Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
JAMA Psychiatry ; 81(5): 456-467, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38353984

ABSTRACT

Importance: Brain aging elicits complex neuroanatomical changes influenced by multiple age-related pathologies. Understanding the heterogeneity of structural brain changes in aging may provide insights into preclinical stages of neurodegenerative diseases. Objective: To derive subgroups with common patterns of variation in participants without diagnosed cognitive impairment (WODCI) in a data-driven manner and relate them to genetics, biomedical measures, and cognitive decline trajectories. Design, Setting, and Participants: Data acquisition for this cohort study was performed from 1999 to 2020. Data consolidation and harmonization were conducted from July 2017 to July 2021. Age-specific subgroups of structural brain measures were modeled in 4 decade-long intervals spanning ages 45 to 85 years using a deep learning, semisupervised clustering method leveraging generative adversarial networks. Data were analyzed from July 2021 to February 2023 and were drawn from the Imaging-Based Coordinate System for Aging and Neurodegenerative Diseases (iSTAGING) international consortium. Individuals WODCI at baseline spanning ages 45 to 85 years were included, with greater than 50 000 data time points. Exposures: Individuals WODCI at baseline scan. Main Outcomes and Measures: Three subgroups, consistent across decades, were identified within the WODCI population. Associations with genetics, cardiovascular risk factors (CVRFs), amyloid ß (Aß), and future cognitive decline were assessed. Results: In a sample of 27 402 individuals (mean [SD] age, 63.0 [8.3] years; 15 146 female [55%]) WODCI, 3 subgroups were identified in contrast with the reference group: a typical aging subgroup, A1, with a specific pattern of modest atrophy and white matter hyperintensity (WMH) load, and 2 accelerated aging subgroups, A2 and A3, with characteristics that were more distinct at age 65 years and older. A2 was associated with hypertension, WMH, and vascular disease-related genetic variants and was enriched for Aß positivity (ages ≥65 years) and apolipoprotein E (APOE) ε4 carriers. A3 showed severe, widespread atrophy, moderate presence of CVRFs, and greater cognitive decline. Genetic variants associated with A1 were protective for WMH (rs7209235: mean [SD] B = -0.07 [0.01]; P value = 2.31 × 10-9) and Alzheimer disease (rs72932727: mean [SD] B = 0.1 [0.02]; P value = 6.49 × 10-9), whereas the converse was observed for A2 (rs7209235: mean [SD] B = 0.1 [0.01]; P value = 1.73 × 10-15 and rs72932727: mean [SD] B = -0.09 [0.02]; P value = 4.05 × 10-7, respectively); variants in A3 were associated with regional atrophy (rs167684: mean [SD] B = 0.08 [0.01]; P value = 7.22 × 10-12) and white matter integrity measures (rs1636250: mean [SD] B = 0.06 [0.01]; P value = 4.90 × 10-7). Conclusions and Relevance: The 3 subgroups showed distinct associations with CVRFs, genetics, and subsequent cognitive decline. These subgroups likely reflect multiple underlying neuropathologic processes and affect susceptibility to Alzheimer disease, paving pathways toward patient stratification at early asymptomatic stages and promoting precision medicine in clinical trials and health care.


Subject(s)
Aging , Brain , Humans , Aged , Female , Male , Middle Aged , Aged, 80 and over , Brain/diagnostic imaging , Brain/pathology , Aging/genetics , Aging/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/diagnostic imaging , Magnetic Resonance Imaging , Cohort Studies , Deep Learning
2.
Nat Commun ; 15(1): 354, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191573

ABSTRACT

Disease heterogeneity has been a critical challenge for precision diagnosis and treatment, especially in neurologic and neuropsychiatric diseases. Many diseases can display multiple distinct brain phenotypes across individuals, potentially reflecting disease subtypes that can be captured using MRI and machine learning methods. However, biological interpretability and treatment relevance are limited if the derived subtypes are not associated with genetic drivers or susceptibility factors. Herein, we describe Gene-SGAN - a multi-view, weakly-supervised deep clustering method - which dissects disease heterogeneity by jointly considering phenotypic and genetic data, thereby conferring genetic correlations to the disease subtypes and associated endophenotypic signatures. We first validate the generalizability, interpretability, and robustness of Gene-SGAN in semi-synthetic experiments. We then demonstrate its application to real multi-site datasets from 28,858 individuals, deriving subtypes of Alzheimer's disease and brain endophenotypes associated with hypertension, from MRI and single nucleotide polymorphism data. Derived brain phenotypes displayed significant differences in neuroanatomical patterns, genetic determinants, biological and clinical biomarkers, indicating potentially distinct underlying neuropathologic processes, genetic drivers, and susceptibility factors. Overall, Gene-SGAN is broadly applicable to disease subtyping and endophenotype discovery, and is herein tested on disease-related, genetically-associated neuroimaging phenotypes.


Subject(s)
Alzheimer Disease , Neuroimaging , Humans , Endophenotypes , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Brain/diagnostic imaging , Cluster Analysis
3.
Proc Natl Acad Sci U S A ; 120(52): e2300842120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38127979

ABSTRACT

Normal and pathologic neurobiological processes influence brain morphology in coordinated ways that give rise to patterns of structural covariance (PSC) across brain regions and individuals during brain aging and diseases. The genetic underpinnings of these patterns remain largely unknown. We apply a stochastic multivariate factorization method to a diverse population of 50,699 individuals (12 studies and 130 sites) and derive data-driven, multi-scale PSCs of regional brain size. PSCs were significantly correlated with 915 genomic loci in the discovery set, 617 of which are newly identified, and 72% were independently replicated. Key pathways influencing PSCs involve reelin signaling, apoptosis, neurogenesis, and appendage development, while pathways of breast cancer indicate potential interplays between brain metastasis and PSCs associated with neurodegeneration and dementia. Using support vector machines, multi-scale PSCs effectively derive imaging signatures of several brain diseases. Our results elucidate genetic and biological underpinnings that influence structural covariance patterns in the human brain.


Subject(s)
Brain Neoplasms , Magnetic Resonance Imaging , Humans , Magnetic Resonance Imaging/methods , Brain/pathology , Brain Mapping/methods , Genomics , Brain Neoplasms/pathology
4.
Comput Methods Programs Biomed ; 242: 107812, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37757566

ABSTRACT

BACKGROUND: Magnetic resonance imaging (MRI), digital pathology imaging (PATH), demographics, and IDH mutation status predict overall survival (OS) in glioma. Identifying and characterizing predictive features in the different modalities may improve OS prediction accuracy. PURPOSE: To evaluate the OS prediction accuracy of combinations of prognostic markers in glioma patients. MATERIALS AND METHODS: Multi-contrast MRI, comprising T1-weighted, T1-weighted post-contrast, T2-weighted, T2 fluid-attenuated-inversion-recovery, and pathology images from glioma patients (n = 160) were retrospectively collected (1983-2008) from TCGA alongside age and sex. Phenotypic profiling of tumors was performed by quantifying the radiographic and histopathologic descriptors extracted from the delineated region-of-interest in MRI and PATH images. A Cox proportional hazard model was trained with the MRI and PATH features, IDH mutation status, and basic demographic variables (age and sex) to predict OS. The performance was evaluated in a split-train-test configuration using the concordance-index, computed between the predicted risk score and observed OS. RESULTS: The average age of patients was 51.2years (women: n = 77, age-range=18-84years; men: n = 83, age-range=21-80years). The median OS of the participants was 494.5 (range,3-4752), 481 (range,7-4752), and 524.5 days (range,3-2869), respectively, in complete dataset, training, and test datasets. The addition of MRI or PATH features improved prediction of OS when compared to models based on age, sex, and mutation status alone or their combination (p < 0.001). The full multi-omics model integrated MRI, PATH, clinical, and genetic profiles and predicted the OS best (c-index= 0.87). CONCLUSION: The combination of imaging, genetic, and clinical profiles leads to a more accurate prognosis than the clinical and/or mutation status.


Subject(s)
Brain Neoplasms , Glioma , Male , Humans , Female , Adolescent , Young Adult , Adult , Middle Aged , Aged , Aged, 80 and over , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Retrospective Studies , Isocitrate Dehydrogenase/genetics , Glioma/diagnostic imaging , Glioma/genetics , Magnetic Resonance Imaging/methods , Phenotype , Mutation , Demography
5.
Sci Rep ; 13(1): 6406, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37076487

ABSTRACT

Effective clinical decision procedures must balance multiple competing objectives such as time-to-decision, acquisition costs, and accuracy. We describe and evaluate POSEIDON, a data-driven method for PrOspective SEquentIal DiagnOsis with Neutral zones to individualize clinical classifications. We evaluated the framework with an application in which the algorithm sequentially proposes to include cognitive, imaging, or molecular markers if a sufficiently more accurate prognosis of clinical decline to manifest Alzheimer's disease is expected. Over a wide range of cost parameter data-driven tuning lead to quantitatively lower total cost compared to ad hoc fixed sets of measurements. The classification accuracy based on all longitudinal data from participants that was acquired over 4.8 years on average was 0.89. The sequential algorithm selected 14 percent of available measurements and concluded after an average follow-up time of 0.74 years at the expense of 0.05 lower accuracy. Sequential classifiers were competitive from a multi-objective perspective since they could dominate fixed sets of measurements by making fewer errors using less resources. Nevertheless, the trade-off of competing objectives depends on inherently subjective prescribed cost parameters. Thus, despite the effectiveness of the method, the implementation into consequential clinical applications will remain controversial and evolve around the choice of cost parameters.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Cognitive Dysfunction/diagnosis , Prospective Studies , Alzheimer Disease/diagnosis , Alzheimer Disease/psychology , Prognosis , Cognition
6.
Neuroimage ; 269: 119911, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36731813

ABSTRACT

To learn multiscale functional connectivity patterns of the aging brain, we built a brain age prediction model of functional connectivity measures at seven scales on a large fMRI dataset, consisting of resting-state fMRI scans of 4186 individuals with a wide age range (22 to 97 years, with an average of 63) from five cohorts. We computed multiscale functional connectivity measures of individual subjects using a personalized functional network computational method, harmonized the functional connectivity measures of subjects from multiple datasets in order to build a functional brain age model, and finally evaluated how functional brain age gap correlated with cognitive measures of individual subjects. Our study has revealed that functional connectivity measures at multiple scales were more informative than those at any single scale for the brain age prediction, the data harmonization significantly improved the brain age prediction performance, and the data harmonization in the functional connectivity measures' tangent space worked better than in their original space. Moreover, brain age gap scores of individual subjects derived from the brain age prediction model were significantly correlated with clinical and cognitive measures. Overall, these results demonstrated that multiscale functional connectivity patterns learned from a large-scale multi-site rsfMRI dataset were informative for characterizing the aging brain and the derived brain age gap was associated with cognitive and clinical measures.


Subject(s)
Aging , Brain , Humans , Young Adult , Adult , Middle Aged , Aged , Aged, 80 and over , Brain Mapping/methods , Learning , Cohort Studies , Magnetic Resonance Imaging/methods
7.
ArXiv ; 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36748000

ABSTRACT

Disease heterogeneity has been a critical challenge for precision diagnosis and treatment, especially in neurologic and neuropsychiatric diseases. Many diseases can display multiple distinct brain phenotypes across individuals, potentially reflecting disease subtypes that can be captured using MRI and machine learning methods. However, biological interpretability and treatment relevance are limited if the derived subtypes are not associated with genetic drivers or susceptibility factors. Herein, we describe Gene-SGAN - a multi-view, weakly-supervised deep clustering method - which dissects disease heterogeneity by jointly considering phenotypic and genetic data, thereby conferring genetic correlations to the disease subtypes and associated endophenotypic signatures. We first validate the generalizability, interpretability, and robustness of Gene-SGAN in semi-synthetic experiments. We then demonstrate its application to real multi-site datasets from 28,858 individuals, deriving subtypes of Alzheimer's disease and brain endophenotypes associated with hypertension, from MRI and SNP data. Derived brain phenotypes displayed significant differences in neuroanatomical patterns, genetic determinants, biological and clinical biomarkers, indicating potentially distinct underlying neuropathologic processes, genetic drivers, and susceptibility factors. Overall, Gene-SGAN is broadly applicable to disease subtyping and endophenotype discovery, and is herein tested on disease-related, genetically-driven neuroimaging phenotypes.

8.
Neuroimage ; 269: 119898, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36702211

ABSTRACT

Generative adversarial networks (GANs) are one powerful type of deep learning models that have been successfully utilized in numerous fields. They belong to the broader family of generative methods, which learn to generate realistic data with a probabilistic model by learning distributions from real samples. In the clinical context, GANs have shown enhanced capabilities in capturing spatially complex, nonlinear, and potentially subtle disease effects compared to traditional generative methods. This review critically appraises the existing literature on the applications of GANs in imaging studies of various neurological conditions, including Alzheimer's disease, brain tumors, brain aging, and multiple sclerosis. We provide an intuitive explanation of various GAN methods for each application and further discuss the main challenges, open questions, and promising future directions of leveraging GANs in neuroimaging. We aim to bridge the gap between advanced deep learning methods and neurology research by highlighting how GANs can be leveraged to support clinical decision making and contribute to a better understanding of the structural and functional patterns of brain diseases.


Subject(s)
Alzheimer Disease , Neurosciences , Humans , Neuroimaging , Aging , Brain
9.
medRxiv ; 2023 Dec 30.
Article in English | MEDLINE | ID: mdl-38234857

ABSTRACT

Brain aging is a complex process influenced by various lifestyle, environmental, and genetic factors, as well as by age-related and often co-existing pathologies. MRI and, more recently, AI methods have been instrumental in understanding the neuroanatomical changes that occur during aging in large and diverse populations. However, the multiplicity and mutual overlap of both pathologic processes and affected brain regions make it difficult to precisely characterize the underlying neurodegenerative profile of an individual from an MRI scan. Herein, we leverage a state-of-the art deep representation learning method, Surreal-GAN, and present both methodological advances and extensive experimental results that allow us to elucidate the heterogeneity of brain aging in a large and diverse cohort of 49,482 individuals from 11 studies. Five dominant patterns of neurodegeneration were identified and quantified for each individual by their respective (herein referred to as) R-indices. Significant associations between R-indices and distinct biomedical, lifestyle, and genetic factors provide insights into the etiology of observed variances. Furthermore, baseline R-indices showed predictive value for disease progression and mortality. These five R-indices contribute to MRI-based precision diagnostics, prognostication, and may inform stratification into clinical trials.

10.
Brain Commun ; 4(3): fcac117, 2022.
Article in English | MEDLINE | ID: mdl-35611306

ABSTRACT

Neuroimaging biomarkers that distinguish between changes due to typical brain ageing and Alzheimer's disease are valuable for determining how much each contributes to cognitive decline. Supervised machine learning models can derive multivariate patterns of brain change related to the two processes, including the Spatial Patterns of Atrophy for Recognition of Alzheimer's Disease (SPARE-AD) and of Brain Aging (SPARE-BA) scores investigated herein. However, the substantial overlap between brain regions affected in the two processes confounds measuring them independently. We present a methodology, and associated results, towards disentangling the two. T1-weighted MRI scans of 4054 participants (48-95 years) with Alzheimer's disease, mild cognitive impairment (MCI), or cognitively normal (CN) diagnoses from the Imaging-based coordinate SysTem for AGIng and NeurodeGenerative diseases (iSTAGING) consortium were analysed. Multiple sets of SPARE scores were investigated, in order to probe imaging signatures of certain clinically or molecularly defined sub-cohorts. First, a subset of clinical Alzheimer's disease patients (n = 718) and age- and sex-matched CN adults (n = 718) were selected based purely on clinical diagnoses to train SPARE-BA1 (regression of age using CN individuals) and SPARE-AD1 (classification of CN versus Alzheimer's disease) models. Second, analogous groups were selected based on clinical and molecular markers to train SPARE-BA2 and SPARE-AD2 models: amyloid-positive Alzheimer's disease continuum group (n = 718; consisting of amyloid-positive Alzheimer's disease, amyloid-positive MCI, amyloid- and tau-positive CN individuals) and amyloid-negative CN group (n = 718). Finally, the combined group of the Alzheimer's disease continuum and amyloid-negative CN individuals was used to train SPARE-BA3 model, with the intention to estimate brain age regardless of Alzheimer's disease-related brain changes. The disentangled SPARE models, SPARE-AD2 and SPARE-BA3, derived brain patterns that were more specific to the two types of brain changes. The correlation between the SPARE-BA Gap (SPARE-BA minus chronological age) and SPARE-AD was significantly reduced after the decoupling (r = 0.56-0.06). The correlation of disentangled SPARE-AD was non-inferior to amyloid- and tau-related measurements and to the number of APOE ε4 alleles but was lower to Alzheimer's disease-related psychometric test scores, suggesting the contribution of advanced brain ageing to the latter. The disentangled SPARE-BA was consistently less correlated with Alzheimer's disease-related clinical, molecular and genetic variables. By employing conservative molecular diagnoses and introducing Alzheimer's disease continuum cases to the SPARE-BA model training, we achieved more dissociable neuroanatomical biomarkers of typical brain ageing and Alzheimer's disease.

11.
JAMA Psychiatry ; 79(5): 464-474, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35262657

ABSTRACT

Importance: Late-life depression (LLD) is characterized by considerable heterogeneity in clinical manifestation. Unraveling such heterogeneity might aid in elucidating etiological mechanisms and support precision and individualized medicine. Objective: To cross-sectionally and longitudinally delineate disease-related heterogeneity in LLD associated with neuroanatomy, cognitive functioning, clinical symptoms, and genetic profiles. Design, Setting, and Participants: The Imaging-Based Coordinate System for Aging and Neurodegenerative Diseases (iSTAGING) study is an international multicenter consortium investigating brain aging in pooled and harmonized data from 13 studies with more than 35 000 participants, including a subset of individuals with major depressive disorder. Multimodal data from a multicenter sample (N = 996), including neuroimaging, neurocognitive assessments, and genetics, were analyzed in this study. A semisupervised clustering method (heterogeneity through discriminative analysis) was applied to regional gray matter (GM) brain volumes to derive dimensional representations. Data were collected from July 2017 to July 2020 and analyzed from July 2020 to December 2021. Main Outcomes and Measures: Two dimensions were identified to delineate LLD-associated heterogeneity in voxelwise GM maps, white matter (WM) fractional anisotropy, neurocognitive functioning, clinical phenotype, and genetics. Results: A total of 501 participants with LLD (mean [SD] age, 67.39 [5.56] years; 332 women) and 495 healthy control individuals (mean [SD] age, 66.53 [5.16] years; 333 women) were included. Patients in dimension 1 demonstrated relatively preserved brain anatomy without WM disruptions relative to healthy control individuals. In contrast, patients in dimension 2 showed widespread brain atrophy and WM integrity disruptions, along with cognitive impairment and higher depression severity. Moreover, 1 de novo independent genetic variant (rs13120336; chromosome: 4, 186387714; minor allele, G) was significantly associated with dimension 1 (odds ratio, 2.35; SE, 0.15; P = 3.14 ×108) but not with dimension 2. The 2 dimensions demonstrated significant single-nucleotide variant-based heritability of 18% to 27% within the general population (N = 12 518 in UK Biobank). In a subset of individuals having longitudinal measurements, those in dimension 2 experienced a more rapid longitudinal change in GM and brain age (Cohen f2 = 0.03; P = .02) and were more likely to progress to Alzheimer disease (Cohen f2 = 0.03; P = .03) compared with those in dimension 1 (N = 1431 participants and 7224 scans from the Alzheimer's Disease Neuroimaging Initiative [ADNI], Baltimore Longitudinal Study of Aging [BLSA], and Biomarkers for Older Controls at Risk for Dementia [BIOCARD] data sets). Conclusions and Relevance: This study characterized heterogeneity in LLD into 2 dimensions with distinct neuroanatomical, cognitive, clinical, and genetic profiles. This dimensional approach provides a potential mechanism for investigating the heterogeneity of LLD and the relevance of the latent dimensions to possible disease mechanisms, clinical outcomes, and responses to interventions.


Subject(s)
Alzheimer Disease , Depressive Disorder, Major , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Brain/diagnostic imaging , Cognition , Depression , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/genetics , Female , Humans , Longitudinal Studies , Magnetic Resonance Imaging/methods , Male , Neuroimaging
12.
J Magn Reson Imaging ; 55(3): 908-916, 2022 03.
Article in English | MEDLINE | ID: mdl-34564904

ABSTRACT

BACKGROUND: In the medical imaging domain, deep learning-based methods have yet to see widespread clinical adoption, in part due to limited generalization performance across different imaging devices and acquisition protocols. The deviation between estimated brain age and biological age is an established biomarker of brain health and such models may benefit from increased cross-site generalizability. PURPOSE: To develop and evaluate a deep learning-based image harmonization method to improve cross-site generalizability of deep learning age prediction. STUDY TYPE: Retrospective. POPULATION: Eight thousand eight hundred and seventy-six subjects from six sites. Harmonization models were trained using all subjects. Age prediction models were trained using 2739 subjects from a single site and tested using the remaining 6137 subjects from various other sites. FIELD STRENGTH/SEQUENCE: Brain imaging with magnetization prepared rapid acquisition with gradient echo or spoiled gradient echo sequences at 1.5 T and 3 T. ASSESSMENT: StarGAN v2, was used to perform a canonical mapping from diverse datasets to a reference domain to reduce site-based variation while preserving semantic information. Generalization performance of deep learning age prediction was evaluated using harmonized, histogram matched, and unharmonized data. STATISTICAL TESTS: Mean absolute error (MAE) and Pearson correlation between estimated age and biological age quantified the performance of the age prediction model. RESULTS: Our results indicated a substantial improvement in age prediction in out-of-sample data, with the overall MAE improving from 15.81 (±0.21) years to 11.86 (±0.11) with histogram matching to 7.21 (±0.22) years with generative adversarial network (GAN)-based harmonization. In the multisite case, across the 5 out-of-sample sites, MAE improved from 9.78 (±6.69) years to 7.74 (±3.03) years with histogram normalization to 5.32 (±4.07) years with GAN-based harmonization. DATA CONCLUSION: While further research is needed, GAN-based medical image harmonization appears to be a promising tool for improving cross-site deep learning generalization. LEVEL OF EVIDENCE: 4 TECHNICAL EFFICACY: Stage 1.


Subject(s)
Deep Learning , Adolescent , Brain/diagnostic imaging , Humans , Image Processing, Computer-Assisted/methods , Research Design , Retrospective Studies
13.
Med Image Anal ; 75: 102304, 2022 01.
Article in English | MEDLINE | ID: mdl-34818611

ABSTRACT

Disease heterogeneity is a significant obstacle to understanding pathological processes and delivering precision diagnostics and treatment. Clustering methods have gained popularity for stratifying patients into subpopulations (i.e., subtypes) of brain diseases using imaging data. However, unsupervised clustering approaches are often confounded by anatomical and functional variations not related to a disease or pathology of interest. Semi-supervised clustering techniques have been proposed to overcome this and, therefore, capture disease-specific patterns more effectively. An additional limitation of both unsupervised and semi-supervised conventional machine learning methods is that they typically model, learn and infer from data using a basis of feature sets pre-defined at a fixed anatomical or functional scale (e.g., atlas-based regions of interest). Herein we propose a novel method, "Multi-scAle heteroGeneity analysIs and Clustering" (MAGIC), to depict the multi-scale presentation of disease heterogeneity, which builds on a previously proposed semi-supervised clustering method, HYDRA. It derives multi-scale and clinically interpretable feature representations and exploits a double-cyclic optimization procedure to effectively drive identification of inter-scale-consistent disease subtypes. More importantly, to understand the conditions under which the clustering model can estimate true heterogeneity related to diseases, we conducted extensive and systematic semi-simulated experiments to evaluate the proposed method on a sizeable healthy control sample from the UK Biobank (N = 4403). We then applied MAGIC to imaging data from Alzheimer's disease (ADNI, N = 1728) and schizophrenia (PHENOM, N = 1166) patients to demonstrate its potential and challenges in dissecting the neuroanatomical heterogeneity of common brain diseases. Taken together, we aim to provide guidance regarding when such analyses can succeed or should be taken with caution. The code of the proposed method is publicly available at https://github.com/anbai106/MAGIC.


Subject(s)
Alzheimer Disease , Brain , Alzheimer Disease/diagnostic imaging , Brain/diagnostic imaging , Cluster Analysis , Humans , Supervised Machine Learning
14.
Article in English | MEDLINE | ID: mdl-36845412

ABSTRACT

Brain age prediction based on functional magnetic resonance imaging (fMRI) data has the potential to serve as a biomarker for quantifying brain health. To predict the brain age based on fMRI data robustly and accurately, we curated a large dataset (n = 4259) of fMRI scans from seven different data acquisition sites and computed personalized functional connectivity measures at multiple scales from each subject's fMRI scan. Particularly, we computed personalized large-scale functional networks and generated functional connectivity measures at multiple scales to characterize each fMRI scan. To account for inter-site effects on the functional connectivity measures, we harmonized the functional connectivity measures in their tangent space and then built brain age prediction models on the harmonized functional connectivity measures. We compared the brain age prediction models with alternatives that were built on the functional connectivity measures computed at a single scale and harmonized using different strategies. Comparison results have demonstrated that the best brain age prediction performance was achieved by the prediction model built on the multi-scale functional connectivity measures that were harmonized in tangent space, indicating that multi-scale functional connectivity measures provided richer information than those computed at any single scales and the harmonization of functional connectivity measures in tangent space improved the brain age prediction.

15.
Nat Commun ; 12(1): 7065, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34862382

ABSTRACT

Heterogeneity of brain diseases is a challenge for precision diagnosis/prognosis. We describe and validate Smile-GAN (SeMI-supervised cLustEring-Generative Adversarial Network), a semi-supervised deep-clustering method, which examines neuroanatomical heterogeneity contrasted against normal brain structure, to identify disease subtypes through neuroimaging signatures. When applied to regional volumes derived from T1-weighted MRI (two studies; 2,832 participants; 8,146 scans) including cognitively normal individuals and those with cognitive impairment and dementia, Smile-GAN identified four patterns or axes of neurodegeneration. Applying this framework to longitudinal data revealed two distinct progression pathways. Measures of expression of these patterns predicted the pathway and rate of future neurodegeneration. Pattern expression offered complementary performance to amyloid/tau in predicting clinical progression. These deep-learning derived biomarkers offer potential for precision diagnostics and targeted clinical trial recruitment.


Subject(s)
Alzheimer Disease/diagnosis , Brain/diagnostic imaging , Cognitive Dysfunction/diagnosis , Deep Learning , Image Processing, Computer-Assisted , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/physiopathology , Brain/physiopathology , Case-Control Studies , Cluster Analysis , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Female , Healthy Volunteers , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging/methods
16.
Alzheimers Dement ; 17(11): 1855-1867, 2021 11.
Article in English | MEDLINE | ID: mdl-34870371

ABSTRACT

We aimed to evaluate the value of ATN biomarker classification system (amyloid beta [A], pathologic tau [T], and neurodegeneration [N]) for predicting conversion from mild cognitive impairment (MCI) to dementia. In a sample of people with MCI (n = 415) we assessed predictive performance of ATN classification using empirical knowledge-based cut-offs for each component of ATN and compared it to two data-driven approaches, logistic regression and RUSBoost machine learning classifiers, which used continuous clinical or biomarker scores. In data-driven approaches, we identified ATN features that distinguish normals from individuals with dementia and used them to classify persons with MCI into dementia-like and normal groups. Both data-driven classification methods performed better than the empirical cut-offs for ATN biomarkers in predicting conversion to dementia. Classifiers that used clinical features performed as well as classifiers that used ATN biomarkers for prediction of progression to dementia. We discuss that data-driven modeling approaches can improve our ability to predict disease progression and might have implications in future clinical trials.


Subject(s)
Alzheimer Disease/classification , Biomarkers , Disease Progression , Machine Learning/classification , Aged , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Cognitive Dysfunction/pathology , Data Collection , Female , Humans , Male , tau Proteins/cerebrospinal fluid
17.
J Cardiovasc Magn Reson ; 23(1): 133, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34758821

ABSTRACT

BACKGROUND: Artificial intelligence can assist in cardiac image interpretation. Here, we achieved a substantial reduction in time required to read a cardiovascular magnetic resonance (CMR) study to estimate left atrial volume without compromising accuracy or reliability. Rather than deploying a fully automatic black-box, we propose to incorporate the automated LA volumetry into a human-centric interactive image-analysis process. METHODS AND RESULTS: Atri-U, an automated data analysis pipeline for long-axis cardiac cine images, computes the atrial volume by: (i) detecting the end-systolic frame, (ii) outlining the endocardial borders of the LA, (iii) localizing the mitral annular hinge points and constructing the longitudinal atrial diameters, equivalent to the usual workup done by clinicians. In every step human interaction is possible, such that the results provided by the algorithm can be accepted, corrected, or re-done from scratch. Atri-U was trained and evaluated retrospectively on a sample of 300 patients and then applied to a consecutive clinical sample of 150 patients with various heart conditions. The agreement of the indexed LA volume between Atri-U and two experts was similar to the inter-rater agreement between clinicians (average overestimation of 0.8 mL/m2 with upper and lower limits of agreement of - 7.5 and 5.8 mL/m2, respectively). An expert cardiologist blinded to the origin of the annotations rated the outputs produced by Atri-U as acceptable in 97% of cases for step (i), 94% for step (ii) and 95% for step (iii), which was slightly lower than the acceptance rate of the outputs produced by a human expert radiologist in the same cases (92%, 100% and 100%, respectively). The assistance of Atri-U lead to an expected reduction in reading time of 66%-from 105 to 34 s, in our in-house clinical setting. CONCLUSIONS: Our proposal enables automated calculation of the maximum LA volume approaching human accuracy and precision. The optional user interaction is possible at each processing step. As such, the assisted process sped up the routine CMR workflow by providing accurate, precise, and validated measurement results.


Subject(s)
Artificial Intelligence , Magnetic Resonance Imaging, Cine , Heart Atria/diagnostic imaging , Humans , Image Interpretation, Computer-Assisted , Magnetic Resonance Spectroscopy , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies
18.
Sci Rep ; 11(1): 14124, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34238951

ABSTRACT

Lobar cerebral microbleeds (CMBs) and localized non-hemorrhage iron deposits in the basal ganglia have been associated with brain aging, vascular disease and neurodegenerative disorders. Particularly, CMBs are small lesions and require multiple neuroimaging modalities for accurate detection. Quantitative susceptibility mapping (QSM) derived from in vivo magnetic resonance imaging (MRI) is necessary to differentiate between iron content and mineralization. We set out to develop a deep learning-based segmentation method suitable for segmenting both CMBs and iron deposits. We included a convenience sample of 24 participants from the MESA cohort and used T2-weighted images, susceptibility weighted imaging (SWI), and QSM to segment the two types of lesions. We developed a protocol for simultaneous manual annotation of CMBs and non-hemorrhage iron deposits in the basal ganglia. This manual annotation was then used to train a deep convolution neural network (CNN). Specifically, we adapted the U-Net model with a higher number of resolution layers to be able to detect small lesions such as CMBs from standard resolution MRI. We tested different combinations of the three modalities to determine the most informative data sources for the detection tasks. In the detection of CMBs using single class and multiclass models, we achieved an average sensitivity and precision of between 0.84-0.88 and 0.40-0.59, respectively. The same framework detected non-hemorrhage iron deposits with an average sensitivity and precision of about 0.75-0.81 and 0.62-0.75, respectively. Our results showed that deep learning could automate the detection of small vessel disease lesions and including multimodal MR data (particularly QSM) can improve the detection of CMB and non-hemorrhage iron deposits with sensitivity and precision that is compatible with use in large-scale research studies.


Subject(s)
Alzheimer Disease/diagnosis , Brain/diagnostic imaging , Image Interpretation, Computer-Assisted , Iron/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Cerebral Hemorrhage/diagnosis , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Female , Humans , Machine Learning , Magnetic Resonance Imaging/statistics & numerical data , Male , Neural Networks, Computer , Neuroimaging/statistics & numerical data
19.
Radiol Imaging Cancer ; 3(4): e200108, 2021 07.
Article in English | MEDLINE | ID: mdl-34296969

ABSTRACT

Purpose To test the hypothesis that combined features from MR and digital histopathologic images more accurately predict overall survival (OS) in patients with glioma compared with MRI or histopathologic features alone. Materials and Methods Multiparametric MR and histopathologic images in patients with a diagnosis of glioma (high- or low-grade glioma [HGG or LGG]) were obtained from The Cancer Imaging Archive (original images acquired 1983-2008). An extensive set of engineered features such as intensity, histogram, and texture were extracted from delineated tumor regions in MR and histopathologic images. Cox proportional hazard regression and support vector machine classification (SVC) models were applied to (a) MRI features only (MRIcox/svc), histopathologic features only (HistoPathcox/svc), and (c) combined MRI and histopathologic features (MRI+HistoPathcox/svc) and evaluated in a split train-test configuration. Results A total of 171 patients (mean age, 51 years ± 15; 91 men) were included with HGG (n = 75) and LGG (n = 96). Median OS was 467 days (range, 3-4752 days) for all patients, 350 days (range, 15-1561 days) for HGG, and 595 days (range, 3-4752 days) for LGG. The MRI+HistoPathcox model demonstrated higher concordance index (C-index) compared with MRIcox and HistoPathcox models on all patients (C-index, 0.79 vs 0.70 [P = .02; MRIcox] and 0.67 [P = .01; HistoPathcox]), patients with HGG (C-index, 0.78 vs 0.68 [P = .03; MRIcox] and 0.64 [P = .01; HistoPathcox]), and patients with LGG (C-index, 0.88 vs 0.62 [P = .008; MRIcox] and 0.62 [P = .006; HistoPathcox]). In binary classification, the MRI+HistoPathsvc model (area under the receiver operating characteristic curve [AUC], 0.86 [95% CI: 0.80, 0.95]) had higher performance than the MRIsvc model (AUC, 0.68 [95% CI: 0.50, 0.81]; P = .01) and the HistoPathsvc model (AUC, 0.72 [95% CI: 0.60, 0.85]; P = .04). Conclusion The model combining features from MR and histopathologic images had higher accuracy in predicting OS compared with the models with MR or histopathologic images alone. Keywords: Survival Prediction, Gliomas, Digital Pathology Imaging, MR Imaging, Machine Learning Supplemental material is available for this article.


Subject(s)
Brain Neoplasms , Glioma , Brain Neoplasms/diagnostic imaging , Glioma/diagnostic imaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Grading , Support Vector Machine
20.
J Am Geriatr Soc ; 69(1): 106-113, 2021 01.
Article in English | MEDLINE | ID: mdl-32936455

ABSTRACT

BACKGROUND/OBJECTIVES: The diagnostic process in a university memory clinic is based largely on cognitive testing. However, input from informants, acquired through interview or questionnaires, may significantly impact diagnosis. We sought to evaluate whether informant questionnaires for basic and instrumental activities of daily living, or for identifying progressive cognitive decline would improve diagnostic predictability of neurodegenerative disorders compared with either the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropsychological test battery or the Mini-Mental State Examination score alone. DESIGN: Retrospective data analysis using logit models. SETTING: University hospital outpatient memory clinic. PARTICIPANTS: A total of 394 patients with dementia, mild cognitive impairment (MCI), depression, or subjective cognitive impairment were assessed. MEASUREMENTS: Bristol Activities of Daily Living Scale, Functional Activities Questionnaire, Informant Questionnaire on Cognitive Decline in the Elderly, and the Physical Self-Maintenance Scale questionnaires were obtained. Analyses through logit models were performed to predict outcome diagnoses, based on cognitive scores alone or in combination with one or more informant questionnaires. RESULTS: The four questionnaires were highly correlated (.31-.86). The addition of informant questionnaires improved diagnostic predictability between differential diagnoses of MCI and dementia, or dementia and depression. However, the misprediction rate was reduced by up to 6 percentage points only. Adding more than one questionnaire or all CERAD subtests instead of their sum score never improved prediction in regularized logit models to a clinically relevant extent. CONCLUSION: Although questionnaires contribute to a statistically better prediction of the outcome diagnosis, for some sets of differential diagnoses, the benefit may not be clinically pertinent when routine semistructured informant interviews are used by trained personnel. However, standardized assessment, particularly when patients are seen longitudinally, should not be underestimated.


Subject(s)
Cognitive Dysfunction/diagnosis , Dementia/diagnosis , Diagnosis, Differential , Neuropsychological Tests/statistics & numerical data , Surveys and Questionnaires , Activities of Daily Living/psychology , Aged , Aged, 80 and over , Caregivers/psychology , Female , Humans , Male , Models, Statistical , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...