Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Front Aging Neurosci ; 16: 1356745, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38813529

RESUMEN

Objectives: Accurately predicting when patients with mild cognitive impairment (MCI) will progress to dementia is a formidable challenge. This work aims to develop a predictive deep learning model to accurately predict future cognitive decline and magnetic resonance imaging (MRI) marker changes over time at the individual level for patients with MCI. Methods: We recruited 657 amnestic patients with MCI from the Samsung Medical Center who underwent cognitive tests, brain MRI scans, and amyloid-ß (Aß) positron emission tomography (PET) scans. We devised a novel deep learning architecture by leveraging an attention mechanism in a recurrent neural network. We trained a predictive model by inputting age, gender, education, apolipoprotein E genotype, neuropsychological test scores, and brain MRI and amyloid PET features. Cognitive outcomes and MRI features of an MCI subject were predicted using the proposed network. Results: The proposed predictive model demonstrated good prediction performance (AUC = 0.814 ± 0.035) in five-fold cross-validation, along with reliable prediction in cognitive decline and MRI markers over time. Faster cognitive decline and brain atrophy in larger regions were forecasted in patients with Aß (+) than with Aß (-). Conclusion: The proposed method provides effective and accurate means for predicting the progression of individuals within a specific period. This model could assist clinicians in identifying subjects at a higher risk of rapid cognitive decline by predicting future cognitive decline and MRI marker changes over time for patients with MCI. Future studies should validate and refine the proposed predictive model further to improve clinical decision-making.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38771688

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative condition that precedes progressive and irreversible dementia; thus, predicting its progression over time is vital for clinical diagnosis and treatment. For this, numerous studies have implemented structural magnetic resonance imaging (MRI) to model AD progression, focusing on three integral aspects: 1) temporal variability; 2) incomplete observations; and 3) temporal geometric characteristics. However, many pioneer deep learning-based approaches addressing data variability and sparsity have yet to consider inherent geometrical properties sufficiently. These properties are integral to modeling as they correlate with brain region size, thickness, volume, and shape in AD progression. The ordinary differential equation-based geometric modeling method (ODE-RGRU) has recently emerged as a promising strategy for modeling time-series data by intertwining a recurrent neural network (RNN) and an ODE in Riemannian space. Despite its achievements, ODE-RGRU encounters limitations when extrapolating positive definite symmetric matrices from incomplete samples, leading to feature reverse occurrences that are particularly problematic, especially within the clinical facet. Therefore, this study proposes a novel geometric learning approach that models longitudinal MRI biomarkers and cognitive scores by combining three modules: topological space shift, ODE-RGRU, and trajectory estimation. We have also developed a training algorithm that integrates the manifold mapping with monotonicity constraints to reflect measurement transition irreversibility. We verify our proposed method's efficacy by predicting clinical labels and cognitive scores over time in regular and irregular settings. Furthermore, we thoroughly analyze our proposed framework through an ablation study.

3.
IEEE Trans Med Imaging ; 43(4): 1400-1411, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38015693

RESUMEN

Deep learning models based on resting-state functional magnetic resonance imaging (rs-fMRI) have been widely used to diagnose brain diseases, particularly autism spectrum disorder (ASD). Existing studies have leveraged the functional connectivity (FC) of rs-fMRI, achieving notable classification performance. However, they have significant limitations, including the lack of adequate information while using linear low-order FC as inputs to the model, not considering individual characteristics (i.e., different symptoms or varying stages of severity) among patients with ASD, and the non-explainability of the decision process. To cover these limitations, we propose a novel explainability-guided region of interest (ROI) selection (EAG-RS) framework that identifies non-linear high-order functional associations among brain regions by leveraging an explainable artificial intelligence technique and selects class-discriminative regions for brain disease identification. The proposed framework includes three steps: (i) inter-regional relation learning to estimate non-linear relations through random seed-based network masking, (ii) explainable connection-wise relevance score estimation to explore high-order relations between functional connections, and (iii) non-linear high-order FC-based diagnosis-informative ROI selection and classifier learning to identify ASD. We validated the effectiveness of our proposed method by conducting experiments using the Autism Brain Imaging Database Exchange (ABIDE) dataset, demonstrating that the proposed method outperforms other comparative methods in terms of various evaluation metrics. Furthermore, we qualitatively analyzed the selected ROIs and identified ASD subtypes linked to previous neuroscientific studies.


Asunto(s)
Trastorno del Espectro Autista , Humanos , Trastorno del Espectro Autista/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Inteligencia Artificial , Encéfalo/diagnóstico por imagen , Mapeo Encefálico/métodos
4.
Schizophr Bull ; 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38085061

RESUMEN

BACKGROUND AND HYPOTHESIS: The brain-predicted age difference (brain-PAD) may serve as a biomarker for neurodegeneration. We investigated the brain-PAD in patients with schizophrenia (SCZ), first-episode schizophrenia spectrum disorders (FE-SSDs), and treatment-resistant schizophrenia (TRS) using structural magnetic resonance imaging (sMRI). STUDY DESIGN: We employed a convolutional network-based regression (SFCNR), and compared its performance with models based on three machine learning (ML) algorithms. We pretrained the SFCNR with sMRI data of 7590 healthy controls (HCs) selected from the UK Biobank. The parameters of the pretrained model were transferred to the next training phase with a new set of HCs (n = 541). The brain-PAD was analyzed in independent HCs (n = 209) and patients (n = 233). Correlations between the brain-PAD and clinical measures were investigated. STUDY RESULTS: The SFCNR model outperformed three commonly used ML models. Advanced brain aging was observed in patients with SCZ, FE-SSDs, and TRS compared to HCs. A significant difference in brain-PAD was observed between FE-SSDs and TRS with ridge regression but not with the SFCNR model. Chlorpromazine equivalent dose and cognitive function were correlated with the brain-PAD in SCZ and FE-SSDs. CONCLUSIONS: Our findings indicate that there is advanced brain aging in patients with SCZ and higher brain-PAD in SCZ can be used as a surrogate marker for cognitive dysfunction. These findings warrant further investigations on the causes of advanced brain age in SCZ. In addition, possible psychosocial and pharmacological interventions targeting brain health should be considered in early-stage SCZ patients with advanced brain age.

5.
Sci Rep ; 13(1): 18588, 2023 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-37903879

RESUMEN

Weakly supervised object localization tasks remain challenging to identify and segment an entire object rather than only discriminative parts of the object. To tackle this problem, corruption-based approaches have been devised, which involve the training of non-discriminative regions by corrupting (e.g., erasing) the input images or intermediate feature maps. However, this approach requires an additional hyperparameter, the corrupting threshold, to determine the degree of corruption and can unfavorably disrupt training. It also tends to localize object regions coarsely. In this paper, we propose a novel approach, Module of Axis-based Nexus Attention (MoANA), which helps to adaptively activate less discriminative regions along with the class-discriminative regions without an additional hyperparameter, and elaborately localizes an entire object. Specifically, MoANA consists of three mechanisms (1) triple-view attentions representation, (2) attentions expansion, and (3) features calibration mechanism. Unlike other attention-based methods that train a coarse attention map with the same values across elements in feature maps, MoANA trains fine-grained values in an attention map by assigning different attention values to each element. We validated MoANA by comparing it with various methods. We also analyzed the effect of each component in MoANA and visualized attention maps to provide insights into the calibration.

6.
Artículo en Inglés | MEDLINE | ID: mdl-37738193

RESUMEN

Transfer learning has attracted considerable attention in medical image analysis because of the limited number of annotated 3-D medical datasets available for training data-driven deep learning models in the real world. We propose Medical Transformer, a novel transfer learning framework that effectively models 3-D volumetric images as a sequence of 2-D image slices. To improve the high-level representation in 3-D-form empowering spatial relations, we use a multiview approach that leverages information from three planes of the 3-D volume, while providing parameter-efficient training. For building a source model generally applicable to various tasks, we pretrain the model using self-supervised learning (SSL) for masked encoding vector prediction as a proxy task, using a large-scale normal, healthy brain magnetic resonance imaging (MRI) dataset. Our pretrained model is evaluated on three downstream tasks: 1) brain disease diagnosis; 2) brain age prediction; and 3) brain tumor segmentation, which are widely studied in brain MRI research. Experimental results demonstrate that our Medical Transformer outperforms the state-of-the-art (SOTA) transfer learning methods, efficiently reducing the number of parameters by up to approximately 92% for classification and regression tasks and 97% for segmentation task, and it also achieves good performance in scenarios where only partial training samples are used.

7.
Artículo en Inglés | MEDLINE | ID: mdl-37708014

RESUMEN

Large amounts of fMRI data are essential to building generalized predictive models for brain disease diagnosis. In order to conduct extensive data analysis, it is often necessary to gather data from multiple organizations. However, the site variation inherent in multisite resting-state functional magnetic resonance imaging (rs-fMRI) leads to unfavorable heterogeneity in data distribution, negatively impacting the identification of biomarkers and the diagnostic decision. Several existing methods have alleviated this shift of domain distribution (i.e., multisite problem). Statistical tuning schemes directly regress out site disparity factors from the data prior to model training. Such methods have a limitation in processing data each time through variance estimation according to the added site. In the model adjustment approaches, domain adaptation (DA) methods adjust the features or models of the source domain according to the target domain during model training. Thus, it is inevitable that it needs updating model parameters according to the samples of a target site, causing great limitations in practical applicability. Meanwhile, the approach of domain generalization (DG) aims to create a universal model that can be quickly adapted to multiple domains. In this study, we propose a novel framework for disease diagnosis that alleviates the multisite problem by adaptively calibrating site-specific features into site-invariant features. Specifically, it applies directly to samples from unseen sites without the need for fine-tuning. With a learning-to-learn strategy that learns how to calibrate the features under the various domain shift environments, our novel modulation mechanism extracts site-invariant features. In our experiments over the Autism Brain Imaging Data Exchange (ABIDE I and II) dataset, we validated the generalization ability of the proposed network by improving diagnostic accuracy in both seen and unseen multisite samples.

8.
Sci Rep ; 13(1): 11664, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468538

RESUMEN

The identification of Alzheimer's disease (AD) using structural magnetic resonance imaging (sMRI) has been studied based on the subtle morphological changes in the brain. One of the typical approaches is a deep learning-based patch-level feature representation. For this approach, however, the predetermined patches before learning the diagnostic model can limit classification performance. To mitigate this problem, we propose the BrainBagNet with a position-based gate (PG), which applies position information of brain images represented through the 3D coordinates. Our proposed method represents the patch-level class evidence based on both MR scan and position information for image-level prediction. To validate the effectiveness of our proposed framework, we conducted comprehensive experiments comparing it with state-of-the-art methods, utilizing two publicly available datasets: the Alzheimer's Disease Neuroimaging Initiative (ADNI) and the Australian Imaging, Biomarkers and Lifestyle (AIBL) dataset. Furthermore, our experimental results demonstrate that our proposed method outperforms the existing competing methods in terms of classification performance for both AD diagnosis and mild cognitive impairment conversion prediction tasks. In addition, we performed various analyses of the results from diverse perspectives to obtain further insights into the underlying mechanisms and strengths of our proposed framework. Based on the results of our experiments, we demonstrate that our proposed framework has the potential to advance deep-learning-based patch-level feature representation studies for AD diagnosis and MCI conversion prediction. In addition, our method provides valuable insights, such as interpretability, and the ability to capture subtle changes, into the underlying pathological processes of AD and MCI, benefiting both researchers and clinicians.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/patología , Australia , Imagen por Resonancia Magnética/métodos , Neuroimagen , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/patología
9.
Neuroimage ; 273: 120073, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37037063

RESUMEN

Identifying Alzheimer's disease (AD) involves a deliberate diagnostic process owing to its innate traits of irreversibility with subtle and gradual progression. These characteristics make AD biomarker identification from structural brain imaging (e.g., structural MRI) scans quite challenging. Using clinically-guided prototype learning, we propose a novel deep-learning approach through eXplainable AD Likelihood Map Estimation (XADLiME) for AD progression modeling over 3D sMRIs. Specifically, we establish a set of topologically-aware prototypes onto the clusters of latent clinical features, uncovering an AD spectrum manifold. Considering this pseudo map as an enriched reference, we employ an estimating network to approximate the AD likelihood map over a 3D sMRI scan. Additionally, we promote the explainability of such a likelihood map by revealing a comprehensible overview from clinical and morphological perspectives. During the inference, this estimated likelihood map served as a substitute for unseen sMRI scans for effectively conducting the downstream task while providing thorough explainable states.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Aprendizaje , Biomarcadores , Neuroimagen/métodos
10.
Eur Radiol ; 33(9): 5924-5932, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37012546

RESUMEN

OBJECTIVES: We aimed to evaluate the effect of hepatic steatosis (HS) on liver volume and to develop a formula to estimate lean liver volume correcting the HS effect. METHODS: This retrospective study included healthy adult liver donors who underwent gadoxetic acid-enhanced MRI and proton density fat fraction (PDFF) measurement from 2015 to 2019. The degree of HS was graded at 5% PDFF intervals from grade 0 (no HS; PDFF < 5.5%). Liver volume was measured with hepatobiliary phase MRI using deep learning algorithm, and standard liver volume (SLV) was calculated as the reference lean liver volume. The association between liver volume and SLV ratio with PDFF grades was evaluated using Spearman's correlation (ρ). The effect of PDFF grades on liver volume was evaluated using the multivariable linear regression model. RESULTS: The study population included 1038 donors (mean age, 31 ± 9 years; 689 men). Mean liver volume to SLV ratio increased according to PDFF grades (ρ = 0.234, p < 0.001). The multivariable analysis indicated that SLV (ß = 1.004, p < 0.001) and PDFF grade*SLV (ß = 0.044, p < 0.001) independently affected liver volume, suggesting a 4.4% increase in liver volume per one-point increment in the PDFF grade. PDFF-adjusted lean liver volume was estimated using the formula, liver volume/[1.004 + 0.044 × PDFF grade]. The mean estimated lean liver volume to SLV ratio approximated to one for all PDFF grades, with no significant association with PDFF grades (p = 0.851). CONCLUSION: HS increases liver volume. The formula to estimate lean liver volume may be useful to adjust for the effect of HS on liver volume. KEY POINTS: • Hepatic steatosis increases liver volume. • The presented formula to estimate lean liver volume using MRI-measured proton density fat fraction and liver volume may be useful to adjust for the effect of hepatic steatosis on measured liver volume.


Asunto(s)
Aprendizaje Profundo , Enfermedad del Hígado Graso no Alcohólico , Adulto , Masculino , Humanos , Adulto Joven , Protones , Enfermedad del Hígado Graso no Alcohólico/diagnóstico por imagen , Estudios Retrospectivos , Hígado/diagnóstico por imagen , Imagen por Resonancia Magnética
11.
IEEE Trans Pattern Anal Mach Intell ; 45(4): 4843-4857, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35947563

RESUMEN

Existing studies on disease diagnostic models focus either on diagnostic model learning for performance improvement or on the visual explanation of a trained diagnostic model. We propose a novel learn-explain-reinforce (LEAR) framework that unifies diagnostic model learning, visual explanation generation (explanation unit), and trained diagnostic model reinforcement (reinforcement unit) guided by the visual explanation. For the visual explanation, we generate a counterfactual map that transforms an input sample to be identified as an intended target label. For example, a counterfactual map can localize hypothetical abnormalities within a normal brain image that may cause it to be diagnosed with Alzheimer's disease (AD). We believe that the generated counterfactual maps represent data-driven knowledge about a target task, i.e., AD diagnosis using structural MRI, which can be a vital source of information to reinforce the generalization of the trained diagnostic model. To this end, we devise an attention-based feature refinement module with the guidance of the counterfactual maps. The explanation and reinforcement units are reciprocal and can be operated iteratively. Our proposed approach was validated via qualitative and quantitative analysis on the ADNI dataset. Its comprehensibility and fidelity were demonstrated through ablation studies and comparisons with existing methods.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Algoritmos , Aprendizaje , Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética , Neuroimagen
12.
Int J Neuropsychopharmacol ; 26(3): 207-216, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36545813

RESUMEN

BACKGROUND: Brain age is a popular brain-based biomarker that offers a powerful strategy for using neuroscience in clinical practice. We investigated the brain-predicted age difference (PAD) in patients with schizophrenia (SCZ), first-episode schizophrenia spectrum disorders (FE-SSDs), and treatment-resistant schizophrenia (TRS) using structural magnetic resonance imaging data. The association between brain-PAD and clinical parameters was also assessed. METHODS: We developed brain age prediction models for the association between 77 average structural brain measures and age in a training sample of controls (HCs) using ridge regression, support vector regression, and relevance vector regression. The trained models in the controls were applied to the test samples of the controls and 3 patient groups to obtain brain-based age estimates. The correlations were tested between the brain PAD and clinical measures in the patient groups. RESULTS: Model performance indicated that, regardless of the type of regression metric, the best model was support vector regression and the worst model was relevance vector regression for the training HCs. Accelerated brain aging was identified in patients with SCZ, FE-SSDs, and TRS compared with the HCs. A significant difference in brain PAD was observed between FE-SSDs and TRS using the ridge regression algorithm. Symptom severity, the Social and Occupational Functioning Assessment Scale, chlorpromazine equivalents, and cognitive function were correlated with the brain PAD in the patient groups. CONCLUSIONS: These findings suggest additional progressive neuronal changes in the brain after SCZ onset. Therefore, pharmacological or psychosocial interventions targeting brain health should be developed and provided during the early course of SCZ.


Asunto(s)
Esquizofrenia , Humanos , Esquizofrenia/diagnóstico por imagen , Esquizofrenia/tratamiento farmacológico , Esquizofrenia Resistente al Tratamiento , Encéfalo , Envejecimiento/fisiología , Imagen por Resonancia Magnética/métodos
13.
IEEE Trans Neural Netw Learn Syst ; 34(2): 739-749, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-34357871

RESUMEN

In recent years, deep learning-based feature representation methods have shown a promising impact on electroencephalography (EEG)-based brain-computer interface (BCI). Nonetheless, owing to high intra- and inter-subject variabilities, many studies on decoding EEG were designed in a subject-specific manner by using calibration samples, with no concern of its practical use, hampered by time-consuming steps and a large data requirement. To this end, recent studies adopted a transfer learning strategy, especially domain adaptation techniques. Among those, we have witnessed the potential of adversarial learning-based transfer learning in BCIs. In the meantime, it is known that adversarial learning-based domain adaptation methods are prone to negative transfer that disrupts learning generalized feature representations, applicable to diverse domains, for example, subjects or sessions in BCIs. In this article, we propose a novel framework that learns class-relevant and subject-invariant feature representations in an information-theoretic manner, without using adversarial learning. To be specific, we devise two operational components in a deep network that explicitly estimate mutual information between feature representations: 1) to decompose features in an intermediate layer into class-relevant and class-irrelevant ones and 2) to enrich class-discriminative feature representation. On two large EEG datasets, we validated the effectiveness of our proposed framework by comparing with several comparative methods in performance. Furthermore, we conducted rigorous analyses by performing an ablation study in regard to the components in our network, explaining our model's decision on input EEG signals via layer-wise relevance propagation, and visualizing the distribution of learned features via t-SNE.


Asunto(s)
Interfaces Cerebro-Computador , Redes Neurales de la Computación , Humanos , Aprendizaje Automático , Electroencefalografía/métodos , Calibración , Algoritmos
14.
IEEE Trans Cybern ; 53(7): 4500-4510, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36063512

RESUMEN

Sleep staging is essential for sleep assessment and plays a vital role as a health indicator. Many recent studies have devised various machine/deep learning methods for sleep staging. However, two key challenges hinder the practical use of those methods: 1) effectively capturing salient waveforms in sleep signals and 2) correctly classifying confusing stages in transitioning epochs. In this study, we propose a novel deep neural-network structure, TransSleep, that captures distinctive local temporal patterns and distinguishes confusing stages using two auxiliary tasks. In particular, TransSleep captures salient waveforms in sleep signals by an attention-based multiscale feature extractor and correctly classifies confusing stages in transitioning epochs, while modeling contextual relationships with two auxiliary tasks. Results show that TransSleep achieves promising performance in automatic sleep staging. The validity of TransSleep is demonstrated by its state-of-the-art performance on two publicly available datasets: 1) Sleep-EDF and 2) MASS. Furthermore, we performed ablations to analyze our results from different perspectives. Based on our overall results, we believe that TransSleep has immense potential to provide new insights into deep-learning-based sleep staging.


Asunto(s)
Electroencefalografía , Redes Neurales de la Computación , Electroencefalografía/métodos , Fases del Sueño , Sueño , Aprendizaje Automático
15.
Korean J Radiol ; 23(12): 1269-1280, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36447415

RESUMEN

OBJECTIVE: This study aimed to evaluate the usefulness of quantitative indices obtained from deep learning analysis of gadoxetic acid-enhanced hepatobiliary phase (HBP) MRI and their longitudinal changes in predicting decompensation and death in patients with advanced chronic liver disease (ACLD). MATERIALS AND METHODS: We included patients who underwent baseline and 1-year follow-up MRI from a prospective cohort that underwent gadoxetic acid-enhanced MRI for hepatocellular carcinoma surveillance between November 2011 and August 2012 at a tertiary medical center. Baseline liver condition was categorized as non-ACLD, compensated ACLD, and decompensated ACLD. The liver-to-spleen signal intensity ratio (LS-SIR) and liver-to-spleen volume ratio (LS-VR) were automatically measured on the HBP images using a deep learning algorithm, and their percentage changes at the 1-year follow-up (ΔLS-SIR and ΔLS-VR) were calculated. The associations of the MRI indices with hepatic decompensation and a composite endpoint of liver-related death or transplantation were evaluated using a competing risk analysis with multivariable Fine and Gray regression models, including baseline parameters alone and both baseline and follow-up parameters. RESULTS: Our study included 280 patients (153 male; mean age ± standard deviation, 57 ± 7.95 years) with non-ACLD, compensated ACLD, and decompensated ACLD in 32, 186, and 62 patients, respectively. Patients were followed for 11-117 months (median, 104 months). In patients with compensated ACLD, baseline LS-SIR (sub-distribution hazard ratio [sHR], 0.81; p = 0.034) and LS-VR (sHR, 0.71; p = 0.01) were independently associated with hepatic decompensation. The ΔLS-VR (sHR, 0.54; p = 0.002) was predictive of hepatic decompensation after adjusting for baseline variables. ΔLS-VR was an independent predictor of liver-related death or transplantation in patients with compensated ACLD (sHR, 0.46; p = 0.026) and decompensated ACLD (sHR, 0.61; p = 0.023). CONCLUSION: MRI indices automatically derived from the deep learning analysis of gadoxetic acid-enhanced HBP MRI can be used as prognostic markers in patients with ACLD.


Asunto(s)
Carcinoma Hepatocelular , Aprendizaje Profundo , Neoplasias Hepáticas , Humanos , Masculino , Estudios Prospectivos , Imagen por Resonancia Magnética , Neoplasias Hepáticas/diagnóstico por imagen
16.
Psychiatry Res ; 317: 114871, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36209668

RESUMEN

BACKGROUND: Few studies have investigated functional connectivity (FC) in patients with psychotic disorder not otherwise specified (PNOS). We sought to identify distinct FC differentiating PNOS from schizophrenia (SZ). METHODS: In total, 49 patients with PNOS, 42 with SZ, and 55 healthy controls (HC) matched for age, sex, and education underwent functional magnetic resonance imaging (fMRI) brain scans and clinical evaluation. Using six functional networks consisting of 40 regions of interest (ROIs), we conducted ROI to ROI and intra- and inter-network FC analyses using resting-state fMRI (rs-fMRI) data. Correlations of altered FC with symptomatology were explored. RESULTS: We found common brain connectomics in PNOS and SZ including thalamo-cortical (especially superior temporal gyrus) hyperconnectivity, thalamo-cerebellar hypoconnectivity, and reduced within-thalamic connectivity compared to HC. Additionally, features differentiating the two patient groups included hyperconnectivity between the thalamic subregion and anterior cingulate cortex in PNOS compared to SZ and hyperconnectivity of the thalamic subregions with the posterior cingulate cortex and precentral gyrus in SZ compared to PNOS. CONCLUSIONS: These findings suggest that PNOS and SZ exhibit both common and differentiating changes in neuronal connectivity. Furthermore, they may support the hypothesis that PNOS should be treated as a separate clinical syndrome with distinct neural connectomics.


Asunto(s)
Conectoma , Trastornos Psicóticos , Esquizofrenia , Humanos , Mapeo Encefálico , Tálamo/diagnóstico por imagen , Conectoma/métodos , Imagen por Resonancia Magnética , Encéfalo
17.
IEEE J Biomed Health Inform ; 26(8): 4270-4280, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35511839

RESUMEN

Electronic health record (EHR) data are sparse and irregular as they are recorded at irregular time intervals, and different clinical variables are measured at each observation point. In this work, to handle irregular multivariate time-series data, we consider the human knowledge of the aspects to be measured and time to measure them in different situations, known as multi-view features, which are indirectly represented in the data. We propose a scheme to realize multi-view features integration learning via a self-attention mechanism. Specifically, we devise a novel multi-integration attention module (MIAM) to extract complex information that is inherent in irregular time-series data. We explicitly learn the relationships among the observed values, missing indicators, and time interval between the consecutive observations in a simultaneous manner. In addition, we build an attention-based decoder as a missing value imputer that helps empower the representation learning of the interrelations among multi-view observations for the prediction task this decoder operates only in the training phase so that the final model is implemented in an imputation-free manner. We validated the effectiveness of our method over the public MIMIC-III and PhysioNet challenge 2012 datasets by comparing with and outperforming the state-of-the-art methods in three downstream tasks i.e., prediction of the in-hospital mortality, prediction of the length of stay, and phenotyping. Moreover, we conduct a layer-wise relevance propagation (LRP) analysis based on case studies to highlight the explainability of the trained model.


Asunto(s)
Registros Electrónicos de Salud , Humanos
18.
Korean J Radiol ; 23(7): 720-731, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35434977

RESUMEN

OBJECTIVE: We aimed to develop and test a deep learning algorithm (DLA) for fully automated measurement of the volume and signal intensity (SI) of the liver and spleen using gadoxetic acid-enhanced hepatobiliary phase (HBP)-magnetic resonance imaging (MRI) and to evaluate the clinical utility of DLA-assisted assessment of functional liver capacity. MATERIALS AND METHODS: The DLA was developed using HBP-MRI data from 1014 patients. Using an independent test dataset (110 internal and 90 external MRI data), the segmentation performance of the DLA was measured using the Dice similarity score (DSS), and the agreement between the DLA and the ground truth for the volume and SI measurements was assessed with a Bland-Altman 95% limit of agreement (LOA). In 276 separate patients (male:female, 191:85; mean age ± standard deviation, 40 ± 15 years) who underwent hepatic resection, we evaluated the correlations between various DLA-based MRI indices, including liver volume normalized by body surface area (LVBSA), liver-to-spleen SI ratio (LSSR), MRI parameter-adjusted LSSR (aLSSR), LSSR × LVBSA, and aLSSR × LVBSA, and the indocyanine green retention rate at 15 minutes (ICG-R15), and determined the diagnostic performance of the DLA-based MRI indices to detect ICG-R15 ≥ 20%. RESULTS: In the test dataset, the mean DSS was 0.977 for liver segmentation and 0.946 for spleen segmentation. The Bland-Altman 95% LOAs were 0.08% ± 3.70% for the liver volume, 0.20% ± 7.89% for the spleen volume, -0.02% ± 1.28% for the liver SI, and -0.01% ± 1.70% for the spleen SI. Among DLA-based MRI indices, aLSSR × LVBSA showed the strongest correlation with ICG-R15 (r = -0.54, p < 0.001), with area under receiver operating characteristic curve of 0.932 (95% confidence interval, 0.895-0.959) to diagnose ICG-R15 ≥ 20%. CONCLUSION: Our DLA can accurately measure the volume and SI of the liver and spleen and may be useful for assessing functional liver capacity using gadoxetic acid-enhanced HBP-MRI.


Asunto(s)
Aprendizaje Profundo , Neoplasias Hepáticas , Adulto , Medios de Contraste , Femenino , Gadolinio DTPA , Humanos , Hígado/diagnóstico por imagen , Hígado/patología , Neoplasias Hepáticas/patología , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
19.
Diagnostics (Basel) ; 12(3)2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35328143

RESUMEN

CT volumetry (CTV) has been widely used for pre-operative graft weight (GW) estimation in living-donor liver transplantation (LDLT), and the use of a deep-learning algorithm (DLA) may further improve its efficiency. However, its accuracy has not been well determined. To evaluate the efficiency and accuracy of DLA-assisted CTV in GW estimation, we performed a retrospective study including 581 consecutive LDLT donors who donated a right-lobe graft. Right-lobe graft volume (GV) was measured on CT using the software implemented with the DLA for automated liver segmentation. In the development group (n = 207), a volume-to-weight conversion formula was constructed by linear regression analysis between the CTV-measured GV and the intraoperative GW. In the validation group (n = 374), the agreement between the estimated and measured GWs was assessed using the Bland-Altman 95% limit-of-agreement (LOA). The mean process time for GV measurement was 1.8 ± 0.6 min (range, 1.3-8.0 min). In the validation group, the GW was estimated using the volume-to-weight conversion formula (estimated GW [g] = 206.3 + 0.653 × CTV-measured GV [mL]), and the Bland-Altman 95% LOA between the estimated and measured GWs was -1.7% ± 17.1%. The DLA-assisted CT volumetry allows for time-efficient and accurate estimation of GW in LDLT.

20.
IEEE Trans Med Imaging ; 41(9): 2348-2359, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35344489

RESUMEN

Imaging genetics, one of the foremost emerging topics in the medical imaging field, analyzes the inherent relations between neuroimaging and genetic data. As deep learning has gained widespread acceptance in many applications, pioneering studies employed deep learning frameworks for imaging genetics. However, existing approaches suffer from some limitations. First, they often adopt a simple strategy for joint learning of phenotypic and genotypic features. Second, their findings have not been extended to biomedical applications, e.g., degenerative brain disease diagnosis and cognitive score prediction. Finally, existing studies perform insufficient and inappropriate analyses from the perspective of data science and neuroscience. In this work, we propose a novel deep learning framework to simultaneously tackle the aforementioned issues. Our proposed framework learns to effectively represent the neuroimaging and the genetic data jointly, and achieves state-of-the-art performance when used for Alzheimer's disease and mild cognitive impairment identification. Furthermore, unlike the existing methods, the framework enables learning the relation between imaging phenotypes and genotypes in a nonlinear way without any prior neuroscientific knowledge. To demonstrate the validity of our proposed framework, we conducted experiments on a publicly available dataset and analyzed the results from diverse perspectives. Based on our experimental results, we believe that the proposed framework has immense potential to provide new insights and perspectives in deep learning-based imaging genetics studies.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Encéfalo/diagnóstico por imagen , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/genética , Aprendizaje Discriminativo , Humanos , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...