Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 437
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Neuroimage ; 295: 120635, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38729542

RESUMEN

In pursuit of cultivating automated models for magnetic resonance imaging (MRI) to aid in diagnostics, an escalating demand for extensive, multisite, and heterogeneous brain imaging datasets has emerged. This potentially introduces biased outcomes when directly applied for subsequent analysis. Researchers have endeavored to address this issue by pursuing the harmonization of MRIs. However, most existing image-based harmonization methods for MRI are tailored for 2D slices, which may introduce inter-slice variations when they are combined into a 3D volume. In this study, we aim to resolve inconsistencies between slices by introducing a pseudo-warping field. This field is created randomly and utilized to transform a slice into an artificially warped subsequent slice. The objective of this pseudo-warping field is to ensure that generators can consistently harmonize adjacent slices to another domain, without being affected by the varying content present in different slices. Furthermore, we construct unsupervised spatial and recycle loss to enhance the spatial accuracy and slice-wise consistency across the 3D images. The results demonstrate that our model effectively mitigates inter-slice variations and successfully preserves the anatomical details of the images during the harmonization process. Compared to generative harmonization models that employ 3D operators, our model exhibits greater computational efficiency and flexibility.


Asunto(s)
Encéfalo , Imagenología Tridimensional , Imagen por Resonancia Magnética , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/normas , Humanos , Imagenología Tridimensional/métodos , Encéfalo/diagnóstico por imagen , Algoritmos , Neuroimagen/métodos , Neuroimagen/normas
2.
Neuroimage ; 292: 120617, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38636639

RESUMEN

A primary challenge to the data-driven analysis is the balance between poor generalizability of population-based research and characterizing more subject-, study- and population-specific variability. We previously introduced a fully automated spatially constrained independent component analysis (ICA) framework called NeuroMark and its functional MRI (fMRI) template. NeuroMark has been successfully applied in numerous studies, identifying brain markers reproducible across datasets and disorders. The first NeuroMark template was constructed based on young adult cohorts. We recently expanded on this initiative by creating a standardized normative multi-spatial-scale functional template using over 100,000 subjects, aiming to improve generalizability and comparability across studies involving diverse cohorts. While a unified template across the lifespan is desirable, a comprehensive investigation of the similarities and differences between components from different age populations might help systematically transform our understanding of the human brain by revealing the most well-replicated and variable network features throughout the lifespan. In this work, we introduced two significant expansions of NeuroMark templates first by generating replicable fMRI templates for infants, adolescents, and aging cohorts, and second by incorporating structural MRI (sMRI) and diffusion MRI (dMRI) modalities. Specifically, we built spatiotemporal fMRI templates based on 6,000 resting-state scans from four datasets. This is the first attempt to create robust ICA templates covering dynamic brain development across the lifespan. For the sMRI and dMRI data, we used two large publicly available datasets including more than 30,000 scans to build reliable templates. We employed a spatial similarity analysis to identify replicable templates and investigate the degree to which unique and similar patterns are reflective in different age populations. Our results suggest remarkably high similarity of the resulting adapted components, even across extreme age differences. With the new templates, the NeuroMark framework allows us to perform age-specific adaptations and to capture features adaptable to each modality, therefore facilitating biomarker identification across brain disorders. In sum, the present work demonstrates the generalizability of NeuroMark templates and suggests the potential of new templates to boost accuracy in mental health research and advance our understanding of lifespan and cross-modal alterations.


Asunto(s)
Encéfalo , Imagen por Resonancia Magnética , Humanos , Adulto , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/normas , Encéfalo/diagnóstico por imagen , Adolescente , Adulto Joven , Masculino , Anciano , Femenino , Persona de Mediana Edad , Lactante , Niño , Envejecimiento/fisiología , Preescolar , Reproducibilidad de los Resultados , Procesamiento de Imagen Asistido por Computador/métodos , Procesamiento de Imagen Asistido por Computador/normas , Anciano de 80 o más Años , Neuroimagen/métodos , Neuroimagen/normas , Imagen de Difusión por Resonancia Magnética/métodos , Imagen de Difusión por Resonancia Magnética/normas
3.
Neuroimage ; 296: 120663, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38843963

RESUMEN

INTRODUCTION: Timely diagnosis and prognostication of Alzheimer's disease (AD) and mild cognitive impairment (MCI) are pivotal for effective intervention. Artificial intelligence (AI) in neuroradiology may aid in such appropriate diagnosis and prognostication. This study aimed to evaluate the potential of novel diffusion model-based AI for enhancing AD and MCI diagnosis through superresolution (SR) of brain magnetic resonance (MR) images. METHODS: 1.5T brain MR scans of patients with AD or MCI and healthy controls (NC) from Alzheimer's Disease Neuroimaging Initiative 1 (ADNI1) were superresolved to 3T using a novel diffusion model-based generative AI (d3T*) and a convolutional neural network-based model (c3T*). Comparisons of image quality to actual 1.5T and 3T MRI were conducted based on signal-to-noise ratio (SNR), naturalness image quality evaluator (NIQE), and Blind/Referenceless Image Spatial Quality Evaluator (BRISQUE). Voxel-based volumetric analysis was then conducted to study whether 3T* images offered more accurate volumetry than 1.5T images. Binary and multiclass classifications of AD, MCI, and NC were conducted to evaluate whether 3T* images offered superior AD classification performance compared to actual 1.5T MRI. Moreover, CNN-based classifiers were used to predict conversion of MCI to AD, to evaluate the prognostication performance of 3T* images. The classification performances were evaluated using accuracy, sensitivity, specificity, F1 score, Matthews correlation coefficient (MCC), and area under the receiver-operating curves (AUROC). RESULTS: Analysis of variance (ANOVA) detected significant differences in image quality among the 1.5T, c3T*, d3T*, and 3T groups across all metrics. Both c3T* and d3T* showed superior image quality compared to 1.5T MRI in NIQE and BRISQUE with statistical significance. While the hippocampal volumes measured in 3T* and 3T images were not significantly different, the hippocampal volume measured in 1.5T images showed significant difference. 3T*-based AD classifications showed superior performance across all performance metrics compared to 1.5T-based AD classification. Classification performance between d3T* and actual 3T was not significantly different. 3T* images offered superior accuracy in predicting the conversion of MCI to AD than 1.5T images did. CONCLUSIONS: The diffusion model-based MRI SR enhances the resolution of brain MR images, significantly improving diagnostic and prognostic accuracy for AD and MCI. Superresolved 3T* images closely matched actual 3T MRIs in quality and volumetric accuracy, and notably improved the prediction performance of conversion from MCI to AD.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/clasificación , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/clasificación , Anciano , Femenino , Masculino , Pronóstico , Anciano de 80 o más Años , Inteligencia Artificial , Imagen por Resonancia Magnética/métodos , Interpretación de Imagen Asistida por Computador/métodos , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Persona de Mediana Edad , Imagen de Difusión por Resonancia Magnética/métodos , Neuroimagen/métodos , Neuroimagen/normas
4.
Hum Brain Mapp ; 45(10): e26768, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38949537

RESUMEN

Structural neuroimaging data have been used to compute an estimate of the biological age of the brain (brain-age) which has been associated with other biologically and behaviorally meaningful measures of brain development and aging. The ongoing research interest in brain-age has highlighted the need for robust and publicly available brain-age models pre-trained on data from large samples of healthy individuals. To address this need we have previously released a developmental brain-age model. Here we expand this work to develop, empirically validate, and disseminate a pre-trained brain-age model to cover most of the human lifespan. To achieve this, we selected the best-performing model after systematically examining the impact of seven site harmonization strategies, age range, and sample size on brain-age prediction in a discovery sample of brain morphometric measures from 35,683 healthy individuals (age range: 5-90 years; 53.59% female). The pre-trained models were tested for cross-dataset generalizability in an independent sample comprising 2101 healthy individuals (age range: 8-80 years; 55.35% female) and for longitudinal consistency in a further sample comprising 377 healthy individuals (age range: 9-25 years; 49.87% female). This empirical examination yielded the following findings: (1) the accuracy of age prediction from morphometry data was higher when no site harmonization was applied; (2) dividing the discovery sample into two age-bins (5-40 and 40-90 years) provided a better balance between model accuracy and explained age variance than other alternatives; (3) model accuracy for brain-age prediction plateaued at a sample size exceeding 1600 participants. These findings have been incorporated into CentileBrain (https://centilebrain.org/#/brainAGE2), an open-science, web-based platform for individualized neuroimaging metrics.


Asunto(s)
Envejecimiento , Encéfalo , Imagen por Resonancia Magnética , Humanos , Adolescente , Femenino , Anciano , Adulto , Niño , Adulto Joven , Masculino , Encéfalo/diagnóstico por imagen , Encéfalo/anatomía & histología , Encéfalo/crecimiento & desarrollo , Anciano de 80 o más Años , Preescolar , Persona de Mediana Edad , Envejecimiento/fisiología , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos , Neuroimagen/normas , Tamaño de la Muestra
5.
Hum Brain Mapp ; 45(9): e26721, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38899549

RESUMEN

With the rise of open data, identifiability of individuals based on 3D renderings obtained from routine structural magnetic resonance imaging (MRI) scans of the head has become a growing privacy concern. To protect subject privacy, several algorithms have been developed to de-identify imaging data using blurring, defacing or refacing. Completely removing facial structures provides the best re-identification protection but can significantly impact post-processing steps, like brain morphometry. As an alternative, refacing methods that replace individual facial structures with generic templates have a lower effect on the geometry and intensity distribution of original scans, and are able to provide more consistent post-processing results by the price of higher re-identification risk and computational complexity. In the current study, we propose a novel method for anonymized face generation for defaced 3D T1-weighted scans based on a 3D conditional generative adversarial network. To evaluate the performance of the proposed de-identification tool, a comparative study was conducted between several existing defacing and refacing tools, with two different segmentation algorithms (FAST and Morphobox). The aim was to evaluate (i) impact on brain morphometry reproducibility, (ii) re-identification risk, (iii) balance between (i) and (ii), and (iv) the processing time. The proposed method takes 9 s for face generation and is suitable for recovering consistent post-processing results after defacing.


Asunto(s)
Imagen por Resonancia Magnética , Humanos , Imagen por Resonancia Magnética/métodos , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/anatomía & histología , Masculino , Femenino , Redes Neurales de la Computación , Imagenología Tridimensional/métodos , Neuroimagen/métodos , Neuroimagen/normas , Anonimización de la Información , Adulto Joven , Procesamiento de Imagen Asistido por Computador/métodos , Procesamiento de Imagen Asistido por Computador/normas , Algoritmos
6.
Hum Brain Mapp ; 45(7): e26692, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38712767

RESUMEN

In neuroimaging studies, combining data collected from multiple study sites or scanners is becoming common to increase the reproducibility of scientific discoveries. At the same time, unwanted variations arise by using different scanners (inter-scanner biases), which need to be corrected before downstream analyses to facilitate replicable research and prevent spurious findings. While statistical harmonization methods such as ComBat have become popular in mitigating inter-scanner biases in neuroimaging, recent methodological advances have shown that harmonizing heterogeneous covariances results in higher data quality. In vertex-level cortical thickness data, heterogeneity in spatial autocorrelation is a critical factor that affects covariance heterogeneity. Our work proposes a new statistical harmonization method called spatial autocorrelation normalization (SAN) that preserves homogeneous covariance vertex-level cortical thickness data across different scanners. We use an explicit Gaussian process to characterize scanner-invariant and scanner-specific variations to reconstruct spatially homogeneous data across scanners. SAN is computationally feasible, and it easily allows the integration of existing harmonization methods. We demonstrate the utility of the proposed method using cortical thickness data from the Social Processes Initiative in the Neurobiology of the Schizophrenia(s) (SPINS) study. SAN is publicly available as an R package.


Asunto(s)
Corteza Cerebral , Imagen por Resonancia Magnética , Esquizofrenia , Humanos , Imagen por Resonancia Magnética/normas , Imagen por Resonancia Magnética/métodos , Esquizofrenia/diagnóstico por imagen , Esquizofrenia/patología , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/anatomía & histología , Neuroimagen/métodos , Neuroimagen/normas , Procesamiento de Imagen Asistido por Computador/métodos , Procesamiento de Imagen Asistido por Computador/normas , Masculino , Femenino , Adulto , Distribución Normal , Grosor de la Corteza Cerebral
7.
Hum Brain Mapp ; 45(8): e26707, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38798082

RESUMEN

Development of deep learning models to evaluate structural brain changes caused by cognitive impairment in MRI scans holds significant translational value. The efficacy of these models often encounters challenges due to variabilities arising from different data generation protocols, imaging equipment, radiological artifacts, and shifts in demographic distributions. Domain generalization (DG) techniques show promise in addressing these challenges by enabling the model to learn from one or more source domains and apply this knowledge to new, unseen target domains. Here we present a framework that utilizes model interpretability to enhance the generalizability of classification models across various cohorts. We used MRI scans and clinical diagnoses from four independent cohorts: Alzheimer's Disease Neuroimaging Initiative (ADNI, n = 1821), the Framingham Heart Study (FHS, n = 304), the Australian Imaging Biomarkers & Lifestyle Study of Ageing (AIBL, n = 661), and the National Alzheimer's Coordinating Center (NACC, n = 4647). With this data, we trained a deep neural network to focus on areas of the brain identified as relevant to the disease for model training. Our approach involved training a classifier to differentiate between structural neurodegeneration in individuals with normal cognition (NC), mild cognitive impairment (MCI), and dementia due to Alzheimer's disease (AD). This was achieved by aligning class-wise attention with a unified visual saliency prior, which was computed offline for each class using all the training data. Our method not only competes with state-of-the-art approaches but also shows improved correlation with postmortem histology. This alignment with the gold standard evidence is a significant step towards validating the effectiveness of DG frameworks, paving the way for their broader application in the field.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Aprendizaje Profundo , Imagen por Resonancia Magnética , Neuroimagen , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/patología , Anciano , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/normas , Femenino , Masculino , Neuroimagen/métodos , Neuroimagen/normas , Anciano de 80 o más Años , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/patología , Estudios de Cohortes
8.
J Integr Neurosci ; 23(5): 100, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38812383

RESUMEN

BACKGROUND: Multiple radiomics models have been proposed for grading glioma using different algorithms, features, and sequences of magnetic resonance imaging. The research seeks to assess the present overall performance of radiomics for grading glioma. METHODS: A systematic literature review of the databases Ovid MEDLINE PubMed, and Ovid EMBASE for publications published on radiomics for glioma grading between 2012 and 2023 was performed. The systematic review was carried out following the criteria of Preferred Reporting Items for Systematic Reviews and Meta-Analysis. RESULTS: In the meta-analysis, a total of 7654 patients from 40 articles, were assessed. R-package mada was used for modeling the joint estimates of specificity (SPE) and sensitivity (SEN). Pooled event rates across studies were performed with a random-effects meta-analysis. The heterogeneity of SPE and SEN were based on the χ2 test. Overall values for SPE and SEN in the differentiation between high-grade gliomas (HGGs) and low-grade gliomas (LGGs) were 84% and 91%, respectively. With regards to the discrimination between World Health Organization (WHO) grade 4 and WHO grade 3, the overall SPE was 81% and the SEN was 89%. The modern non-linear classifiers showed a better trend, whereas textural features tend to be the best-performing (29%) and the most used. CONCLUSIONS: Our findings confirm that present radiomics' diagnostic performance for glioma grading is superior in terms of SEN and SPE for the HGGs vs. LGGs discrimination task when compared to the WHO grade 4 vs. 3 task.


Asunto(s)
Neoplasias Encefálicas , Glioma , Imagen por Resonancia Magnética , Clasificación del Tumor , Glioma/diagnóstico por imagen , Glioma/patología , Humanos , Imagen por Resonancia Magnética/normas , Imagen por Resonancia Magnética/métodos , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Neuroimagen/normas , Neuroimagen/métodos , Radiómica
9.
Neuroimage ; 247: 118786, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34906711

RESUMEN

Here we investigate the crucial role of trials in task-based neuroimaging from the perspectives of statistical efficiency and condition-level generalizability. Big data initiatives have gained popularity for leveraging a large sample of subjects to study a wide range of effect magnitudes in the brain. On the other hand, most task-based FMRI designs feature a relatively small number of subjects, so that resulting parameter estimates may be associated with compromised precision. Nevertheless, little attention has been given to another important dimension of experimental design, which can equally boost a study's statistical efficiency: the trial sample size. The common practice of condition-level modeling implicitly assumes no cross-trial variability. Here, we systematically explore the different factors that impact effect uncertainty, drawing on evidence from hierarchical modeling, simulations and an FMRI dataset of 42 subjects who completed a large number of trials of cognitive control task. We find that, due to an approximately symmetric hyperbola-relationship between trial and subject sample sizes in the presence of relatively large cross-trial variability, 1) trial sample size has nearly the same impact as subject sample size on statistical efficiency; 2) increasing both the number of trials and subjects improves statistical efficiency more effectively than focusing on subjects alone; 3) trial sample size can be leveraged alongside subject sample size to improve the cost-effectiveness of an experimental design; 4) for small trial sample sizes, trial-level modeling, rather than condition-level modeling through summary statistics, may be necessary to accurately assess the standard error of an effect estimate. We close by making practical suggestions for improving experimental designs across neuroimaging and behavioral studies.


Asunto(s)
Encéfalo/diagnóstico por imagen , Ensayos Clínicos como Asunto/normas , Neuroimagen/normas , Tamaño de la Muestra , Interpretación Estadística de Datos , Humanos , Proyectos de Investigación/normas
10.
Neuroimage ; 246: 118751, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34848299

RESUMEN

BACKGROUND: Large-scale longitudinal and multi-centre studies are used to explore neuroimaging markers of normal ageing, and neurodegenerative and mental health disorders. Longitudinal changes in brain structure are typically small, therefore the reliability of automated techniques is crucial. Determining the effects of different factors on reliability allows investigators to control those adversely affecting reliability, calculate statistical power, or even avoid particular brain measures with low reliability. This study examined the impact of several image acquisition and processing factors and documented the test-retest reliability of structural MRI measurements. METHODS: In Phase I, 20 healthy adults (11 females; aged 20-30 years) were scanned on two occasions three weeks apart on the same scanner using the ADNI-3 protocol. On each occasion, individuals were scanned twice (repetition), after re-entering the scanner (reposition) and after tilting their head forward. At one year follow-up, nine returning individuals and 11 new volunteers were recruited for Phase II (11 females; aged 22-31 years). Scans were acquired on two different scanners using the ADNI-2 and ADNI-3 protocols. Structural images were processed using FreeSurfer (v5.3.0, 6.0.0 and 7.1.0) to provide subcortical and cortical volume, cortical surface area and thickness measurements. Intra-class correlation coefficients (ICC) were calculated to estimate test-retest reliability. We examined the effect of repetition, reposition, head tilt, time between scans, MRI sequence and scanner on reliability of structural brain measurements. Mean percentage differences were also calculated in supplementary analyses. RESULTS: Using the FreeSurfer v7.1.0 longitudinal pipeline, we observed high reliability for subcortical and cortical volumes, and cortical surface areas at repetition, reposition, three weeks and one year (mean ICCs>0.97). Cortical thickness reliability was lower (mean ICCs>0.82). Head tilt had the greatest adverse impact on ICC estimates, for example reducing mean right cortical thickness to ICC=0.74. In contrast, changes in ADNI sequence or MRI scanner had a minimal effect. We observed an increase in reliability for updated FreeSurfer versions, with the longitudinal pipeline consistently having a higher reliability than the cross-sectional pipeline. DISCUSSION: Longitudinal studies should monitor or control head tilt to maximise reliability. We provided the ICC estimates and mean percentage differences for all FreeSurfer brain regions, which may inform power analyses for clinical studies and have implications for the design of future longitudinal studies.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética/normas , Neuroimagen/normas , Adulto , Femenino , Humanos , Estudios Longitudinales , Imagen por Resonancia Magnética/métodos , Masculino , Neuroimagen/métodos , Reproducibilidad de los Resultados , Adulto Joven
11.
Neuroimage ; 249: 118871, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34995797

RESUMEN

Convolutional neural networks (CNN) can accurately predict chronological age in healthy individuals from structural MRI brain scans. Potentially, these models could be applied during routine clinical examinations to detect deviations from healthy ageing, including early-stage neurodegeneration. This could have important implications for patient care, drug development, and optimising MRI data collection. However, existing brain-age models are typically optimised for scans which are not part of routine examinations (e.g., volumetric T1-weighted scans), generalise poorly (e.g., to data from different scanner vendors and hospitals etc.), or rely on computationally expensive pre-processing steps which limit real-time clinical utility. Here, we sought to develop a brain-age framework suitable for use during routine clinical head MRI examinations. Using a deep learning-based neuroradiology report classifier, we generated a dataset of 23,302 'radiologically normal for age' head MRI examinations from two large UK hospitals for model training and testing (age range = 18-95 years), and demonstrate fast (< 5 s), accurate (mean absolute error [MAE] < 4 years) age prediction from clinical-grade, minimally processed axial T2-weighted and axial diffusion-weighted scans, with generalisability between hospitals and scanner vendors (Δ MAE < 1 year). The clinical relevance of these brain-age predictions was tested using 228 patients whose MRIs were reported independently by neuroradiologists as showing atrophy 'excessive for age'. These patients had systematically higher brain-predicted age than chronological age (mean predicted age difference = +5.89 years, 'radiologically normal for age' mean predicted age difference = +0.05 years, p < 0.0001). Our brain-age framework demonstrates feasibility for use as a screening tool during routine hospital examinations to automatically detect older-appearing brains in real-time, with relevance for clinical decision-making and optimising patient pathways.


Asunto(s)
Envejecimiento , Encéfalo/diagnóstico por imagen , Desarrollo Humano , Imagen por Resonancia Magnética , Neuroimagen , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , Envejecimiento/fisiología , Aprendizaje Profundo , Desarrollo Humano/fisiología , Humanos , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/normas , Persona de Mediana Edad , Neuroimagen/métodos , Neuroimagen/normas , Adulto Joven
12.
Neuroimage ; 249: 118835, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34936923

RESUMEN

Quantitative susceptibility mapping (QSM) is an MRI-based, computational method for anatomically localizing and measuring concentrations of specific biomarkers in tissue such as iron. Growing research suggests QSM is a viable method for evaluating the impact of iron overload in neurological disorders and on cognitive performance in aging. Several software toolboxes are currently available to reconstruct QSM maps from 3D GRE MR Images. However, few if any software packages currently exist that offer fully automated pipelines for QSM-based data analyses: from DICOM images to region-of-interest (ROI) based QSM values. Even less QSM-based software exist that offer quality control measures for evaluating the QSM output. Here, we address these gaps in the field by introducing and demonstrating the reliability and external validity of Ironsmith; an open-source, fully automated pipeline for creating and processing QSM maps, extracting QSM values from subcortical and cortical brain regions (89 ROIs) and evaluating the quality of QSM data using SNR measures and assessment of outlier regions on phase images. Ironsmith also features automatic filtering of QSM outlier values and precise CSF-only QSM reference masks that minimize partial volume effects. Testing of Ironsmith revealed excellent intra- and inter-rater reliability. Finally, external validity of Ironsmith was demonstrated via an anatomically selective relationship between motor performance and Ironsmith-derived QSM values in motor cortex. In sum, Ironsmith provides a freely-available, reliable, turn-key pipeline for QSM-based data analyses to support research on the impact of brain iron in aging and neurodegenerative disease.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Procesamiento de Imagen Asistido por Computador/métodos , Hierro/metabolismo , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos , Programas Informáticos , Encéfalo/diagnóstico por imagen , Humanos , Procesamiento de Imagen Asistido por Computador/normas , Imagen por Resonancia Magnética/normas , Neuroimagen/normas
13.
Hum Brain Mapp ; 43(1): 255-277, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-32596977

RESUMEN

The ENIGMA group on Generalized Anxiety Disorder (ENIGMA-Anxiety/GAD) is part of a broader effort to investigate anxiety disorders using imaging and genetic data across multiple sites worldwide. The group is actively conducting a mega-analysis of a large number of brain structural scans. In this process, the group was confronted with many methodological challenges related to study planning and implementation, between-country transfer of subject-level data, quality control of a considerable amount of imaging data, and choices related to statistical methods and efficient use of resources. This report summarizes the background information and rationale for the various methodological decisions, as well as the approach taken to implement them. The goal is to document the approach and help guide other research groups working with large brain imaging data sets as they develop their own analytic pipelines for mega-analyses.


Asunto(s)
Trastornos de Ansiedad/diagnóstico por imagen , Corteza Cerebral/diagnóstico por imagen , Interpretación Estadística de Datos , Metaanálisis como Asunto , Estudios Multicéntricos como Asunto , Neuroimagen , Humanos , Estudios Multicéntricos como Asunto/métodos , Estudios Multicéntricos como Asunto/normas , Neuroimagen/métodos , Neuroimagen/normas
14.
Hum Brain Mapp ; 43(3): 929-939, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34704337

RESUMEN

White matter hyperintensities (WMHs) represent the most common neuroimaging marker of cerebral small vessel disease (CSVD). The volume and location of WMHs are important clinical measures. We present a pipeline using deep fully convolutional network and ensemble models, combining U-Net, SE-Net, and multi-scale features, to automatically segment WMHs and estimate their volumes and locations. We evaluated our method in two datasets: a clinical routine dataset comprising 60 patients (selected from Chinese National Stroke Registry, CNSR) and a research dataset composed of 60 patients (selected from MICCAI WMH Challenge, MWC). The performance of our pipeline was compared with four freely available methods: LGA, LPA, UBO detector, and U-Net, in terms of a variety of metrics. Additionally, to access the model generalization ability, another research dataset comprising 40 patients (from Older Australian Twins Study and Sydney Memory and Aging Study, OSM), was selected and tested. The pipeline achieved the best performance in both research dataset and the clinical routine dataset with DSC being significantly higher than other methods (p < .001), reaching .833 and .783, respectively. The results of model generalization ability showed that the model trained on the research dataset (DSC = 0.736) performed higher than that trained on the clinical dataset (DSC = 0.622). Our method outperformed widely used pipelines in WMHs segmentation. This system could generate both image and text outputs for whole brain, lobar and anatomical automatic labeling WMHs. Additionally, software and models of our method are made publicly available at https://www.nitrc.org/projects/what_v1.


Asunto(s)
Leucoaraiosis/diagnóstico por imagen , Leucoaraiosis/patología , Imagen por Resonancia Magnética/métodos , Redes Neurales de la Computación , Neuroimagen/métodos , Anciano , Conjuntos de Datos como Asunto , Humanos , Imagen por Resonancia Magnética/normas , Neuroimagen/normas
15.
Hum Brain Mapp ; 43(1): 244-254, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-32841457

RESUMEN

The problem of poor reproducibility of scientific findings has received much attention over recent years, in a variety of fields including psychology and neuroscience. The problem has been partly attributed to publication bias and unwanted practices such as p-hacking. Low statistical power in individual studies is also understood to be an important factor. In a recent multisite collaborative study, we mapped brain anatomical left-right asymmetries for regional measures of surface area and cortical thickness, in 99 MRI datasets from around the world, for a total of over 17,000 participants. In the present study, we revisited these hemispheric effects from the perspective of reproducibility. Within each dataset, we considered that an effect had been reproduced when it matched the meta-analytic effect from the 98 other datasets, in terms of effect direction and significance threshold. In this sense, the results within each dataset were viewed as coming from separate studies in an "ideal publishing environment," that is, free from selective reporting and p hacking. We found an average reproducibility rate of 63.2% (SD = 22.9%, min = 22.2%, max = 97.0%). As expected, reproducibility was higher for larger effects and in larger datasets. Reproducibility was not obviously related to the age of participants, scanner field strength, FreeSurfer software version, cortical regional measurement reliability, or regional size. These findings constitute an empirical illustration of reproducibility in the absence of publication bias or p hacking, when assessing realistic biological effects in heterogeneous neuroscience data, and given typically-used sample sizes.


Asunto(s)
Corteza Cerebral/anatomía & histología , Corteza Cerebral/diagnóstico por imagen , Imagen por Resonancia Magnética/normas , Neuroimagen/normas , Adolescente , Adulto , Anciano , Grosor de la Corteza Cerebral , Conjuntos de Datos como Asunto , Humanos , Persona de Mediana Edad , Estudios Multicéntricos como Asunto/normas , Sesgo de Publicación , Reproducibilidad de los Resultados , Adulto Joven
16.
Hum Brain Mapp ; 43(4): 1179-1195, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34904312

RESUMEN

To acquire larger samples for answering complex questions in neuroscience, researchers have increasingly turned to multi-site neuroimaging studies. However, these studies are hindered by differences in images acquired across multiple sites. These effects have been shown to bias comparison between sites, mask biologically meaningful associations, and even introduce spurious associations. To address this, the field has focused on harmonizing data by removing site-related effects in the mean and variance of measurements. Contemporaneously with the increase in popularity of multi-center imaging, the use of machine learning (ML) in neuroimaging has also become commonplace. These approaches have been shown to provide improved sensitivity, specificity, and power due to their modeling the joint relationship across measurements in the brain. In this work, we demonstrate that methods for removing site effects in mean and variance may not be sufficient for ML. This stems from the fact that such methods fail to address how correlations between measurements can vary across sites. Data from the Alzheimer's Disease Neuroimaging Initiative is used to show that considerable differences in covariance exist across sites and that popular harmonization techniques do not address this issue. We then propose a novel harmonization method called Correcting Covariance Batch Effects (CovBat) that removes site effects in mean, variance, and covariance. We apply CovBat and show that within-site correlation matrices are successfully harmonized. Furthermore, we find that ML methods are unable to distinguish scanner manufacturer after our proposed harmonization is applied, and that the CovBat-harmonized data retain accurate prediction of disease group.


Asunto(s)
Corteza Cerebral/anatomía & histología , Corteza Cerebral/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador , Estudios Multicéntricos como Asunto , Neuroimagen , Conjuntos de Datos como Asunto , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Procesamiento de Imagen Asistido por Computador/normas , Aprendizaje Automático , Modelos Teóricos , Estudios Multicéntricos como Asunto/métodos , Estudios Multicéntricos como Asunto/normas , Neuroimagen/métodos , Neuroimagen/normas
17.
Hum Brain Mapp ; 43(1): 234-243, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33067842

RESUMEN

As stroke mortality rates decrease, there has been a surge of effort to study poststroke dementia (PSD) to improve long-term quality of life for stroke survivors. Hippocampal volume may be an important neuroimaging biomarker in poststroke dementia, as it has been associated with many other forms of dementia. However, studying hippocampal volume using MRI requires hippocampal segmentation. Advances in automated segmentation methods have allowed for studying the hippocampus on a large scale, which is important for robust results in the heterogeneous stroke population. However, most of these automated methods use a single atlas-based approach and may fail in the presence of severe structural abnormalities common in stroke. Hippodeep, a new convolutional neural network-based hippocampal segmentation method, does not rely solely on a single atlas-based approach and thus may be better suited for stroke populations. Here, we compared quality control and the accuracy of segmentations generated by Hippodeep and two well-accepted hippocampal segmentation methods on stroke MRIs (FreeSurfer 6.0 whole hippocampus and FreeSurfer 6.0 sum of hippocampal subfields). Quality control was performed using a stringent protocol for visual inspection of the segmentations, and accuracy was measured as volumetric correlation with manual segmentations. Hippodeep performed significantly better than both FreeSurfer methods in terms of quality control. All three automated segmentation methods had good correlation with manual segmentations and no one method was significantly more correlated than the others. Overall, this study suggests that both Hippodeep and FreeSurfer may be useful for hippocampal segmentation in stroke rehabilitation research, but Hippodeep may be more robust to stroke lesion anatomy.


Asunto(s)
Hipocampo/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Redes Neurales de la Computación , Neuroimagen/métodos , Accidente Cerebrovascular/diagnóstico por imagen , Conjuntos de Datos como Asunto , Hipocampo/patología , Humanos , Procesamiento de Imagen Asistido por Computador/normas , Imagen por Resonancia Magnética/normas , Neuroimagen/normas , Control de Calidad , Accidente Cerebrovascular/patología
18.
Hum Brain Mapp ; 43(1): 207-233, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33368865

RESUMEN

Structural hippocampal abnormalities are common in many neurological and psychiatric disorders, and variation in hippocampal measures is related to cognitive performance and other complex phenotypes such as stress sensitivity. Hippocampal subregions are increasingly studied, as automated algorithms have become available for mapping and volume quantification. In the context of the Enhancing Neuro Imaging Genetics through Meta Analysis Consortium, several Disease Working Groups are using the FreeSurfer software to analyze hippocampal subregion (subfield) volumes in patients with neurological and psychiatric conditions along with data from matched controls. In this overview, we explain the algorithm's principles, summarize measurement reliability studies, and demonstrate two additional aspects (subfield autocorrelation and volume/reliability correlation) with illustrative data. We then explain the rationale for a standardized hippocampal subfield segmentation quality control (QC) procedure for improved pipeline harmonization. To guide researchers to make optimal use of the algorithm, we discuss how global size and age effects can be modeled, how QC steps can be incorporated and how subfields may be aggregated into composite volumes. This discussion is based on a synopsis of 162 published neuroimaging studies (01/2013-12/2019) that applied the FreeSurfer hippocampal subfield segmentation in a broad range of domains including cognition and healthy aging, brain development and neurodegeneration, affective disorders, psychosis, stress regulation, neurotoxicity, epilepsy, inflammatory disease, childhood adversity and posttraumatic stress disorder, and candidate and whole genome (epi-)genetics. Finally, we highlight points where FreeSurfer-based hippocampal subfield studies may be optimized.


Asunto(s)
Hipocampo/anatomía & histología , Hipocampo/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Neuroimagen , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Procesamiento de Imagen Asistido por Computador/normas , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/normas , Estudios Multicéntricos como Asunto , Neuroimagen/métodos , Neuroimagen/normas , Control de Calidad
19.
Hum Brain Mapp ; 43(2): 816-832, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34708477

RESUMEN

The UK Biobank (UKB) is a highly promising dataset for brain biomarker research into population mental health due to its unprecedented sample size and extensive phenotypic, imaging, and biological measurements. In this study, we aimed to provide a shared foundation for UKB neuroimaging research into mental health with a focus on anxiety and depression. We compared UKB self-report measures and revealed important timing effects between scan acquisition and separate online acquisition of some mental health measures. To overcome these timing effects, we introduced and validated the Recent Depressive Symptoms (RDS-4) score which we recommend for state-dependent and longitudinal research in the UKB. We furthermore tested univariate and multivariate associations between brain imaging-derived phenotypes (IDPs) and mental health. Our results showed a significant multivariate relationship between IDPs and mental health, which was replicable. Conversely, effect sizes for individual IDPs were small. Test-retest reliability of IDPs was stronger for measures of brain structure than for measures of brain function. Taken together, these results provide benchmarks and guidelines for future UKB research into brain biomarkers of mental health.


Asunto(s)
Bancos de Muestras Biológicas , Encéfalo/diagnóstico por imagen , Bases de Datos Factuales , Depresión/diagnóstico , Trastornos Mentales/diagnóstico , Neuroimagen/normas , Autoinforme , Anciano , Bancos de Muestras Biológicas/normas , Bases de Datos Factuales/normas , Depresión/diagnóstico por imagen , Femenino , Humanos , Masculino , Trastornos Mentales/diagnóstico por imagen , Persona de Mediana Edad , Neuroimagen/métodos , Reproducibilidad de los Resultados , Autoinforme/normas , Reino Unido
20.
Hum Brain Mapp ; 43(1): 555-565, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33064342

RESUMEN

To identify neuroimaging biomarkers of alcohol dependence (AD) from structural magnetic resonance imaging, it may be useful to develop classification models that are explicitly generalizable to unseen sites and populations. This problem was explored in a mega-analysis of previously published datasets from 2,034 AD and comparison participants spanning 27 sites curated by the ENIGMA Addiction Working Group. Data were grouped into a training set used for internal validation including 1,652 participants (692 AD, 24 sites), and a test set used for external validation with 382 participants (146 AD, 3 sites). An exploratory data analysis was first conducted, followed by an evolutionary search based feature selection to site generalizable and high performing subsets of brain measurements. Exploratory data analysis revealed that inclusion of case- and control-only sites led to the inadvertent learning of site-effects. Cross validation methods that do not properly account for site can drastically overestimate results. Evolutionary-based feature selection leveraging leave-one-site-out cross-validation, to combat unintentional learning, identified cortical thickness in the left superior frontal gyrus and right lateral orbitofrontal cortex, cortical surface area in the right transverse temporal gyrus, and left putamen volume as final features. Ridge regression restricted to these features yielded a test-set area under the receiver operating characteristic curve of 0.768. These findings evaluate strategies for handling multi-site data with varied underlying class distributions and identify potential biomarkers for individuals with current AD.


Asunto(s)
Alcoholismo/diagnóstico por imagen , Corteza Cerebral/diagnóstico por imagen , Aprendizaje Automático , Imagen por Resonancia Magnética , Estudios Multicéntricos como Asunto , Neuroimagen , Putamen/diagnóstico por imagen , Corteza Cerebral/patología , Humanos , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/normas , Estudios Multicéntricos como Asunto/métodos , Estudios Multicéntricos como Asunto/normas , Neuroimagen/métodos , Neuroimagen/normas , Putamen/patología , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA