in a stereotypic pattern. Sixty-two participants, including 22 cognitively normal subjects, 22 individuals with subjective cognitive decline, and 18 with mild cognitive impairment were included in the study. WM characteristics were inferred by classic diffusion tensor imaging (DTI) and a complementary diffusion compartment model - neurite orientation dispersion and density imaging (NODDI) that provides a proxy for axonal density. A data-driven iterative searching (DDIS) approach, coupled with whole-brain graph theory analyses, was developed to continuously track tau-WM association patterns. Without applying prior knowledge of the tau spread, we observed a distinct spatial pattern that resembled the typical propagation of tau pathology in AD. Such association pattern was not observed between diffusion and amyloid-ß PET signal. Tau-related WM degeneration is characterized by an increase in the mean diffusivity (with a dominant change in the radial direction) and a decrease in the intra-axonal volume fraction. These findings suggest that cortical tau deposition (as measured in tau PET) is associated with a lower axonal packing density and greater diffusion freedom. In conclusion, our in vivo findings using a data-driven method on cross-sectional data underline the important role of WM alterations in the AD pathological cascade with an association pattern similar to the postmortem Braak staging of AD. Future studies will focus on longitudinal analyses to provide in vivo evidence of tau pathology spreads along neuroanatomically connected brain areas.


Asunto(s)
Encéfalo/diagnóstico por imagen , Disfunción Cognitiva/diagnóstico por imagen , Degeneración Nerviosa/diagnóstico por imagen , Sustancia Blanca/diagnóstico por imagen , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Encéfalo/metabolismo , Encéfalo/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Estudios Transversales , Imagen de Difusión Tensora , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Tomografía de Emisión de Positrones , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
10.
Hum Brain Mapp ; 42(11): 3500-3516, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33949732

RESUMEN

Functional connectivity, as estimated using resting state functional MRI, has shown potential in bridging the gap between pathophysiology and cognition. However, clinical use of functional connectivity biomarkers is impeded by unreliable estimates of individual functional connectomes and lack of generalizability of models predicting cognitive outcomes from connectivity. To address these issues, we combine the frameworks of connectome predictive modeling and differential identifiability. Using the combined framework, we show that enhancing the individual fingerprint of resting state functional connectomes leads to robust identification of functional networks associated to cognitive outcomes and also improves prediction of cognitive outcomes from functional connectomes. Using a comprehensive spectrum of cognitive outcomes associated to Alzheimer's disease (AD), we identify and characterize functional networks associated to specific cognitive deficits exhibited in AD. This combined framework is an important step in making individual level predictions of cognition from resting state functional connectomes and in understanding the relationship between cognition and connectivity.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Disfunción Cognitiva/diagnóstico por imagen , Conectoma/métodos , Red Nerviosa/diagnóstico por imagen , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/fisiopatología , Disfunción Cognitiva/fisiopatología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Red Nerviosa/fisiopatología
11.
Bioinformatics ; 36(Suppl_1): i371-i379, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32657360

RESUMEN

MOTIVATION: Brain imaging genetics studies the complex associations between genotypic data such as single nucleotide polymorphisms (SNPs) and imaging quantitative traits (QTs). The neurodegenerative disorders usually exhibit the diversity and heterogeneity, originating from which different diagnostic groups might carry distinct imaging QTs, SNPs and their interactions. Sparse canonical correlation analysis (SCCA) is widely used to identify bi-multivariate genotype-phenotype associations. However, most existing SCCA methods are unsupervised, leading to an inability to identify diagnosis-specific genotype-phenotype associations. RESULTS: In this article, we propose a new joint multitask learning method, named MT-SCCALR, which absorbs the merits of both SCCA and logistic regression. MT-SCCALR learns genotype-phenotype associations of multiple tasks jointly, with each task focusing on identifying one diagnosis-specific genotype-phenotype pattern. Meanwhile, MT-SCCALR cannot only select relevant SNPs and imaging QTs for each diagnostic group alone, but also allows the selection of those shared by multiple diagnostic groups. We derive an efficient optimization algorithm whose convergence to a local optimum is guaranteed. Compared with two state-of-the-art methods, MT-SCCALR yields better or similar canonical correlation coefficients and classification performances. In addition, it owns much better discriminative canonical weight patterns of great interest than competitors. This demonstrates the power and capability of MTSCCAR in identifying diagnostically heterogeneous genotype-phenotype patterns, which would be helpful to understand the pathophysiology of brain disorders. AVAILABILITY AND IMPLEMENTATION: The software is publicly available at https://github.com/dulei323/MTSCCALR. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Enfermedad de Alzheimer , Neuroimagen , Algoritmos , Estudios de Asociación Genética , Humanos , Análisis Multivariante , Fenotipo , Polimorfismo de Nucleótido Simple
12.
Bioinformatics ; 36(8): 2554-2560, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31860065

RESUMEN

MOTIVATION: Brain imaging genetics aims to reveal genetic effects on brain phenotypes, where most studies examine phenotypes defined on anatomical or functional regions of interest (ROIs) given their biologically meaningful interpretation and modest dimensionality compared with voxelwise approaches. Typical ROI-level measures used in these studies are summary statistics from voxelwise measures in the region, without making full use of individual voxel signals. RESULTS: In this article, we propose a flexible and powerful framework for mining regional imaging genetic associations via voxelwise enrichment analysis, which embraces the collective effect of weak voxel-level signals and integrates brain anatomical annotation information. Our proposed method achieves three goals at the same time: (i) increase the statistical power by substantially reducing the burden of multiple comparison correction; (ii) employ brain annotation information to enable biologically meaningful interpretation and (iii) make full use of fine-grained voxelwise signals. We demonstrate our method on an imaging genetic analysis using data from the Alzheimer's Disease Neuroimaging Initiative, where we assess the collective regional genetic effects of voxelwise FDG-positron emission tomography measures between 116 ROIs and 565 373 single-nucleotide polymorphisms. Compared with traditional ROI-wise and voxelwise approaches, our method identified 2946 novel imaging genetic associations in addition to 33 ones overlapping with the two benchmark methods. In particular, two newly reported variants were further supported by transcriptome evidences from region-specific expression analysis. This demonstrates the promise of the proposed method as a flexible and powerful framework for exploring imaging genetic effects on the brain. AVAILABILITY AND IMPLEMENTATION: The R code and sample data are freely available at https://github.com/lshen/RIGEA. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Enfermedad de Alzheimer , Neuroimagen , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Encéfalo/diagnóstico por imagen , Humanos , Polimorfismo de Nucleótido Simple , Tomografía de Emisión de Positrones
13.
Nature ; 520(7546): 224-9, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25607358

RESUMEN

The highly complex structure of the human brain is strongly shaped by genetic influences. Subcortical brain regions form circuits with cortical areas to coordinate movement, learning, memory and motivation, and altered circuits can lead to abnormal behaviour and disease. To investigate how common genetic variants affect the structure of these brain regions, here we conduct genome-wide association studies of the volumes of seven subcortical regions and the intracranial volume derived from magnetic resonance images of 30,717 individuals from 50 cohorts. We identify five novel genetic variants influencing the volumes of the putamen and caudate nucleus. We also find stronger evidence for three loci with previously established influences on hippocampal volume and intracranial volume. These variants show specific volumetric effects on brain structures rather than global effects across structures. The strongest effects were found for the putamen, where a novel intergenic locus with replicable influence on volume (rs945270; P = 1.08 × 10(-33); 0.52% variance explained) showed evidence of altering the expression of the KTN1 gene in both brain and blood tissue. Variants influencing putamen volume clustered near developmental genes that regulate apoptosis, axon guidance and vesicle transport. Identification of these genetic variants provides insight into the causes of variability in human brain development, and may help to determine mechanisms of neuropsychiatric dysfunction.


Asunto(s)
Encéfalo/anatomía & histología , Variación Genética/genética , Estudio de Asociación del Genoma Completo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Apoptosis/genética , Núcleo Caudado/anatomía & histología , Niño , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Sitios Genéticos/genética , Hipocampo/anatomía & histología , Humanos , Imagen por Resonancia Magnética , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Tamaño de los Órganos/genética , Putamen/anatomía & histología , Caracteres Sexuales , Cráneo/anatomía & histología , Adulto Joven
14.
BMC Bioinformatics ; 21(Suppl 21): 496, 2020 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-33371874

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most common type of dementia, typically characterized by memory loss followed by progressive cognitive decline and functional impairment. Many clinical trials of potential therapies for AD have failed, and there is currently no approved disease-modifying treatment. Biomarkers for early detection and mechanistic understanding of disease course are critical for drug development and clinical trials. Amyloid has been the focus of most biomarker research. Here, we developed a deep learning-based framework to identify informative features for AD classification using tau positron emission tomography (PET) scans. RESULTS: The 3D convolutional neural network (CNN)-based classification model of AD from cognitively normal (CN) yielded an average accuracy of 90.8% based on five-fold cross-validation. The LRP model identified the brain regions in tau PET images that contributed most to the AD classification from CN. The top identified regions included the hippocampus, parahippocampus, thalamus, and fusiform. The layer-wise relevance propagation (LRP) results were consistent with those from the voxel-wise analysis in SPM12, showing significant focal AD associated regional tau deposition in the bilateral temporal lobes including the entorhinal cortex. The AD probability scores calculated by the classifier were correlated with brain tau deposition in the medial temporal lobe in MCI participants (r = 0.43 for early MCI and r = 0.49 for late MCI). CONCLUSION: A deep learning framework combining 3D CNN and LRP algorithms can be used with tau PET images to identify informative features for AD classification and may have application for early detection during prodromal stages of AD.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Aprendizaje Profundo , Procesamiento de Imagen Asistido por Computador/métodos , Tomografía de Emisión de Positrones , Proteínas tau/metabolismo , Anciano , Enfermedad de Alzheimer/fisiopatología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/fisiopatología , Cognición , Progresión de la Enfermedad , Diagnóstico Precoz , Femenino , Humanos , Masculino
15.
BMC Genomics ; 21(Suppl 11): 896, 2020 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-33372590

RESUMEN

BACKGROUND: Genome-wide association studies (GWAS) have identified many individual genes associated with brain imaging quantitative traits (QTs) in Alzheimer's disease (AD). However single marker level association discovery may not be able to address the underlying biological interactions with disease mechanism. RESULTS: In this paper, we used the MGAS (Multivariate Gene-based Association test by extended Simes procedure) tool to perform multivariate GWAS on eight AD-relevant subcortical imaging measures. We conducted multiple iPINBPA (integrative Protein-Interaction-Network-Based Pathway Analysis) network analyses on MGAS findings using protein-protein interaction (PPI) data, and identified five Consensus Modules (CMs) from the PPI network. Functional annotation and network analysis were performed on the identified CMs. The MGAS yielded significant hits within APOE, TOMM40 and APOC1 genes, which were known AD risk factors, as well as a few new genes such as LAMA1, XYLB, HSD17B7P2, and NPEPL1. The identified five CMs were enriched by biological processes related to disorders such as Alzheimer's disease, Legionellosis, Pertussis, and Serotonergic synapse. CONCLUSIONS: The statistical power of coupling MGAS with iPINBPA was higher than traditional GWAS method, and yielded new findings that were missed by GWAS. This study provides novel insights into the molecular mechanism of Alzheimer's Disease and will be of value to novel gene discovery and functional genomic studies.


Asunto(s)
Enfermedad de Alzheimer , Estudio de Asociación del Genoma Completo , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Encéfalo/diagnóstico por imagen , Predisposición Genética a la Enfermedad , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple , Mapas de Interacción de Proteínas
16.
Brief Bioinform ; 19(6): 1370-1381, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-28679163

RESUMEN

In the past decade, significant progress has been made in complex disease research across multiple omics layers from genome, transcriptome and proteome to metabolome. There is an increasing awareness of the importance of biological interconnections, and much success has been achieved using systems biology approaches. However, because of the typical focus on one single omics layer at a time, existing systems biology findings explain only a modest portion of complex disease. Recent advances in multi-omics data collection and sharing present us new opportunities for studying complex diseases in a more comprehensive fashion, and yet simultaneously create new challenges considering the unprecedented data dimensionality and diversity. Here, our goal is to review extant and emerging network approaches that can be applied across multiple biological layers to facilitate a more comprehensive and integrative multilayered omics analysis of complex diseases.


Asunto(s)
Genómica , Proteómica , Biología de Sistemas , Transcriptoma , Minería de Datos , Epistasis Genética , Aprendizaje Automático
17.
Bioinformatics ; 35(14): i474-i483, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31510645

RESUMEN

MOTIVATION: Identifying the genetic basis of the brain structure, function and disorder by using the imaging quantitative traits (QTs) as endophenotypes is an important task in brain science. Brain QTs often change over time while the disorder progresses and thus understanding how the genetic factors play roles on the progressive brain QT changes is of great importance and meaning. Most existing imaging genetics methods only analyze the baseline neuroimaging data, and thus those longitudinal imaging data across multiple time points containing important disease progression information are omitted. RESULTS: We propose a novel temporal imaging genetic model which performs the multi-task sparse canonical correlation analysis (T-MTSCCA). Our model uses longitudinal neuroimaging data to uncover that how single nucleotide polymorphisms (SNPs) play roles on affecting brain QTs over the time. Incorporating the relationship of the longitudinal imaging data and that within SNPs, T-MTSCCA could identify a trajectory of progressive imaging genetic patterns over the time. We propose an efficient algorithm to solve the problem and show its convergence. We evaluate T-MTSCCA on 408 subjects from the Alzheimer's Disease Neuroimaging Initiative database with longitudinal magnetic resonance imaging data and genetic data available. The experimental results show that T-MTSCCA performs either better than or equally to the state-of-the-art methods. In particular, T-MTSCCA could identify higher canonical correlation coefficients and capture clearer canonical weight patterns. This suggests that T-MTSCCA identifies time-consistent and time-dependent SNPs and imaging QTs, which further help understand the genetic basis of the brain QT changes over the time during the disease progression. AVAILABILITY AND IMPLEMENTATION: The software and simulation data are publicly available at https://github.com/dulei323/TMTSCCA. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Neuroimagen , Algoritmos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Encéfalo/anatomía & histología , Humanos , Estudios Longitudinales , Imagen por Resonancia Magnética
18.
BMC Med Inform Decis Mak ; 20(1): 319, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33267852

RESUMEN

BACKGROUND: Cognitive assessments represent the most common clinical routine for the diagnosis of Alzheimer's Disease (AD). Given a large number of cognitive assessment tools and time-limited office visits, it is important to determine a proper set of cognitive tests for different subjects. Most current studies create guidelines of cognitive test selection for a targeted population, but they are not customized for each individual subject. In this manuscript, we develop a machine learning paradigm enabling personalized cognitive assessments prioritization. METHOD: We adapt a newly developed learning-to-rank approach [Formula: see text] to implement our paradigm. This method learns the latent scoring function that pushes the most effective cognitive assessments onto the top of the prioritization list. We also extend [Formula: see text] to better separate the most effective cognitive assessments and the less effective ones. RESULTS: Our empirical study on the ADNI data shows that the proposed paradigm outperforms the state-of-the-art baselines on identifying and prioritizing individual-specific cognitive biomarkers. We conduct experiments in cross validation and level-out validation settings. In the two settings, our paradigm significantly outperforms the best baselines with improvement as much as 22.1% and 19.7%, respectively, on prioritizing cognitive features. CONCLUSIONS: The proposed paradigm achieves superior performance on prioritizing cognitive biomarkers. The cognitive biomarkers prioritized on top have great potentials to facilitate personalized diagnosis, disease subtyping, and ultimately precision medicine in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Cognición/fisiología , Disfunción Cognitiva/patología , Interpretación de Imagen Asistida por Computador/métodos , Imagen por Resonancia Magnética/métodos , Enfermedad de Alzheimer/diagnóstico , Biomarcadores , Disfunción Cognitiva/diagnóstico , Biología Computacional , Humanos , Aprendizaje Automático
19.
Alzheimers Dement ; 16(9): 1213-1223, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32755048

RESUMEN

INTRODUCTION: Abnormal gene expression patterns may contribute to the onset and progression of late-onset Alzheimer's disease (LOAD). METHODS: We performed transcriptome-wide meta-analysis (N = 1440) of blood-based microarray gene expression profiles as well as neuroimaging and cerebrospinal fluid (CSF) endophenotype analysis. RESULTS: We identified and replicated five genes (CREB5, CD46, TMBIM6, IRAK3, and RPAIN) as significantly dysregulated in LOAD. The most significantly altered gene, CREB5, was also associated with brain atrophy and increased amyloid beta (Aß) accumulation, especially in the entorhinal cortex region. cis-expression quantitative trait loci mapping analysis of CREB5 detected five significant associations (P < 5 × 10-8 ), where rs56388170 (most significant) was also significantly associated with global cortical Aß deposition measured by [18 F]Florbetapir positron emission tomography and CSF Aß1-42 . DISCUSSION: RNA from peripheral blood indicated a differential gene expression pattern in LOAD. Genes identified have been implicated in biological processes relevant to Alzheimer's disease. CREB, in particular, plays a key role in nervous system development, cell survival, plasticity, and learning and memory.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/genética , Perfilación de la Expresión Génica , Anciano , Enfermedad de Alzheimer/sangre , Péptidos beta-Amiloides/líquido cefalorraquídeo , Péptidos beta-Amiloides/metabolismo , Compuestos de Anilina , Atrofia/patología , Encéfalo/patología , Corteza Entorrinal/patología , Glicoles de Etileno , Femenino , Técnicas de Genotipaje , Humanos , Masculino , Tomografía de Emisión de Positrones
20.
Bioinformatics ; 34(17): i866-i874, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30423101

RESUMEN

Motivation: The identification of quantitative trait loci (QTL) is critical to the study of causal relationships between genetic variations and disease abnormalities. We focus on identifying the QTLs associated to the brain endophenotypes in imaging genomics study for Alzheimer's Disease (AD). Existing research works mainly depict the association between single nucleotide polymorphisms (SNPs) and the brain endophenotypes via the linear methods, which may introduce high bias due to the simplicity of the models. Since the influence of QTLs on brain endophenotypes is quite complex, it is desired to design the appropriate non-linear models to investigate the associations of genotypes and endophenotypes. Results: In this paper, we propose a new additive model to learn the non-linear associations between SNPs and brain endophenotypes in Alzheimer's disease. Our model can be flexibly employed to explain the non-linear influence of QTLs, thus is more adaptive for the complex distribution of the high-throughput biological data. Meanwhile, as an important computational learning theory contribution, we provide the generalization error analysis for the proposed approach. Unlike most previous theoretical analysis under independent and identically distributed samples assumption, our error bound is based on m-dependent observations, which is more appropriate for the high-throughput and noisy biological data. Experiments on the data from Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort demonstrate the promising performance of our approach for identifying biological meaningful SNPs. Availability and implementation: An executable is available at https://github.com/littleq1991/additive_FNNRW.


Asunto(s)
Encéfalo , Sitios de Carácter Cuantitativo , Enfermedad de Alzheimer/genética , Encéfalo/metabolismo , Endofenotipos , Genotipo , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple , Programas Informáticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA