[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (64)

Search Parameters:
Keywords = DID

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1465 KiB  
Article
Alzheimer’s Multiclassification Using Explainable AI Techniques
by Kamese Jordan Junior, Kouayep Sonia Carole, Tagne Poupi Theodore Armand, Hee-Cheol Kim and The Alzheimer’s Disease Neuroimaging Initiative
Appl. Sci. 2024, 14(18), 8287; https://doi.org/10.3390/app14188287 (registering DOI) - 14 Sep 2024
Viewed by 365
Abstract
In this study, we address the early detection challenges of Alzheimer’s disease (AD) using explainable artificial intelligence (XAI) techniques. AD, characterized by amyloid plaques and tau tangles, leads to cognitive decline and remains hard to diagnose due to genetic and environmental factors. Utilizing [...] Read more.
In this study, we address the early detection challenges of Alzheimer’s disease (AD) using explainable artificial intelligence (XAI) techniques. AD, characterized by amyloid plaques and tau tangles, leads to cognitive decline and remains hard to diagnose due to genetic and environmental factors. Utilizing deep learning models, we analyzed brain MRI scans from the ADNI database, categorizing them into normal cognition (NC), mild cognitive impairment (MCI), and AD. The ResNet-50 architecture was employed, enhanced by a channel-wise attention mechanism to improve feature extraction. To ensure model transparency, we integrated local interpretable model-agnostic explanations (LIMEs) and gradient-weighted class activation mapping (Grad-CAM), highlighting significant image regions contributing to predictions. Our model achieved 85% accuracy, effectively distinguishing between the classes. The LIME and Grad-CAM visualizations provided insights into the model’s decision-making process, particularly emphasizing changes near the hippocampus for MCI. These XAI methods enhance the interpretability of AI-driven AD diagnosis, fostering trust and aiding clinical decision-making. Our approach demonstrates the potential of combining deep learning with XAI for reliable and transparent medical applications. Full article
(This article belongs to the Special Issue Future Information & Communication Engineering 2024)
Show Figures

Figure 1

Figure 1
<p>Showing the system model and workflow.</p>
Full article ">Figure 2
<p>Raw MRI samples for normal cognition (NC) in the first row, mild cognitive impairment (MCI) in the middle row, and Alzheimer’s disease (AD) in the bottom row.</p>
Full article ">Figure 3
<p>Model accuracy against the count of epochs during training and validation.</p>
Full article ">Figure 4
<p>Confusion matrix for the pre-trained model.</p>
Full article ">Figure 5
<p>Output prediction from the ResNet-50 model; positive for mild cognitive impairment with 58.94% confidence.</p>
Full article ">Figure 6
<p>Perturbed instances from the predicted image in <a href="#applsci-14-08287-f005" class="html-fig">Figure 5</a> showing deactivated pixels.</p>
Full article ">Figure 7
<p>Activated pixels displaying relevant features for the positive MCI prediction.</p>
Full article ">Figure 8
<p>Jet heatmap of positive values using self-attention for class-specific interpretability with gradient-weighted class activation mapping. The highest level of intensity in the heatmap is observed in close proximity to the hippocampus.</p>
Full article ">Figure 9
<p>Comparison between Grad-CAM without channel-wise attention (<b>Left</b>), which highlights a generalized region, and Grad-CAM with the attention mechanism (<b>Right</b>), which is more localized close to the hippocampal region.</p>
Full article ">
27 pages, 1592 KiB  
Review
A Glimpse into Humoral Response and Related Therapeutic Approaches of Takayasu’s Arteritis
by Shuning Guo, Yixiao Tian, Jing Li and Xiaofeng Zeng
Int. J. Mol. Sci. 2024, 25(12), 6528; https://doi.org/10.3390/ijms25126528 - 13 Jun 2024
Viewed by 948
Abstract
Takayasu’s arteritis (TAK) manifests as an insidiously progressive and debilitating form of granulomatous inflammation including the aorta and its major branches. The precise etiology of TAK remains elusive, with current understanding suggesting an autoimmune origin primarily driven by T cells. Notably, a growing [...] Read more.
Takayasu’s arteritis (TAK) manifests as an insidiously progressive and debilitating form of granulomatous inflammation including the aorta and its major branches. The precise etiology of TAK remains elusive, with current understanding suggesting an autoimmune origin primarily driven by T cells. Notably, a growing body of evidence bears testimony to the widespread effects of B cells on disease pathogenesis and progression. Distinct alterations in peripheral B cell subsets have been described in individuals with TAK. Advancements in technology have facilitated the identification of novel autoantibodies in TAK. Moreover, emerging data suggest that dysregulated signaling cascades downstream of B cell receptor families, including interactions with innate pattern recognition receptors such as toll-like receptors, as well as co-stimulatory molecules like CD40, CD80 and CD86, may result in the selection and proliferation of autoreactive B cell clones in TAK. Additionally, ectopic lymphoid neogenesis within the aortic wall of TAK patients exhibits functional characteristics. In recent decades, therapeutic interventions targeting B cells, notably utilizing the anti-CD20 monoclonal antibody rituximab, have demonstrated efficacy in TAK. Despite the importance of the humoral immune response, a systematic understanding of how autoreactive B cells contribute to the pathogenic process is still lacking. This review provides a comprehensive overview of the biological significance of B cell-mediated autoimmunity in TAK pathogenesis, as well as insights into therapeutic strategies targeting the humoral response. Furthermore, it examines the roles of T-helper and T follicular helper cells in humoral immunity and their potential contributions to disease mechanisms. We believe that further identification of the pathogenic role of autoimmune B cells and the underlying regulation system will lead to deeper personalized management of TAK patients. We believe that further elucidation of the pathogenic role of autoimmune B cells and the underlying regulatory mechanisms holds promise for the development of personalized approaches to managing TAK patients. Full article
Show Figures

Figure 1

Figure 1
<p>Two pathways of naïve B cells into antibody-secreting cells. In the follicular response (<b>left</b>), activated B cells engage in interactions with Th cells and follicle dendritic cells to form GC in secondary lymphoid organs. Following iterative rounds of somatic hypermutation and antigen affinity-driven selection, resting naïve B cells differentiate into antibody secreting cells or switched memory B cells derived from the germinal center. Extrafollicular responses (<b>right</b>) emerge preceding the formation of germinal centers, displaying distinctive phenotypic and transcriptional profiles compared to GC B cells. In healthy individuals, TLR7 and IFN-γ induce resting naïve B cells to differentiate into activated counterparts, DN2 cells and antibody-secreting cells in an IL-21-dependent manner. Neither pathway is T cell-dependent. In particular, the extrafollicular response includes a T cell-independent pathway. In addition, both pathways have mainly been reported in systemic lupus erythematosus. In TAK, the pathogenic role of extrafollicular responses is unknown. Therefore, we have marked a question mark on extrafollicular responses. Th: T helper; FDC: follicle dendritic cell; Mø: macrophage; DN2: double negative 2 cells; Ab: antibody; TLR7: toll-like receptor 7; IFNγ: interferon gamma; IL21: interleukin 21; TAK: Takayasu’s arteritis; GC: germinal center.</p>
Full article ">Figure 2
<p>A profile of artery involvement in TAK. In the left part, the color gradient illustrates the typical frequency of arterial segment involvement in TAK, with a predilection for the brachiocephalic arteries, as well as the thoracic and abdominal arterial territories. The right part shows the profile of the peripheral blood and vascular wall of TAK. The pathological process of TAK initiates in the vasa vasorum of the adventitia and is marked by the rupture of elastic laminae and smooth muscle cell migration. Several immune cells including memory B cells, antigen-experienced B cells as well as Tfh cells infiltrate the adventitia. The granulomas are located in the medial layer, and TLOs are distributed deeper within the adventitial layer which involves a dense network of HEVs. TLO: tertiary lymphoid organ; HEV: high endothelial venule; DC: dendritic cell; RBC: red blood cell; Tfh: T follicular helper; TAK: Takayasu’s arteritis.</p>
Full article ">Figure 3
<p>Abnormal activation of B cell checkpoints in TAK. The activation of BCRs, TLRs and several co-stimulatory molecules (including CD40, CD80 and CD86) was documented in TAK. Serum APRIL and BAFF levels and cytokines related to humoral immunity, including IL2, IL4, IL6, IL9, IL21, IL23 and IFN-γ, exhibited enhanced levels in TAK patients compared with healthy individuals. IL-5 induces B cell development and Ig secretion, the role of which is unclear in TAK. The bottom half of the figure is the cytokines and their receptors that are involved in B cell activation. The top half of the figure includes BCRs, TLRs and several co-stimulatory molecules. IL: interleukin; IFNγ: interferon-gamma; R: receptor; BAFF: B cell activating factor; BCMA: B cell maturation antigen; APRIL: A proliferation-inducing ligand; TACI: transmembrane activator and calcium modulator and cyclophilin ligand interactor; BCR: B cell receptor; TLR: toll-like receptor; Ig: immunoglobulin; gp130: glycoprotein 130; TAK: Takayasu’s arteritis; PAMP: pathogen-associated molecular pattern; DAMP: damage-associated molecular patterns.</p>
Full article ">
15 pages, 2580 KiB  
Article
Brain Network Modularity and Resilience Signaled by Betweenness Centrality Percolation Spiking
by Parker Kotlarz, Marcelo Febo, Juan C. Nino and on behalf of the Alzheimer’s Disease Neuroimaging Initiative
Appl. Sci. 2024, 14(10), 4197; https://doi.org/10.3390/app14104197 - 15 May 2024
Viewed by 1131
Abstract
Modularity and resilience are fundamental properties of brain network organization and function. The interplay of these network characteristics is integral to understanding brain vulnerability, network efficiency, and neurocognitive disorders. One potential methodology to explore brain network modularity and resilience is through percolation theory, [...] Read more.
Modularity and resilience are fundamental properties of brain network organization and function. The interplay of these network characteristics is integral to understanding brain vulnerability, network efficiency, and neurocognitive disorders. One potential methodology to explore brain network modularity and resilience is through percolation theory, a sub-branch of graph theory that simulates lesions across brain networks. In this work, percolation theory is applied to connectivity matrices derived from functional MRI from human, mice, and null networks. Nodes, or regions, with the highest betweenness centrality, a graph theory quantifier that examines shortest paths, were sequentially removed from the network. This attack methodology led to a rapid fracturing of the network, resulting in two terminal modules connected by one transfer module. Additionally, preceding the rapid network fracturing, the average betweenness centrality of the network peaked in value, indicating a critical point in brain network functionality. Thus, this work introduces a methodological perspective to identify hubs within the brain based on critical points that can be used as an architectural framework for a neural network. By applying percolation theory to functional brain networks through a network phase-transition lens, network sub-modules are identified using local spikes in betweenness centrality as an indicator of brain criticality. This modularity phase transition provides supporting evidence of the brain functioning at a near-critical point while showcasing a formalism to understand the computational efficiency of the brain as a neural network. Full article
(This article belongs to the Section Applied Neuroscience and Neural Engineering)
Show Figures

Figure 1

Figure 1
<p>Overview of connectomic pipeline from human neuroimaging to network generation. (1) The brain of each subject was extracted using FSL BET. (2) fMRI time series were then despiked, motion-corrected, and detrended. Nuisance signals were removed using ICA-based regression and the signal underwent low-pass filtering and spatial smoothing. (3) Preprocessed anatomical and fMRI scans were aligned to the MNI152 template (2 mm version). (4) Linear and nonlinear warping matrices for fMRI-to-anatomical alignment were applied to individual scans in the time series, then the merged 4D functional time series were moved to the atlas space using the prior anatomical-to-template transformation matrices. (5) Nodes were generated under the guidance of the Schaefer 300 functional parcellation [<a href="#B29-applsci-14-04197" class="html-bibr">29</a>]. (6) Signal time series were extracted from preprocessed fMRI scans with the assistance of ROI mask overlays. (7) The time-series files were used in cross-correlations and in calculations of Pearson r coefficients for every pairwise combination of ROIs (1dCorrelate in AFNI). The resulting matrices were (8) normalized and (9) thresholded across different graph density thresholds (5–30%) to remove spurious edges and negative correlations. Lastly, (10) network quantifiers were calculated, and percolation analysis was conducted.</p>
Full article ">Figure 2
<p>Individual-level subject percolation showing the average betweenness centrality spike coinciding with a significant drop in the largest cluster across different thresholds (5–30%). The gray shaded area represents the phase transition through the critical point. Plots with two gray shaded areas represent thresholds that have two potential phase transitions. The shaded area with the red asterisk is the phase transition shown in the brain inset. In the brain inset, the orange nodes show nodes part of the largest clusters while the red nodes in the after image are completely disconnected from the orange cluster. As seen throughout different thresholds, the regions identified from the phase transitions are relatively consistent, with one region located near the central gyrus and the subsequent region located in the anterior and posterior parts of the brain.</p>
Full article ">Figure 3
<p>(<b>A</b>) Representative connectivity matrix example illustrating key features shown by the following heatmaps from an individual subject. Nodes (x and y axes) are rearranged based on different modularity methods with the module denoted on the right-hand side; however, within-module node rearrangement is arbitrary. The matrices are colored by modules found using the phase transition modularity method, with black denoting no connections found at the node-to-node edge. Connections near the diagonal (light and navy blue) represent dense and sparse module connectivity, respectively. Off-diagonal clusters (green) represent between-module connections. Vis: visual; SomMot: somatomator; DorsAttn: dorsal attention; SalVent: salience/ventral attention; TempPar: temporoparietal. (<b>B</b>) Connectivity matrix organized by the Schaefer 300 node functional parcellation [<a href="#B29-applsci-14-04197" class="html-bibr">29</a>] used to create the original connectivity matrices. (<b>C</b>) Connectivity matrix organized by identifying features of the rich club: rich nodes, feeder nodes, and local nodes [<a href="#B49-applsci-14-04197" class="html-bibr">49</a>,<a href="#B50-applsci-14-04197" class="html-bibr">50</a>,<a href="#B51-applsci-14-04197" class="html-bibr">51</a>]. (<b>D</b>) Connectivity matrix organized by Newman’s modularity [<a href="#B3-applsci-14-04197" class="html-bibr">3</a>]. (<b>E</b>) Connectivity matrix organized by phase transition modularity. The large black squares demonstrate the reliance on the transfer module (yellow) to connect the terminal modules.</p>
Full article ">Figure 4
<p>(<b>A</b>) Visual representation of the modules identified using individual-level subject percolation. The red and orange nodes represent nodes part of their respective terminal module, while the yellow nodes represent nodes in the transfer module. (<b>B</b>) Neural network representation as a potential application of phase transition modularity. Transfer modules identified using phase transition modularity can act as hidden layers, while terminal layers function as either input or output layers.</p>
Full article ">
17 pages, 2973 KiB  
Article
Predicting the Conversion from Mild Cognitive Impairment to Alzheimer’s Disease Using an Explainable AI Approach
by Gerasimos Grammenos, Aristidis G. Vrahatis, Panagiotis Vlamos, Dean Palejev, Themis Exarchos and for the Alzheimer’s Disease Neuroimaging Initiative
Information 2024, 15(5), 249; https://doi.org/10.3390/info15050249 - 28 Apr 2024
Viewed by 1403
Abstract
Mild Cognitive Impairment (MCI) is a cognitive state frequently observed in older adults, characterized by significant alterations in memory, thinking, and reasoning abilities that extend beyond typical cognitive decline. It is worth noting that around 10–15% of individuals with MCI are projected to [...] Read more.
Mild Cognitive Impairment (MCI) is a cognitive state frequently observed in older adults, characterized by significant alterations in memory, thinking, and reasoning abilities that extend beyond typical cognitive decline. It is worth noting that around 10–15% of individuals with MCI are projected to develop Alzheimer’s disease, effectively positioning MCI as an early stage of Alzheimer’s. In this study, a novel approach is presented involving the utilization of eXtreme Gradient Boosting to predict the onset of Alzheimer’s disease during the MCI stage. The methodology entails utilizing data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Through the analysis of longitudinal data, spanning from the baseline visit to the 12-month follow-up, a predictive model was constructed. The proposed model calculates, over a 36-month period, the likelihood of progression from MCI to Alzheimer’s disease, achieving an accuracy rate of 85%. To further enhance the precision of the model, this study implements feature selection using the Recursive Feature Elimination technique. Additionally, the Shapley method is employed to provide insights into the model’s decision-making process, thereby augmenting the transparency and interpretability of the predictions. Full article
Show Figures

Figure 1

Figure 1
<p>Demographics summary.</p>
Full article ">Figure 2
<p>Sankey Diagram featuring patient’s clinical state from the baseline until the 48th-month visit.</p>
Full article ">Figure 3
<p>Missing values at baseline visit.</p>
Full article ">Figure 4
<p>Prediction method using baseline and first annual visit data.</p>
Full article ">Figure 5
<p>Training workflow used for creating the model.</p>
Full article ">Figure 6
<p>Recursive Feature Elimination workflow as implemented in scikit-learn.</p>
Full article ">Figure 7
<p>Summarized confusion matrix from 5-fold cross-validation.</p>
Full article ">Figure 8
<p>SHAP summary plot.</p>
Full article ">Figure 9
<p>SHAP feature importance plot.</p>
Full article ">
12 pages, 546 KiB  
Article
Identifying Progression-Specific Alzheimer’s Subtypes Using Multimodal Transformer
by Diego Machado Reyes, Hanqing Chao, Juergen Hahn, Li Shen, Pingkun Yan and for the Alzheimer’s Disease Neuroimaging Initiative
J. Pers. Med. 2024, 14(4), 421; https://doi.org/10.3390/jpm14040421 - 15 Apr 2024
Viewed by 1282
Abstract
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disease, yet its current treatments are limited to stopping disease progression. Moreover, the effectiveness of these treatments remains uncertain due to the heterogeneity of the disease. Therefore, it is essential to identify disease subtypes at [...] Read more.
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disease, yet its current treatments are limited to stopping disease progression. Moreover, the effectiveness of these treatments remains uncertain due to the heterogeneity of the disease. Therefore, it is essential to identify disease subtypes at a very early stage. Current data-driven approaches can be used to classify subtypes during later stages of AD or related disorders, but making predictions in the asymptomatic or prodromal stage is challenging. Furthermore, the classifications of most existing models lack explainability, and these models rely solely on a single modality for assessment, limiting the scope of their analysis. Thus, we propose a multimodal framework that utilizes early-stage indicators, including imaging, genetics, and clinical assessments, to classify AD patients into progression-specific subtypes at an early stage. In our framework, we introduce a tri-modal co-attention mechanism (Tri-COAT) to explicitly capture cross-modal feature associations. Data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) (slow progressing = 177, intermediate = 302, and fast = 15) were used to train and evaluate Tri-COAT using a 10-fold stratified cross-testing approach. Our proposed model outperforms baseline models and sheds light on essential associations across multimodal features supported by known biological mechanisms. The multimodal design behind Tri-COAT allows it to achieve the highest classification area under the receiver operating characteristic curve while simultaneously providing interpretability to the model predictions through the co-attention mechanism. Full article
(This article belongs to the Section Mechanisms of Diseases)
Show Figures

Figure 1

Figure 1
<p>The three main multimodal fusion strategies, early, intermediate, and late fusion, for deep learning methods.</p>
Full article ">Figure 2
<p>Illustration of the proposed framework for AD subtyping consisting of two main sections: single-modality encoding and tri-modal attention with joint encoding.</p>
Full article ">Figure 3
<p>AD progression-specific subtype clusters based on a decrease in the MMSE at each visit. (<b>a</b>) Each line represents the average score across patients for each cluster, and the shadow represents one standard deviation. (<b>b</b>) Individual lines per patient are plotted.</p>
Full article ">Figure 4
<p>Cross-modal associations of key AD biomarkers visualized from the learned co-attention.</p>
Full article ">
14 pages, 387 KiB  
Article
Imputation-Based Variable Selection Method for Block-Wise Missing Data When Integrating Multiple Longitudinal Studies
by Zhongzhe Ouyang, Lu Wang and Alzheimer’s Disease Neuroimaging Initiative
Mathematics 2024, 12(7), 951; https://doi.org/10.3390/math12070951 - 23 Mar 2024
Viewed by 842
Abstract
When integrating data from multiple sources, a common challenge is block-wise missing. Most existing methods address this issue only in cross-sectional studies. In this paper, we propose a method for variable selection when combining datasets from multiple sources in longitudinal studies. To account [...] Read more.
When integrating data from multiple sources, a common challenge is block-wise missing. Most existing methods address this issue only in cross-sectional studies. In this paper, we propose a method for variable selection when combining datasets from multiple sources in longitudinal studies. To account for block-wise missing in covariates, we impute the missing values multiple times based on combinations of samples from different missing pattern and predictors from different data sources. We then use these imputed data to construct estimating equations, and aggregate the information across subjects and sources with the generalized method of moments. We employ the smoothly clipped absolute deviation penalty in variable selection and use the extended Bayesian Information Criterion criteria for tuning parameter selection. We establish the asymptotic properties of the proposed estimator, and demonstrate the superior performance of the proposed method through numerical experiments. Furthermore, we apply the proposed method in the Alzheimer’s Disease Neuroimaging Initiative study to identify sensitive early-stage biomarkers of Alzheimer’s Disease, which is crucial for early disease detection and personalized treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Example of block-wise missing data in longitudinal studies.</p>
Full article ">Figure 2
<p>Two imputation approaches for missing covariates of source 3 in pattern 2. In the left figure, samples from pattern 1 and covariates in source 1 and source 2 are used to train the model, which is subsequently used to predict the missing covariates in pattern 2. Similarly, in the right figure, samples from pattern 1 and pattern 3 and covariates in source 1 are used to train the model.</p>
Full article ">
10 pages, 599 KiB  
Brief Report
The Association among Hypothalamic Subnits, Gonadotropic and Sex Hormone Plasmas Levels in Alzheimer’s Disease
by Edward Ofori, Anamaria Solis, Nahid Punjani and on behalf of the Alzheimer’s Disease Neuroimaging Initiative
Brain Sci. 2024, 14(3), 276; https://doi.org/10.3390/brainsci14030276 - 14 Mar 2024
Viewed by 1194
Abstract
This study investigates the sex-specific role of the Hypothalamic–Pituitary–Gonadal axis in Alzheimer’s disease progression, utilizing ADNI1 data for 493 individuals, analyzing plasma levels of gonadotropic and sex hormones, and examining neurodegeneration-related brain structures. We assessed plasma levels of follicle stimulating hormone (FSH), luteinizing [...] Read more.
This study investigates the sex-specific role of the Hypothalamic–Pituitary–Gonadal axis in Alzheimer’s disease progression, utilizing ADNI1 data for 493 individuals, analyzing plasma levels of gonadotropic and sex hormones, and examining neurodegeneration-related brain structures. We assessed plasma levels of follicle stimulating hormone (FSH), luteinizing hormone (LH), progesterone (P4), and testosterone (T), along with volumetric measures of the hippocampus, entorhinal cortex, and hypothalamic subunits, to explore their correlation with Alzheimer’s disease markers across different cognitive statuses and sexes. Significant cognitive status effects were observed for all volumetric measures, with a distinct sex-by-cognitive status interaction for hypothalamic volume, indicating a decrease in males but not in females across cognitive impairment stages. Regression analyses showed specific hypothalamic subunit volume related to hormone levels, accounting for up to approximately 40% of the variance (p < 0.05). The findings highlight sex differences in neurodegeneration and hormonal regulation, suggesting potential for personalized treatments and advancing the understanding of Alzheimer’s disease etiology. Full article
(This article belongs to the Section Neurodegenerative Diseases)
Show Figures

Figure 1

Figure 1
<p>Box plots of hypothalamic (<b>A</b>) and entorhinal volume (<b>B</b>) across cognitive status on the x-axis. Green boxes are male plots and orange boxes are female plots. Numerical values in box plots are the % of intracranial volume reflective of the unique condition. A red x indicates a male (M) point and a blue x indicates an individual female (F) point. The asterisk indicates the significance at <span class="html-italic">p</span> &lt; 0.05 of the sex-by-cognitive status interaction.</p>
Full article ">
20 pages, 2660 KiB  
Article
Sex Differences in Conversion Risk from Mild Cognitive Impairment to Alzheimer’s Disease: An Explainable Machine Learning Study with Random Survival Forests and SHAP
by Alessia Sarica, Assunta Pelagi, Federica Aracri, Fulvia Arcuri, Aldo Quattrone, Andrea Quattrone and for the Alzheimer’s Disease Neuroimaging Initiative
Brain Sci. 2024, 14(3), 201; https://doi.org/10.3390/brainsci14030201 - 22 Feb 2024
Cited by 1 | Viewed by 1311
Abstract
Alzheimer’s disease (AD) exhibits sex-linked variations, with women having a higher prevalence, and little is known about the sexual dimorphism in progressing from Mild Cognitive Impairment (MCI) to AD. The main aim of our study was to shed light on the sex-specific conversion-to-AD [...] Read more.
Alzheimer’s disease (AD) exhibits sex-linked variations, with women having a higher prevalence, and little is known about the sexual dimorphism in progressing from Mild Cognitive Impairment (MCI) to AD. The main aim of our study was to shed light on the sex-specific conversion-to-AD risk factors using Random Survival Forests (RSF), a Machine Learning survival approach, and Shapley Additive Explanations (SHAP) on dementia biomarkers in stable (sMCI) and progressive (pMCI) patients. With this purpose, we built two separate models for male (M-RSF) and female (F-RSF) cohorts to assess whether global explanations differ between the sexes. Similarly, SHAP local explanations were obtained to investigate changes across sexes in feature contributions to individual risk predictions. The M-RSF achieved higher performance on the test set (0.87) than the F-RSF (0.79), and global explanations of male and female models had limited similarity (<71.1%). Common influential variables across the sexes included brain glucose metabolism and CSF biomarkers. Conversely, the M-RSF had a notable contribution from hippocampus, which had a lower impact on the F-RSF, while verbal memory and executive function were key contributors only in F-RSF. Our findings confirmed that females had a higher risk of progressing to dementia; moreover, we highlighted distinct sex-driven patterns of variable importance, uncovering different feature contribution risks across sexes that decrease/increase the conversion-to-AD risk. Full article
(This article belongs to the Special Issue Neuroscience Meets Artificial Intelligence)
Show Figures

Figure 1

Figure 1
<p>Performance per timepoint on the test set by RSF trained on (<b>a</b>) male MCI patients; (<b>b</b>) female MCI patients. Upper: plot over time of expected number of MCI patients at risk of conversion to AD, estimated survival curve by Kaplan-Meier in gray. Bottom: Integrated Brier error curve (IBS, critical cut-off limit of 0.25 in red). <span class="html-italic">C</span>-index on the test set, cross-validated (<span class="html-italic">cv</span>) <span class="html-italic">c</span>-index on the training set (mean ± standard deviation), Root Mean Square Error (RMSE), and median and mean absolute error are also reported.</p>
Full article ">Figure 2
<p>Random Survival Forests (RSF) global explanations trained on (<b>a</b>) male MCI (M-RSF) and (<b>b</b>) female MCI (F-RSF) patients. From left to right: RSF feature importance (VIMP), permutation importance (mean value), and SHAP importance (mean |SHAP| value). (<b>c</b>) Rank-Biased Overlap (RBO) curves assessing the overlap between male and female (M vs. F) variable rankings at different numbers of the important features considered (depth <span class="html-italic">d</span>): RSF feature importance (in plum), mean permutation importance (in violet), and mean |SHAP| importance (in purple).</p>
Full article ">Figure 3
<p>SHAP bar plots of (<b>a</b>) male MCI (M-RSF) and (<b>b</b>) female MCI test patients (F-RSF).</p>
Full article ">Figure 4
<p>Random Survival Forests (RSF) local explanations trained on male MCI patients. (<b>a</b>) Histograms of male sMCI and pMCI patients’ risk distribution predicted by M-RSF. Patients were stratified by risk grade: low (in green, range 1.39–2), medium (in orange, range 2–2.6), high (in red, range 2.6–3.47). (<b>b</b>) RSF survival functions of male pMCI patients per risk score: M-pMCI#1 high risk (score 3.262, converted to AD after 12 months), M-pMCI#2 medium risk (score 1.962, converted to AD after 24 months), M-pMCI#3 low risk (score 1.395, converted to AD after 36 months). SHAP waterfall plot of (<b>c</b>) patient M-pMCI#1, (<b>d</b>) patient M-pMCI#2, (<b>e</b>) patient M-pMCI#3, and (<b>f</b>) stable MCI patient who does not convert to AD within 36 months (M-sMCI, risk score 0.459). Features that decrease the risk are in blue, while those that increase it are in red. Average predicted risk <span class="html-italic">E</span>[<span class="html-italic">f</span>(<span class="html-italic">x</span>)] = 1.769. Actual value of the feature is in gray.</p>
Full article ">Figure 5
<p>Random Survival Forests (RSF) local explanations trained on female MCI patients. (<b>a</b>) Histograms of female sMCI and pMCI patients’ risk distribution predicted by F-RSF. Patients were stratified by risk grade: low (in green, range 1.51–2.3), medium (in orange, range 2.3–3.7), high (in red, range 3.7–5.05). (<b>b</b>) RSF survival functions of female pMCI patients per risk score: F-pMCI#1 high risk (score 4.683, converted to AD after 6 months), F-pMCI#2 medium risk (score 2.799, converted to AD after 12 months), F-pMCI#3 low risk (score 1.51, converted to AD after 24 months). SHAP waterfall plot of (<b>c</b>) patient F-pMCI#1, (<b>d</b>) patient F-pMCI#2, (<b>e</b>) patient F-pMCI#3, and (<b>f</b>) stable MCI patient who does not convert to AD within 36 months (F-sMCI, risk score 0.528). Features that decrease the risk are in blue, while those that increase it are in red. Average predicted risk <span class="html-italic">E</span>[<span class="html-italic">f</span>(<span class="html-italic">x</span>)] = 2.534. Actual value of the feature is in gray.</p>
Full article ">
21 pages, 4449 KiB  
Article
CD163-Mediated Small-Vessel Injury in Alzheimer’s Disease: An Exploration from Neuroimaging to Transcriptomics
by Yuewei Chen, Peiwen Lu, Shengju Wu, Jie Yang, Wanwan Liu, Zhijun Zhang and Qun Xu
Int. J. Mol. Sci. 2024, 25(4), 2293; https://doi.org/10.3390/ijms25042293 - 14 Feb 2024
Cited by 1 | Viewed by 1650
Abstract
Patients with Alzheimer’s disease (AD) often present with imaging features indicative of small-vessel injury, among which, white-matter hyperintensities (WMHs) are the most prevalent. However, the underlying mechanism of the association between AD and small-vessel injury is still obscure. The aim of this study [...] Read more.
Patients with Alzheimer’s disease (AD) often present with imaging features indicative of small-vessel injury, among which, white-matter hyperintensities (WMHs) are the most prevalent. However, the underlying mechanism of the association between AD and small-vessel injury is still obscure. The aim of this study is to investigate the mechanism of small-vessel injury in AD. Differential gene expression analyses were conducted to identify the genes related to WMHs separately in mild cognitive impairment (MCI) and cognitively normal (CN) subjects from the ADNI database. The WMH-related genes identified in patients with MCI were considered to be associated with small-vessel injury in early AD. Functional enrichment analyses and a protein–protein interaction (PPI) network were performed to explore the pathway and hub genes related to the mechanism of small-vessel injury in MCI. Subsequently, the Boruta algorithm and support vector machine recursive feature elimination (SVM-RFE) algorithm were performed to identify feature-selection genes. Finally, the mechanism of small-vessel injury was analyzed in MCI from the immunological perspectives; the relationship of feature-selection genes with various immune cells and neuroimaging indices were also explored. Furthermore, 5×FAD mice were used to demonstrate the genes related to small-vessel injury. The results of the logistic regression analyses suggested that WMHs significantly contributed to MCI, the early stage of AD. A total of 276 genes were determined as WMH-related genes in patients with MCI, while 203 WMH-related genes were obtained in CN patients. Among them, only 15 genes overlapped and were thus identified as the crosstalk genes. By employing the Boruta and SVM-RFE algorithms, CD163, ALDH3B1, MIR22HG, DTX2, FOLR2, ALDH2, and ZNF23 were recognized as the feature-selection genes linked to small-vessel injury in MCI. After considering the results from the PPI network, CD163 was finally determined as the critical WMH-related gene in MCI. The expression of CD163 was correlated with fractional anisotropy (FA) values in regions that are vulnerable to small-vessel injury in AD. The immunostaining and RT-qPCR results from the verifying experiments demonstrated that the indicators of small-vessel injury presented in the cortical tissue of 5×FAD mice and related to the upregulation of CD163 expression. CD163 may be the most pivotal candidates related to small-vessel injury in early AD. Full article
Show Figures

Figure 1

Figure 1
<p>The study workflow chart. Abbreviations: WMH, white-matter hyperintensities; DTI_ROI, diffusion tensor imaging region of interest; CN, cognitively normal; MCI, mild cognitive impairment; AD, Alzheimer’s disease; ln_WMH_TCV, natural logarithm of standardized white-matter hyperintensities (WMH) to total cerebrum cranial volume (TCV); MCI_WMH+, mild cognitive impairment with severe white-matter hyperintensities, whose values of ‘ ln_WMH_TCV’ were above the median; MCI_WMH–, mild cognitive impairment with no or mild white-matter hyperintensities, whose values of ‘ ln_WMH_TCV’ were below the median.</p>
Full article ">Figure 2
<p>The difference among CN, MCI, and dementia in DTI indices and the prediction accuracy of ln_WMH_TCV and TCB_TCV in predicting AD progression: (<b>A</b>) The intersection of difference in FA, MD, RD, and AxD indices were considered the most fragile fiber bundles; (<b>B</b>) The predictive effectiveness of cerebral atrophy and small-vessel injury at different stages of AD diagnosis; (<b>C</b>) different cross-sectional views of the fragile fiber bundles. Red: bilateral splenium of the corpus callosum (SUMSCC, SCC_R, SCC_L), blue: bilateral fornix (SUMFX), violet: posterior thalamic radiation_left (PTR_L), yellow: fornix (cres)/stria terminalis_left (FX_ST_L, FX_L), cyan: tapetum (TAP_R, TAP_L). Abbreviations: FA, fractional anisotropy; MD, mean diffusivity; RD, radial diffusivity; AxD, axial diffusivity, TAP_R, tapetum right; TAP_L, tapetum left; SUMSCC, bilateral splenium of the corpus callosum; SUMFX, bilateral fornix; SCC_R, splenium of corpus callosum right; SCC_L, splenium of corpus callosum left; PTR_L, posterior thalamic radiation left; FX_ST_L, fornix (cres)/stria terminalis left; FX_L, fornix left; CN, cognitively normal; MCI, mild cognitive impairment; AD, Alzheimer’s disease; ROC, receiver operating characteristic; AUC, area under the curve; TCB_TCV, standardized total cerebrum brain volume (TCB) to total cerebrum cranial volume (TCV); ln_WMH_TCV, natural logarithm of standardized white-matter hyperintensities (WMH) to total cerebrum cranial volume (TCV).</p>
Full article ">Figure 3
<p>Identification of DEGs and functional enrichment analysis: (<b>A</b>) Volcano plot of DEGs constructed using the fold-change values (0.12) and <span class="html-italic">p</span>-value (0.05); red-orange color dots represent genes upregulated in MCI_WMH+, gray dots represent genes not differing significantly between MCI_WMH+ and MCI_WMH–, and cyan dots represent genes downregulated in MCI_WMH+. (<b>B</b>) Volcano plot of DEGs constructed using the fold-change values (0.12) and <span class="html-italic">p</span>-value (0.05); red-orange color dots represent genes upregulated in CN_WMH+ group, gray dots represent genes not differing significantly between CN_WMH+ and CN_WMH– groups, and cyan dots represent genes downregulated in CN_WMH+ group. (<b>C</b>) Venn plot. Only fifteen genes shared between WMH-related genes in CN patients and those with MCI. (<b>D</b>,<b>E</b>) The results of GSEA analysis of KEGG in MCI_WMH+ samples. Abbreviations: MCI_WMH+, mild cognitive impairment with severe white-matter hyperintensities; MCI_WMH–, mild cognitive impairment with no or mild white-matter hyperintensities; CN_WMH+, cognitively normal with severe white-matter hyperintensities; CN_WMH–, cognitively normal with no or mild white-matter hyperintensities; KEGG_GSEA, gene set enrichment analysis (GSEA) of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways; DEGs, differentially expressed genes.</p>
Full article ">Figure 4
<p>Screening of closely related genes and hub genes of WMH-related genes in MCI group using cytoHubba and MCODE plugins: (<b>A</b>) Macroscopic display of PPI networks for all DEGs of WMH-related genes in MCI group, with a redder color indicating a higher degree score; the gene nodes in the topological characteristics of this PPI network were ranked in descending order of degree value (<b>B</b>–<b>D</b>), with a deeper purple color indicating a higher score. The top three modules’ genes are filtered by MCODE; (<b>E</b>) A redder color indicates a higher score and a yellower color indicates a lower score. The hub genes are filtered by the MCC for the top 30 genes; (<b>F</b>) Venn plot the common genes are both filtered by MCODE and MCC. Abbreviations: MCODE, molecular complex detection; MCC, maximal clique centrality.</p>
Full article ">Figure 5
<p>Screening feature-selection genes from DEGs between mild and severe small-vessel injury in MCI group: (<b>A</b>) 15 feature-selection genes were screened by Boruta algorithm; (<b>B</b>) 22 feature-selected genes were screened by SVM-RFE algorithm; (<b>C</b>) Venn plot, seven variables including CD163, FOLR2, ALDH3B1, DTX2, ALDH2, ZNF23, and MIR22HG intersected by Boruta and SVM-RFE algorithms; (<b>D</b>) the ROC curve of seven machine learning models, and the AUC value represents the model predictive effectiveness in testing set; (<b>E</b>) the expression level of feature-selection genes CD163, FOLR2, ALDH3B1, DTX2, ALDH2, ZNF23, and MIR22HG in the MCI between mild and severe small-vessel injury. CD163, <span class="html-italic">p</span>-value = 1.2 × 10<sup>−4</sup>; FOLR2, <span class="html-italic">p</span>-value = 6.4 × 10<sup>−4</sup>; ALDH3B1, <span class="html-italic">p</span>-value = 4.7 × 10<sup>−4</sup>; DTX2, <span class="html-italic">p</span>-value = 1.8 × 10<sup>−3</sup>; ALDH2, <span class="html-italic">p</span>-value = 3 × 10<sup>−3</sup>; ZNF23, <span class="html-italic">p</span>-value = 5.1 × 10<sup>−4</sup>; MIR22HG, <span class="html-italic">p</span>-value = 8.6 × 10<sup>−5</sup>. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001. Abbreviations: SVM-RFE, support vector machine recursive feature elimination; RMSE, root mean square error; AUC, area under the curve; gbm, gradient boosting machine; rf, random forest; KNN, k-nearest neighbors; SVM, support vector machine; GLM, generalized linear model; XGboost, extreme gradient boosting; rpart, recursive partition tree.</p>
Full article ">Figure 6
<p>Alterations in the abundance of immune cells in MCI group with severe small-vessel injury (MCI_WMH+) and correlation analysis between hub genes with the abundance of immune cell and FA values: (<b>A</b>) Estimated proportions of 28 immune cell types between two groups in MCI group; (<b>B</b>) Correlation analysis of hub genes with different immune cell types; (<b>C</b>) Correlation analysis of hub genes with differential brain-area FA values; (<b>D</b>) Venn plot, two genes (CD163 and FOLR2) intersected by Feature-selection algorithms and PPI. (<b>E</b>) The correlation between CD163, FOLR2, and ln_WMH_TCV. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. Abbreviations: FA, fractional anisotropy; SUMFX, bilateral fornix; TAP_R, tapetum right; SCC_L, splenium of corpus callosum left; SCC_R, splenium of corpus callosum right; SUMSCC, bilateral splenium of the corpus callosum; PTR_L, posterior thalamic radiation left; FX_ST_L, fornix (cres)/stria terminalis left; FX_L, fornix left; TAP_L, tapetum left; ln_WMH_TCV, natural logarithm of standardized white-matter hyperintensities (WMH) to total cerebrum cranial volume (TCV).</p>
Full article ">Figure 7
<p>AD pathology leads to small-vessel injury and elevates CD163 expression. Immunofluorescence staining results of (<b>A</b>) 5×FAD cerebral cortex (left scale bar = 25 um; right scale bar = 10 um) and (<b>B</b>) WT cerebral cortex, green: CD31; red: AQP4; magenta: Aβ; blue: DAPI. (<b>C</b>–<b>E</b>) Calculation of the width of the perivascular spaces and the comparation between WT and 5×FAD. (<b>F</b>) rt-qPCR results from cortical tissues exhibited an upregulation of CD163 expression in 5×FAD mice. (<b>G</b>) Immunofluorescence staining: control vs. Aβ-treated rat primary microglia (scale bar = 50 um), green: CD163; red: iba-1; blue: DAPI. (<b>H</b>) Immunofluorescence staining results of CD163 showing the MFI and the percentage of cells in different MFIs. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001. Abbreviations: MFI, mean fluorescence intensity; Aβ, amyloid beta.</p>
Full article ">
16 pages, 1592 KiB  
Article
Predicting Conversion from Mild Cognitive Impairment to Alzheimer’s Disease Using K-Means Clustering on MRI Data
by Miranda Bellezza, Azzurra di Palma and Andrea Frosini
Information 2024, 15(2), 96; https://doi.org/10.3390/info15020096 - 8 Feb 2024
Viewed by 1437
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that leads to the loss of cognitive functions due to the deterioration of brain tissue. Current diagnostic methods are often invasive or costly, limiting their widespread use. Developing non-invasive and cost-effective screening methods is [...] Read more.
Alzheimer’s disease (AD) is a neurodegenerative disorder that leads to the loss of cognitive functions due to the deterioration of brain tissue. Current diagnostic methods are often invasive or costly, limiting their widespread use. Developing non-invasive and cost-effective screening methods is crucial, especially for identifying patients with mild cognitive impairment (MCI) at the risk of developing Alzheimer’s disease. This study employs a Machine Learning (ML) approach, specifically K-means clustering, on a subset of pixels common to all magnetic resonance imaging (MRI) images to rapidly classify subjects with AD and those with normal Normal Cognitive (NC). In particular, we benefited from defining significant pixels, a narrow subset of points (in the range of 1.5% to 6% of the total) common to all MRI images and related to more intense degeneration of white or gray matter. We performed K-means clustering, with k = 2, on the significant pixels of AD and NC MRI images to separate subjects belonging to the two classes and detect the class centroids. Subsequently, we classified subjects with MCI using only the significant pixels. This approach enables quick classification of subjects with AD and NC, and more importantly, it predicts MCI-to-AD conversion with high accuracy and low computational cost, making it a rapid and effective diagnostic tool for real-time assessments. Full article
(This article belongs to the Section Information Applications)
Show Figures

Figure 1

Figure 1
<p>Block diagram of the study design. The data collected from the <span class="html-italic">ADNI</span> database first undergoes a pre-processing stage where white and gray matter are segmented. Then, a permutation test on the white and gray matter of <math display="inline"><semantics> <mrow> <mi>N</mi> <mi>C</mi> </mrow> </semantics></math> and <math display="inline"><semantics> <mrow> <mi>A</mi> <mi>D</mi> </mrow> </semantics></math> subjects allows significant pixels to be detected and to restrict the dataset accordingly. The involved classes are defined in <a href="#sec3-information-15-00096" class="html-sec">Section 3</a> and <a href="#sec3dot2-information-15-00096" class="html-sec">Section 3.2</a>. Finally, a ML model, <span class="html-italic">K</span>-means, is trained, tested, and employed to distinguish between normal aging and <span class="html-italic">AD</span> degeneration, as well as predict candidates exhibiting the <span class="html-italic">MCI</span>-to-<span class="html-italic">AD</span> pattern.</p>
Full article ">Figure 2
<p>The axial slice 58 view of gray matter, white matter, and fluid (from left to right) extracted from MRI data of an <span class="html-italic">AD</span> subject. The pixel classification is performed with the Matlab CONN toolbox.</p>
Full article ">Figure 3
<p>From left to right, the axial slice 58 view of gray matter at <math display="inline"><semantics> <msub> <mi>t</mi> <mn>0</mn> </msub> </semantics></math>, <math display="inline"><semantics> <msub> <mi>t</mi> <mn>2</mn> </msub> </semantics></math> time acquisitions and their difference, highlighted in red. The data are from the MRI of an <span class="html-italic">AD</span> subject.</p>
Full article ">Figure 4
<p>Significant pixels’ distribution for the white and gray matter according to the three different <math display="inline"><semantics> <mi>α</mi> </semantics></math> thresholds.</p>
Full article ">Figure 5
<p>The confusion matrices computed after the <span class="html-italic">K</span>-means clustering of the <math display="inline"><semantics> <msup> <mover accent="true"> <mrow> <mi>A</mi> <mi>D</mi> </mrow> <mo>^</mo> </mover> <mi>g</mi> </msup> </semantics></math> and <math display="inline"><semantics> <msup> <mover accent="true"> <mrow> <mi>N</mi> <mi>C</mi> </mrow> <mo>^</mo> </mover> <mi>g</mi> </msup> </semantics></math> classes with respect to the four (Euclidean, Czekanowski, Chebyshev, and City Block) distances (the first one outperforming the others). The distances led to convergence after 2, 8, 10, and 6 iterations, respectively.</p>
Full article ">Figure 6
<p>Subject-to-centroid distance distributions inside each cluster.</p>
Full article ">Figure 7
<p>Slice 58 of an <math display="inline"><semantics> <mrow> <mi>A</mi> <mi>D</mi> </mrow> </semantics></math> subject. (<b>Left</b>) white and gray matter locations. (<b>Right</b>) the significant pixels of the same slice and the brain areas in which they are located, divided according to Brodmann areas. The vast majority of significant pixels are located in the fusiform gyros, hyppocampus, amigdala, parahippocampal gyrus, and orbitofrontal lobe.</p>
Full article ">
14 pages, 2836 KiB  
Article
Uptake of 18F-AV45 in the Putamen Provides Additional Insights into Alzheimer’s Disease beyond the Cortex
by Zhengshi Yang, Jefferson W. Kinney, Dietmar Cordes and The Alzheimer’s Disease Neuroimaging Initiative
Biomolecules 2024, 14(2), 157; https://doi.org/10.3390/biom14020157 - 29 Jan 2024
Cited by 1 | Viewed by 1403
Abstract
Cortical uptake in brain amyloid positron emission tomography (PET) is increasingly used for the biological diagnosis of Alzheimer’s disease (AD); however, the clinical and biological relevance of the striatum beyond the cortex in amyloid PET scans remains unclear. A total of 513 amyloid-positive [...] Read more.
Cortical uptake in brain amyloid positron emission tomography (PET) is increasingly used for the biological diagnosis of Alzheimer’s disease (AD); however, the clinical and biological relevance of the striatum beyond the cortex in amyloid PET scans remains unclear. A total of 513 amyloid-positive participants having 18F-AV45 amyloid PET scans available were included in the analysis. The associations between cognitive scores and striatal uptake were analyzed. The participants were categorized into three groups based on the residual from the linear fitting between 18F-AV45 uptake in the putamen and the cortex in the order of HighP > MidP > LowP group. We then examined the differences between these three groups in terms of clinical diagnosis, APOE genotype, CSF phosphorylated tau (ptau) concentration, hippocampal volume, entorhinal thickness, and cognitive decline rate to evaluate the additional insights provided by the putamen beyond the cortex. The 18F-AV45 uptake in the putamen was more strongly associated with ADAS-cog13 and MoCA scores (p < 0.001) compared to the uptake in the caudate nucleus. Despite comparable cortical uptakes, the HighP group had a two-fold higher risk of being ε4-homozygous or diagnosed with AD dementia compared to the LowP group. These three groups had significantly different CSF ptau concentration, hippocampal volume, entorhinal thickness, and cognitive decline rate. These findings suggest that the assessment of 18F-AV45 uptake in the putamen is of unique value for evaluating disease severity and predicting disease progression. Full article
(This article belongs to the Special Issue The Role of Amyloid in Neurological Disorders)
Show Figures

Figure 1

Figure 1
<p>Violin plots of original SUVRs in the cortex (<b>a</b>), putamen (<b>b</b>), and caudate nucleus (<b>c</b>). A comparison between CN- and all amyloid-positive participants (including clinical diagnosis as CN, MCI, or dementia) was conducted with the Cohen’s d marked in the figure.</p>
Full article ">Figure 2
<p>Association analysis between striatal SUVRs and cognitive scores/age. ADAS-cog13 and MoCA were used for cognitive assessment. All amyloid-positive participants, including CN (gray dot), MCI (blue diamond), and dementia (red triangle), were used for analysis. The linear fitting curves with 95% confidence interval (gray areas), together with scatter plots, were shown in the figure. The slopes β from the linear fitting and the corresponding significance level are marked in the figure. The putamen had stronger association with cognitive scores, and the caudate nucleus had stronger association with age (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Participant categorization based on the association of SUVRs in the cortex and putamen. (<b>a</b>) Scatter plot between cortical SUVR and putamen SUVR. The linear fitting curve (solid blue line) is also shown in the figure. Based on the standardized residual (<span class="html-italic">r<sub>std</sub></span>) from the linear fitting, the participants were grouped into LowP (<span class="html-italic">r<sub>std</sub></span> ≤ −0.5; blue), MidP (|<span class="html-italic">r<sub>std</sub></span>| &lt; 0.5; gray), or HighP (<span class="html-italic">r<sub>std</sub></span> ≥ 0.5; red) groups along the direction (black arrow) perpendicular to the fitting curve. (<b>b</b>) Violin plots of original cortical SUVR in LowP, MidP, and HighP groups. There was no difference in cortical SUVR between these three groups (<span class="html-italic">p</span> &gt; 0.05). (<b>c</b>) Violin plots of the residual from the linear fitting. The dashed blue lines in (<b>a</b>,<b>c</b>) indicate <span class="html-italic">r<sub>std</sub></span> = ±0.5.</p>
Full article ">Figure 4
<p>Proportions of clinical diagnosis (<b>top panel</b>) and <span class="html-italic">APOE</span> genotype (<b>bottom panel</b>) in LowP, MidP, and HighP groups. Clinical diagnosis and <span class="html-italic">APOE</span> genotypes are significantly different in LowP, MidP, and HighP groups.</p>
Full article ">Figure 5
<p>Group comparisons of hippocampal volume (<b>a</b>), entorhinal thickness (<b>b</b>), and CSF ptau concentration (<b>c</b>) between LowP, MidP, and HighP groups. One-way ANOVA found that these measures were significantly different between three groups. The horizontal line in (<b>c</b>) indicates the cut-off concentration for tau positivity. The Cohen’s d values for the pairwise comparisons having significant differences were marked in the figure. * <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 1 × 10<sup>−5</sup>.</p>
Full article ">
16 pages, 6078 KiB  
Article
Explainable Machine Learning with Pairwise Interactions for Predicting Conversion from Mild Cognitive Impairment to Alzheimer’s Disease Utilizing Multi-Modalities Data
by Jiaxin Cai, Weiwei Hu, Jiaojiao Ma, Aima Si, Shiyu Chen, Lingmin Gong, Yong Zhang, Hong Yan, Fangyao Chen and for the Alzheimer’s Disease Neuroimaging Initiative
Brain Sci. 2023, 13(11), 1535; https://doi.org/10.3390/brainsci13111535 - 31 Oct 2023
Cited by 1 | Viewed by 1829
Abstract
Background: Predicting cognition decline in patients with mild cognitive impairment (MCI) is crucial for identifying high-risk individuals and implementing effective management. To improve predicting MCI-to-AD conversion, it is necessary to consider various factors using explainable machine learning (XAI) models which provide interpretability while [...] Read more.
Background: Predicting cognition decline in patients with mild cognitive impairment (MCI) is crucial for identifying high-risk individuals and implementing effective management. To improve predicting MCI-to-AD conversion, it is necessary to consider various factors using explainable machine learning (XAI) models which provide interpretability while maintaining predictive accuracy. This study used the Explainable Boosting Machine (EBM) model with multimodal features to predict the conversion of MCI to AD during different follow-up periods while providing interpretability. Methods: This retrospective case-control study is conducted with data obtained from the ADNI database, with records of 1042 MCI patients from 2006 to 2022 included. The exposures included in this study were MRI biomarkers, cognitive scores, demographics, and clinical features. The main outcome was AD conversion from aMCI during follow-up. The EBM model was utilized to predict aMCI converting to AD based on three feature combinations, obtaining interpretability while ensuring accuracy. Meanwhile, the interaction effect was considered in the model. The three feature combinations were compared in different follow-up periods with accuracy, sensitivity, specificity, and AUC-ROC. The global and local explanations are displayed by importance ranking and feature interpretability plots. Results: The five-years prediction accuracy reached 85% (AUC = 0.92) using both cognitive scores and MRI markers. Apart from accuracies, we obtained features’ importance in different follow-up periods. In early stage of AD, the MRI markers play a major role, while for middle-term, the cognitive scores are more important. Feature risk scoring plots demonstrated insightful nonlinear interactive associations between selected factors and outcome. In one-year prediction, lower right inferior temporal volume (<9000) is significantly associated with AD conversion. For two-year prediction, low left inferior temporal thickness (<2) is most critical. For three-year prediction, higher FAQ scores (>4) is the most important. During four-year prediction, APOE4 is the most critical. For five-year prediction, lower right entorhinal volume (<1000) is the most critical feature. Conclusions: The established glass-box model EBMs with multimodal features demonstrated a superior ability with detailed interpretability in predicting AD conversion from MCI. Multi features with significant importance were identified. Further study may be of significance to determine whether the established prediction tool would improve clinical management for AD patients. Full article
(This article belongs to the Topic Translational Advances in Neurodegenerative Dementias)
Show Figures

Figure 1

Figure 1
<p>The flow chart of the whole study.</p>
Full article ">Figure 2
<p>Accuracy, sensitivity, specificity, and AUC, with (<b>a</b>–<b>d</b>), for classifier performance from 1 to 5 years of follow-up. Error bars show the associated standard deviation.</p>
Full article ">Figure 3
<p>The rankings of the overall feature importance and mean absolute score from 1 to 5 years of follow-up. With (<b>a</b>–<b>e</b>) corresponding to 1, 2, 3, 4, and 5-year follow-up periods.</p>
Full article ">Figure 4
<p>The uni-factor interpretation from 1 to 5 years of follow-up, with (<b>a</b>–<b>e</b>) corresponding to 1, 2, 3, 4, and 5-year follow up periods.</p>
Full article ">Figure 5
<p>The heatmap of interaction ranking 5 (<b>a</b>) and 6 (<b>b</b>) in 3-year follow up period.</p>
Full article ">
16 pages, 2290 KiB  
Article
Genome-Wide Association Analysis across Endophenotypes in Alzheimer’s Disease: Main Effects and Disease Stage-Specific Interactions
by Thea J. Rosewood, Kwangsik Nho, Shannon L. Risacher, Sujuan Gao, Li Shen, Tatiana Foroud, Andrew J. Saykin and on behalf of the Alzheimer’s Disease Neuroimaging Initiative
Genes 2023, 14(11), 2010; https://doi.org/10.3390/genes14112010 - 27 Oct 2023
Viewed by 1629
Abstract
The underlying genetic susceptibility for Alzheimer’s disease (AD) is not yet fully understood. The heterogeneous nature of the disease challenges genetic association studies. Endophenotype approaches can help to address this challenge by more direct interrogation of biological traits related to the disease. AD [...] Read more.
The underlying genetic susceptibility for Alzheimer’s disease (AD) is not yet fully understood. The heterogeneous nature of the disease challenges genetic association studies. Endophenotype approaches can help to address this challenge by more direct interrogation of biological traits related to the disease. AD endophenotypes based on amyloid-β, tau, and neurodegeneration (A/T/N) biomarkers and cognitive performance were selected from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort (N = 1565). A genome-wide association study (GWAS) of quantitative phenotypes was performed using an SNP main effect and an SNP by Diagnosis interaction (SNP × DX) model to identify disease stage-specific genetic effects. Nine loci were identified as study-wide significant with one or more A/T/N endophenotypes in the main effect model, as well as additional findings significantly associated with cognitive measures. These nine loci include SNPs in or near the genes APOE, SRSF10, HLA-DQB1, XKR3, and KIAA1671. The SNP × DX model identified three study-wide significant genetic loci (BACH2, EP300, and PACRG-AS1) with a neuroprotective effect in later AD stage endophenotypes. An endophenotype approach identified novel genetic associations and provided insight into the molecular mechanisms underlying the genetic associations that may otherwise be missed using conventional case-control study designs. Full article
Show Figures

Figure 1

Figure 1
<p>Matrix of main effect analysis results. Each row indicates the top SNP for a genetic region after LD trimming, and each column represents an AD endophenotype. Ordered based on minimum <span class="html-italic">p</span>-value across the row. The asterisks represent the <span class="html-italic">p</span>-value of the association with [***] indicating meeting the study-wide significant threshold (<span class="html-italic">p</span> ≤ 8.33 × 10<sup>−9</sup>), [**] the conventional genome-wide threshold (<span class="html-italic">p</span> ≤ 5 × 10<sup>−8</sup>), and [*] the suggestive association threshold (<span class="html-italic">p</span> ≤ 1 × 10<sup>−5</sup>). The color and box size relate to the β value effect size for a given association, with larger box size relating to distance from zero in either positive (blue, suggesting neuroprotective) or negative (red, suggesting neuropathological effect) direction. <sup>†</sup> SNP retains significance when including DX as a covariate.</p>
Full article ">Figure 2
<p>Matrix of SNP x Diagnosis analysis results. Each row indicates the top SNP for a genetic region after LD trimming, and each column represents an AD endophenotype. Endophenotypes showing no level of significance were removed for clarity. The asterisks represent the <span class="html-italic">p</span>-value of the association with [***] indicating meeting the study-wide significant threshold (<span class="html-italic">p</span> ≤ 8.33 × 10<sup>−9</sup>), [**] the conventional genome-wide threshold (<span class="html-italic">p</span> ≤ 5 × 10<sup>−8</sup>), and [*] the suggestive threshold (<span class="html-italic">p</span> ≤ 1 × 10<sup>−5</sup>). The color and box size relate to the β value effect size for a given association, with larger box size relating to distance from zero in either positive (blue, suggesting neuroprotective) or negative (red, suggesting neuropathological effect) direction.</p>
Full article ">Figure 3
<p>Violin and boxplot distribution of Parietal Lobe Cortical Thickness, stratified by (<b>A</b>) rs1065272 SNP, (<b>B</b>) Diagnosis, and (<b>C</b>) SNP and Diagnosis. (<b>A</b>) represents the main effect analysis, (<b>B</b>) the association with Diagnosis, and (<b>C</b>) the SNP × DX interaction association, with DX codes as an ordinal logistic variable of distinct categories with known ordinal relation (CN &lt; EMCI &lt; LMCI &lt; AD). The asterisks represent the <span class="html-italic">p</span>-value of the association with [***] indicating meeting the study-wide significant threshold (<span class="html-italic">p</span> ≤ 8.33 × 10<sup>−9</sup>).</p>
Full article ">
13 pages, 1269 KiB  
Article
Self-Supervised Contrastive Learning to Predict the Progression of Alzheimer’s Disease with 3D Amyloid-PET
by Min Gu Kwak, Yi Su, Kewei Chen, David Weidman, Teresa Wu, Fleming Lure, Jing Li and for the Alzheimer’s Disease Neuroimaging Initiative
Bioengineering 2023, 10(10), 1141; https://doi.org/10.3390/bioengineering10101141 - 28 Sep 2023
Cited by 1 | Viewed by 1479
Abstract
Early diagnosis of Alzheimer’s disease (AD) is an important task that facilitates the development of treatment and prevention strategies, and may potentially improve patient outcomes. Neuroimaging has shown great promise, including the amyloid-PET, which measures the accumulation of amyloid plaques in the brain—a [...] Read more.
Early diagnosis of Alzheimer’s disease (AD) is an important task that facilitates the development of treatment and prevention strategies, and may potentially improve patient outcomes. Neuroimaging has shown great promise, including the amyloid-PET, which measures the accumulation of amyloid plaques in the brain—a hallmark of AD. It is desirable to train end-to-end deep learning models to predict the progression of AD for individuals at early stages based on 3D amyloid-PET. However, commonly used models are trained in a fully supervised learning manner, and they are inevitably biased toward the given label information. To this end, we propose a selfsupervised contrastive learning method to accurately predict the conversion to AD for individuals with mild cognitive impairment (MCI) with 3D amyloid-PET. The proposed method, SMoCo, uses both labeled and unlabeled data to capture general semantic representations underlying the images. As the downstream task is given as classification of converters vs. non-converters, unlike the general self-supervised learning problem that aims to generate task-agnostic representations, SMoCo additionally utilizes the label information in the pre-training. To demonstrate the performance of our method, we conducted experiments on the Alzheimer’s Disease Neuroimaging Initiative (ADNI) dataset. The results confirmed that the proposed method is capable of providing appropriate data representations, resulting in accurate classification. SMoCo showed the best classification performance over the existing methods, with AUROC = 85.17%, accuracy = 81.09%, sensitivity = 77.39%, and specificity = 82.17%. While SSL has demonstrated great success in other application domains of computer vision, this study provided the initial investigation of using a proposed self-supervised contrastive learning model, SMoCo, to effectively predict MCI conversion to AD based on 3D amyloid-PET. Full article
(This article belongs to the Special Issue Artificial Intelligence in Biomedical Imaging)
Show Figures

Figure 1

Figure 1
<p>Distributions of demographic and clinical variables in ADNI dataset.</p>
Full article ">Figure 2
<p>Graphical overview of SMoCo. For a given image <math display="inline"><semantics> <msub> <mi>x</mi> <mi>i</mi> </msub> </semantics></math>, two augmentations are applied to generate a positive instance <math display="inline"><semantics> <msubsup> <mi>x</mi> <mi>i</mi> <mo>+</mo> </msubsup> </semantics></math> and an anchor <math display="inline"><semantics> <msubsup> <mi>x</mi> <mi>i</mi> <mi>a</mi> </msubsup> </semantics></math>. Both instances are fed into 3D ResNet-50 encoders <math display="inline"><semantics> <msub> <mi>f</mi> <mi>ϕ</mi> </msub> </semantics></math> and <math display="inline"><semantics> <msub> <mi>f</mi> <mi>θ</mi> </msub> </semantics></math> to obtain representations <math display="inline"><semantics> <msubsup> <mi>z</mi> <mi>i</mi> <mo>+</mo> </msubsup> </semantics></math> and <math display="inline"><semantics> <msubsup> <mi>z</mi> <mi>i</mi> <mi>a</mi> </msubsup> </semantics></math>, respectively. <math display="inline"><semantics> <msubsup> <mi mathvariant="script">L</mi> <mrow> <mi>i</mi> </mrow> <mrow> <mi>M</mi> <mi>o</mi> <mi>C</mi> <mi>o</mi> </mrow> </msubsup> </semantics></math> aims to pull <math display="inline"><semantics> <msubsup> <mi>z</mi> <mi>i</mi> <mo>+</mo> </msubsup> </semantics></math> toward <math display="inline"><semantics> <msubsup> <mi>z</mi> <mi>i</mi> <mi>a</mi> </msubsup> </semantics></math> because they are created from the same image, while pushing other instances in the memory queue away from <math display="inline"><semantics> <msubsup> <mi>z</mi> <mi>i</mi> <mi>a</mi> </msubsup> </semantics></math>. <math display="inline"><semantics> <msubsup> <mi mathvariant="script">L</mi> <mrow> <mi>i</mi> </mrow> <mrow> <mi>L</mi> <mi>a</mi> <mi>b</mi> <mi>e</mi> <mi>l</mi> </mrow> </msubsup> </semantics></math> leverages label information from the memory queue, ensuring the representations from the same class are pulled closer to <math display="inline"><semantics> <msubsup> <mi>z</mi> <mi>i</mi> <mi>a</mi> </msubsup> </semantics></math>. <math display="inline"><semantics> <msubsup> <mi mathvariant="script">L</mi> <mrow> <mi>i</mi> </mrow> <mrow> <mi>M</mi> <mi>o</mi> <mi>C</mi> <mi>o</mi> </mrow> </msubsup> </semantics></math> and <math display="inline"><semantics> <msubsup> <mi mathvariant="script">L</mi> <mrow> <mi>i</mi> </mrow> <mrow> <mi>L</mi> <mi>a</mi> <mi>b</mi> <mi>e</mi> <mi>l</mi> </mrow> </msubsup> </semantics></math> are combined as the final loss in SMoCo, <math display="inline"><semantics> <msubsup> <mi mathvariant="script">L</mi> <mrow> <mi>i</mi> </mrow> <mrow> <mi>S</mi> <mi>M</mi> <mi>o</mi> <mi>C</mi> <mi>o</mi> </mrow> </msubsup> </semantics></math>.</p>
Full article ">Figure 3
<p>(<b>a</b>) Structure of ResNet-50 encoder used for SMoCo (the same encoder is used for <math display="inline"><semantics> <msub> <mi>f</mi> <mi>ϕ</mi> </msub> </semantics></math> and <math display="inline"><semantics> <msub> <mi>f</mi> <mi>θ</mi> </msub> </semantics></math>). The numbers in a bracket denote <math display="inline"><semantics> <msub> <mi>K</mi> <mn>1</mn> </msub> </semantics></math>, <math display="inline"><semantics> <msub> <mi>K</mi> <mn>2</mn> </msub> </semantics></math>, and <math display="inline"><semantics> <msub> <mi>K</mi> <mn>3</mn> </msub> </semantics></math> of a 3D residual block, respectively. (<b>b</b>) Structure of 3D residual block in the encoder.</p>
Full article ">Figure 4
<p>UMAP visualization of the representations of training images. (<b>a</b>) SMoCo; (<b>b</b>) MoCo. Grey, blue, and red points refer to the unlabeled images, converters, and non-converters, respectively.</p>
Full article ">Figure 5
<p>Comparing AUROC of SMoCo and MoCo across training epochs. SMoCo shows faster and efficient training, as well as higher performance than MoCo.</p>
Full article ">
16 pages, 427 KiB  
Article
Taking Rational Numbers at Random
by Nicola Cufaro Petroni
AppliedMath 2023, 3(3), 648-663; https://doi.org/10.3390/appliedmath3030034 - 1 Sep 2023
Viewed by 1451
Abstract
In this article, some prescriptions to define a distribution on the set Q0 of all rational numbers in [0,1] are outlined. We explored a few properties of these distributions and the possibility of making these rational numbers asymptotically [...] Read more.
In this article, some prescriptions to define a distribution on the set Q0 of all rational numbers in [0,1] are outlined. We explored a few properties of these distributions and the possibility of making these rational numbers asymptotically equiprobable in a suitable sense. In particular, it will be shown that in the said limit—albeit no absolutely continuous uniform distribution can be properly defined in Q0—the probability allotted to every single qQ0 asymptotically vanishes, while that of the subset of Q0 falling in an interval [a,b]Q0 goes to ba. We finally present some hints to complete sequencing without repeating the numbers in Q0 as a prerequisite to laying down more distributions on it. Full article
(This article belongs to the Special Issue Applications of Number Theory to the Sciences and Mathematics)
Show Figures

Figure 1

Figure 1
<p>Probabilities (<a href="#FD17-appliedmath-03-00034" class="html-disp-formula">17</a>) attributed to rational numbers as a function of the irreducible, geometrically distributed denominators <span class="html-italic">m</span>, and for decreasing (<math display="inline"><semantics> <mrow> <mn>0.9</mn> <mo>,</mo> <mn>0.5</mn> <mo>,</mo> <mn>0.1</mn> <mo>,</mo> <mn>0.01</mn> <mo>,</mo> <mn>0.001</mn> </mrow> </semantics></math>) values of <span class="html-italic">w</span>: by choosing different <span class="html-italic">m</span> intervals, the pictures show how these probabilities level down to infinitesimal equiprobability for <math display="inline"><semantics> <mrow> <mi>w</mi> <mo>→</mo> <mn>0</mn> </mrow> </semantics></math>.</p>
Full article ">Figure 2
<p>Numerosity <math display="inline"><semantics> <msub> <mi>ν</mi> <mi>m</mi> </msub> </semantics></math> of the different rational numbers <math display="inline"><semantics> <mrow> <mi>q</mi> <mo>≐</mo> <mspace width="0.166667em"/> <msup> <mspace width="-0.166667em"/> <mi>n</mi> </msup> <msub> <mo>/</mo> <mi>m</mi> </msub> </mrow> </semantics></math> sharing a common, irreducible denominator <span class="html-italic">m</span>.</p>
Full article ">Figure 3
<p>Typical histogram of the relative frequencies of a sample of <math display="inline"><semantics> <msup> <mn>10</mn> <mn>5</mn> </msup> </semantics></math> random rationals generated following the procedure described in <a href="#sec6dot2-appliedmath-03-00034" class="html-sec">Section 6.2</a>: here, the maximum value of the equiprobable denominators is chosen to be <math display="inline"><semantics> <mrow> <mi>k</mi> <mo>=</mo> <msup> <mn>10</mn> <mn>5</mn> </msup> </mrow> </semantics></math>.</p>
Full article ">
Back to TopTop