Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 506
Filter
1.
Article in English | MEDLINE | ID: mdl-38742150

ABSTRACT

Glioblastoma (GBM) is most aggressive and common adult brain tumor. The standard treatments typically include maximal surgical resection, followed adjuvant radiotherapy and chemotherapy. However, the efficacy of these treatment is often limited, as tumor often infiltrate into the surrounding brain tissue, often extending beyond the radiologically defined margins. This infiltration contributes to the high recurrence rate and poor prognosis associated with GBM patients, necessitating advanced methods for early and accurate detection of tumor infiltration. Despite the great promise traditional supervised machine learning shows in predicting tumor infiltration beyond resectable margins, these methods are heavily reliant on expert-drawn Regions of Interest (ROIs), which are used to construct multi-variate models of different Magnetic Resonance (MR) signal characteristics associated with tumor infiltration. This process is both time consuming and resource intensive. Addressing this limitation, our study proposes a novel integration of fully automatic methods for generating ROIs with deep learning algorithms to create predictive maps of tumor infiltration. This approach uses pre-operative multi-parametric MRI (mpMRI) scans, encompassing T1, T1Gd, T2, T2-FLAIR, and ADC sequences, to fully leverage the knowledge from previously drawn ROIs. Subsequently, a patch based Convolutional Neural Network (CNN) model is trained on these automatically generated ROIs to predict areas of potential tumor infiltration. The performance of this model was evaluated using a leave-one-out cross-validation approach. Generated predictive maps binarized for comparison against post-recurrence mpMRI scans. The model demonstrates robust predictive capability, evidenced by the average cross-validated accuracy of 0.87, specificity of 0.88, and sensitivity of 0.90. Notably, the odds ratio of 8.62 indicates that regions identified as high-risk on the predictive map were significantly more likely to exhibit tumor recurrence than low-risk regions. The proposed method demonstrates that a fully automatic mpMRI analysis using deep learning can successfully predict tumor infiltration in peritumoral region for GBM patients while bypassing the intensive requirement for expert-drawn ROIs.

2.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746228

ABSTRACT

Personalized functional networks (FNs) derived from functional magnetic resonance imaging (fMRI) data are useful for characterizing individual variations in the brain functional topography associated with the brain development, aging, and disorders. To facilitate applications of the personalized FNs with enhanced reliability and reproducibility, we develop an open-source toolbox that is user-friendly, extendable, and includes rigorous quality control (QC), featuring multiple user interfaces (graphics, command line, and a step-by-step guideline) and job-scheduling for high performance computing (HPC) clusters. Particularly, the toolbox, named personalized functional network modeling (pNet), takes fMRI inputs in either volumetric or surface type, ensuring compatibility with multiple fMRI data formats, and computes personalized FNs using two distinct modeling methods: one method optimizes the functional coherence of FNs, while the other enhances their independence. Additionally, the toolbox provides HTML-based reports for QC and visualization of personalized FNs. The toolbox is developed in both MATLAB and Python platforms with a modular design to facilitate extension and modification by users familiar with either programming language. We have evaluated the toolbox on two fMRI datasets and demonstrated its effectiveness and user-friendliness with interactive and scripting examples. pNet is publicly available at https://github.com/MLDataAnalytics/pNet .

3.
Biol Psychiatry ; 2024 May 06.
Article in English | MEDLINE | ID: mdl-38718880

ABSTRACT

Machine learning has been increasingly used to obtain individualized neuroimaging signatures for disease diagnosis, prognosis, and response to treatment in neuropsychiatric and neurodegenerative disorders. Therefore, it has contributed to a better understanding of disease heterogeneity by identifying disease subtypes with different brain phenotypic measures. In this Review, we first present a systematic literature overview of studies using machine learning and multimodal MRI to unravel disease heterogeneity in various neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, schizophrenia, major depressive disorder, autism spectrum disorder, multiple sclerosis, as well as their potential in a transdiagnostic framework, where neuroanatomical and neurobiological commonalities were assessed across diagnostic boundaries. Subsequently, we summarize relevant machine learning methodologies and their clinical interpretability. We discuss the potential clinical implications of the current findings and envision future research avenues. Finally, we discuss an emerging paradigm called dimensional neuroimaging endophenotypes (DNE). DNE dissects the neurobiological heterogeneity of neuropsychiatric and neurodegenerative disorders into low-dimensional yet informative, quantitative brain phenotypic representations, serving as robust intermediate phenotypes (i.e., endophenotypes) presumably reflecting the interplay of underlying genetic, lifestyle, and environmental processes associated with the disease etiology.

4.
Article in English | MEDLINE | ID: mdl-38656243

ABSTRACT

It is not well understood how neighborhood disadvantage is associated with specific domains of cognitive function and underlying brain health within older adults. Thus, the objective was to examine associations between neighborhood disadvantage, brain health, and cognitive performance, and examine whether associations were more pronounced among women. The study included 136 older adults who underwent cognitive testing and MRI. Neighborhood disadvantage was characterized using the Area Deprivation Index (ADI). Descriptive statistics, bivariate correlations, and multiple regressions were run. Multiple regressions, adjusted for age, sex, education, and depression, showed that higher ADI state rankings (greater disadvantage) were associated with poorer working memory performance (p < .01) and lower hippocampal volumes (p < .01), but not total, frontal, and white matter lesion volumes, nor visual and verbal memory performance. There were no significant sex interactions. Findings suggest that greater neighborhood disadvantage may play a role in working memory and underlying brain structure.

5.
Article in English | MEDLINE | ID: mdl-38650741

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive brain tumor with short overall survival (OS) of about 15 months. Understanding the causal factors affecting the patient survival is crucial for disease prognosis and treatment planning. Although previous efforts on survival prediction using multi-omics data has yielded useful predictive models, the causation of the correlated genetic risk factors has not been addressed. Recent advances in causal deep learning models enable the study of causality from complex dataset. In this paper, we leverage the recently proposed structural causal model (SCM) with normalizing flows parameterized by deep networks to perform the counterfactual query to investigate the causal relationship between gene mutation and OS with the presence of other confounders including sex, age and radiomics features. The query amounts to the question that what the survival days will be if the gene mutation status has been changed, i.e., from mutant to non-mutant and vice versa. The trained causal model will infer the counterfactual outcome given the intervention on specific gene mutation. We apply multivariate Cox-PH model to find the genes associated with survival, and investigate the causal genetic effect by comparing the original and counterfactual survival days in a bi-directional fashion. Particularly, the following two scenarios are considered: (1) intervention on a specific gene with non-mutant status to generate the counterfactual survival days as if the gene is mutant, with which the original survival days of the subjects with that mutant gene will be compared; (2) intervention on the gene with mutant status and perform the comparison with survival days of subjects with that non-mutant gene. Our experimental results show that no causation of two correlated genes (NF1, RB1) was revealed in the cohort (n=181), while their genetic effects on OS in terms of prolonging or shortening are generally in accordance with clinical findings.

6.
Article in English | MEDLINE | ID: mdl-38651004

ABSTRACT

Radiomics has been widely recognized for its effectiveness in decoding tumor phenotypes through the extraction of quantitative imaging features. However, the robustness of radiomic methods to estimate clinically relevant biomarkers non-invasively remains largely untested. In this study, we propose Cascaded Data Processing Network (CDPNet), a radiomic feature learning method to predict tumor molecular status from medical images. We apply CDPNet to an epigenetic case, specifically targeting the estimation of O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation from Magnetic Resonance Imaging (MRI) scans of glioblastoma patients. CDPNet has three components: 1) Principal Component Analysis (PCA), 2) Fisher Linear Discriminant (FLD), and 3) a combination of hashing and blockwise histograms. The outlined architectural framework capitalizes on PCA to reconstruct input image patches, followed by FLD to extract discriminative filter banks, and finally using binary hashing and blockwise histogram module for indexing, pooling, and feature generation. To validate the effectiveness of CDPNet, we conducted an exhaustive evaluation on a comprehensive retrospective cohort comprising 484 IDH-wildtype glioblastoma patients with pre-operative multi-parametric MRI scans (T1, T1-Gd, T2, and T2-FLAIR). The prediction of MGMT promoter methylation status was cast as a binary classification problem. The developed model underwent rigorous training via 10-fold cross-validation on a discovery cohort of 446 patients. Subsequently, the model's performance was evaluated on a distinct and previously unseen replication cohort of 38 patients. Our method achieved an accuracy of 70.11% and an area under the curve of 0.71 (95% CI: 0.65 - 0.74).

7.
Article in English | MEDLINE | ID: mdl-38684319

ABSTRACT

BACKGROUND: Understanding sex-based differences in glioblastoma patients is necessary for accurate personalized treatment planning to improve patient outcomes. PURPOSE: To investigate sex-specific differences in molecular, clinical and radiological tumor parameters, as well as survival outcomes in glioblastoma, isocitrate dehydrogenase-1 wildtype (IDH1-WT), grade 4 patients. METHODS: Retrospective data of 1832 glioblastoma, IDH1-WT patients with comprehensive information on tumor parameters was acquired from the Radiomics Signatures for Precision Oncology in Glioblastoma (ReSPOND) consortium. Data imputation was performed for missing values. Sex-based differences in tumor parameters, such as, age, molecular parameters, pre-operative KPS score, tumor volumes, epicenter and laterality were assessed through non-parametric tests. Spatial atlases were generated using pre-operative MRI maps to visualize tumor characteristics. Survival time analysis was performed through log-rank tests and Cox proportional hazard analyses. RESULTS: GBM was diagnosed at a median age of 64 years in females compared to 61.9 years in males (FDR = 0.003). Males had a higher Karnofsky Performance Score (above 80) as compared to females (60.4% females Vs 69.7% males, FDR = 0.044). Females had lower tumor volumes in enhancing (16.7 cm3 Vs. 20.6 cm3 in males, FDR = 0.001), necrotic core (6.18 cm3 Vs. 7.76 cm3 in males, FDR = 0.001) and edema regions (46.9 cm3 Vs. 59.2 cm3 in males, FDR = 0.0001). Right temporal region was the most common tumor epicenter in the overall population. Right as well as left temporal lobes were more frequently involved in males. There were no significant differences in survival outcomes and mortality ratios. Higher age, unmethylated O6-methylguanine-DNAmethyltransferase (MGMT) promoter and undergoing subtotal resection increased the mortality risk in both males and females. CONCLUSIONS: Our study demonstrates significant sex-based differences in clinical and radiological tumor parameters of glioblastoma, IDH1-WT, grade 4 patients. Sex is not an independent prognostic factor for survival outcomes and the tumor parameters influencing patient outcomes are identical for males and females. ABBREVIATIONS: IDH1-WT = isocitrate dehydrogenase-1 wildtype; MGMTp = O6-methylguanine-DNA-methyltransferase promoter; KPS = Karnofsky performance score; EOR = extent of resection; WHO = world health organization; FDR = false discovery rate.

8.
Article in English | MEDLINE | ID: mdl-38584725

ABSTRACT

We introduce an informative metric, called morphometric correlation, as a measure of shared neuroanatomic similarity between two cognitive traits. Traditional estimates of trait correlations can be confounded by factors beyond brain morphology. To exclude these confounding factors, we adopt a Gaussian kernel to measure the morphological similarity between individuals and compare pure neuroanatomic correlations among cognitive traits. In our empirical study, we employ a multiscale strategy. Given a set of cognitive traits, we first perform morphometric correlation analysis for each pair of traits to reveal their shared neuroanatomic correlation at the whole brain (or global) level. After that, we extend our whole brain concept to regional morphometric correlation and estimate shared neuroanatomic similarity between two cognitive traits at the regional (or local) level. Our results demonstrate that morphometric correlation can provide insights into shared neuroanatomic architecture between cognitive traits. Furthermore, we also estimate the morphometricity of each cognitive trait at both global and local levels, which can be used to better understand how neuroanatomic changes influence individuals' cognitive status.

9.
Alzheimers Res Ther ; 16(1): 94, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689358

ABSTRACT

BACKGROUND: Although blood-based biomarkers have been identified as cost-effective and scalable alternatives to PET and CSF markers of neurodegenerative disease, little is known about how these biomarkers predict future brain atrophy and cognitive decline in cognitively unimpaired individuals. Using data from the Baltimore Longitudinal Study of Aging (BLSA), we examined whether plasma biomarkers of Alzheimer's disease (AD) pathology (amyloid-ß [Aß42/40], phosphorylated tau [pTau-181]), astrogliosis (glial fibrillary acidic protein [GFAP]), and neuronal injury (neurofilament light chain [NfL]) were associated with longitudinal brain volume loss and cognitive decline. Additionally, we determined whether sex, APOEε4 status, and plasma amyloid-ß status modified these associations. METHODS: Plasma biomarkers were measured using Quanterix SIMOA assays. Regional brain volumes were measured by 3T MRI, and a battery of neuropsychological tests assessed five cognitive domains. Linear mixed effects models adjusted for demographic factors, kidney function, and intracranial volume (MRI analyses) were completed to relate baseline plasma biomarkers to baseline and longitudinal brain volume and cognitive performance. RESULTS: Brain volume analyses included 622 participants (mean age ± SD: 70.9 ± 10.2) with an average of 3.3 MRI scans over 4.7 years. Cognitive performance analyses included 674 participants (mean age ± SD: 71.2 ± 10.0) with an average of 3.9 cognitive assessments over 5.7 years. Higher baseline pTau-181 was associated with steeper declines in total gray matter volume and steeper regional declines in several medial temporal regions, whereas higher baseline GFAP was associated with greater longitudinal increases in ventricular volume. Baseline Aß42/40 and NfL levels were not associated with changes in brain volume. Lower baseline Aß42/40 (higher Aß burden) was associated with a faster decline in verbal memory and visuospatial performance, whereas higher baseline GFAP was associated with a faster decline in verbal fluency. Results were generally consistent across sex and APOEε4 status. However, the associations of higher pTau-181 with increasing ventricular volume and memory declines were significantly stronger among individuals with higher Aß burden, as was the association of higher GFAP with memory decline. CONCLUSIONS: Among cognitively unimpaired older adults, plasma biomarkers of AD pathology (pTau-181) and astrogliosis (GFAP), but not neuronal injury (NfL), serve as markers of future brain atrophy and cognitive decline.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Atrophy , Biomarkers , Brain , Cognitive Dysfunction , tau Proteins , Humans , Female , Male , Biomarkers/blood , Aged , Atrophy/pathology , Brain/pathology , Brain/diagnostic imaging , Alzheimer Disease/blood , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/blood , Cognitive Dysfunction/blood , Cognitive Dysfunction/pathology , tau Proteins/blood , tau Proteins/cerebrospinal fluid , Longitudinal Studies , Glial Fibrillary Acidic Protein/blood , Middle Aged , Aged, 80 and over , Neurofilament Proteins/blood , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/pathology , Neuropsychological Tests , Magnetic Resonance Imaging , Peptide Fragments/blood
10.
Nat Commun ; 15(1): 2604, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38521789

ABSTRACT

The complex biological mechanisms underlying human brain aging remain incompletely understood. This study investigated the genetic architecture of three brain age gaps (BAG) derived from gray matter volume (GM-BAG), white matter microstructure (WM-BAG), and functional connectivity (FC-BAG). We identified sixteen genomic loci that reached genome-wide significance (P-value < 5×10-8). A gene-drug-disease network highlighted genes linked to GM-BAG for treating neurodegenerative and neuropsychiatric disorders and WM-BAG genes for cancer therapy. GM-BAG displayed the most pronounced heritability enrichment in genetic variants within conserved regions. Oligodendrocytes and astrocytes, but not neurons, exhibited notable heritability enrichment in WM and FC-BAG, respectively. Mendelian randomization identified potential causal effects of several chronic diseases on brain aging, such as type 2 diabetes on GM-BAG and AD on WM-BAG. Our results provide insights into the genetics of human brain aging, with clinical implications for potential lifestyle and therapeutic interventions. All results are publicly available at https://labs.loni.usc.edu/medicine .


Subject(s)
Diabetes Mellitus, Type 2 , White Matter , Humans , Brain , Gray Matter , Magnetic Resonance Imaging/methods , White Matter/physiology , Mendelian Randomization Analysis
11.
iScience ; 27(3): 109212, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38433927

ABSTRACT

Traditional loss functions such as cross-entropy loss often quantify the penalty for each mis-classified training sample without adequately considering its distance from the ground truth class distribution in the feature space. Intuitively, the larger this distance is, the higher the penalty should be. With this observation, we propose a penalty called distance-weighted Sinkhorn (DWS) loss. For each mis-classified training sample (with predicted label A and true label B), its contribution to the DWS loss positively correlates to the distance the training sample needs to travel to reach the ground truth distribution of all the A samples. We apply the DWS framework with a neural network to classify different stages of Alzheimer's disease. Our empirical results demonstrate that the DWS framework outperforms the traditional neural network loss functions and is comparable or better to traditional machine learning methods, highlighting its potential in biomedical informatics and data science.

12.
Sci Rep ; 14(1): 4922, 2024 02 28.
Article in English | MEDLINE | ID: mdl-38418494

ABSTRACT

Glioblastoma is a highly heterogeneous disease, with variations observed at both phenotypical and molecular levels. Personalized therapies would be facilitated by non-invasive in vivo approaches for characterizing this heterogeneity. In this study, we developed unsupervised joint machine learning between radiomic and genomic data, thereby identifying distinct glioblastoma subtypes. A retrospective cohort of 571 IDH-wildtype glioblastoma patients were included in the study, and pre-operative multi-parametric MRI scans and targeted next-generation sequencing (NGS) data were collected. L21-norm minimization was used to select a subset of 12 radiomic features from the MRI scans, and 13 key driver genes from the five main signal pathways most affected in glioblastoma were selected from the genomic data. Subtypes were identified using a joint learning approach called Anchor-based Partial Multi-modal Clustering on both radiomic and genomic modalities. Kaplan-Meier analysis identified three distinct glioblastoma subtypes: high-risk, medium-risk, and low-risk, based on overall survival outcome (p < 0.05, log-rank test; Hazard Ratio = 1.64, 95% CI 1.17-2.31, Cox proportional hazard model on high-risk and low-risk subtypes). The three subtypes displayed different phenotypical and molecular characteristics in terms of imaging histogram, co-occurrence of genes, and correlation between the two modalities. Our findings demonstrate the synergistic value of integrated radiomic signatures and molecular characteristics for glioblastoma subtyping. Joint learning on both modalities can aid in better understanding the molecular basis of phenotypical signatures of glioblastoma, and provide insights into the biological underpinnings of tumor formation and progression.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/diagnostic imaging , Glioblastoma/genetics , Retrospective Studies , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Prognosis , Magnetic Resonance Imaging/methods , Genomics
13.
J Alzheimers Dis ; 98(1): 231-246, 2024.
Article in English | MEDLINE | ID: mdl-38393899

ABSTRACT

Background: Blood-based biomarkers (BBMs) are of growing interest in the field of Alzheimer's disease (AD) and related dementias. Objective: This study aimed to assess the ability of plasma biomarkers to 1) predict disease progression from mild cognitive impairment (MCI) to dementia and 2) improve the predictive ability of magnetic resonance imaging (MRI) and cerebrospinal fluid (CSF) measures when combined. Methods: We used data from the Alzheimer's Disease Neuroimaging Initiative. Machine learning models were trained using the data from participants who remained cognitively stable (CN-s) and with Dementia diagnosis at 2-year follow-up visit. The models were used to predict progression to dementia in MCI individuals. We assessed the performance of models with plasma biomarkers against those with CSF and MRI measures, and also in combination with them. Results: Our models with plasma biomarkers classified CN-s individuals from AD with an AUC of 0.75±0.03 and could predict conversion to dementia in MCI individuals with an AUC of 0.64±0.03 (17.1% BP, base prevalence). Models with plasma biomarkers performed better when combined with CSF and MRI measures (CN versus AD: AUC of 0.89±0.02; MCI-to-AD: AUC of 0.76±0.03, 21.5% BP). Conclusions: Our results highlight the potential of plasma biomarkers in predicting conversion to dementia in MCI individuals. While plasma biomarkers could improve the predictive ability of CSF and MRI measures when combined, they also show the potential to predict non-progression to AD when considered alone. The predictive ability of plasma biomarkers is crucially linked to reducing the costly and effortful collection of CSF and MRI measures.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/cerebrospinal fluid , Disease Progression , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Magnetic Resonance Imaging/methods , Amyloid beta-Peptides/cerebrospinal fluid , tau Proteins/cerebrospinal fluid
14.
ArXiv ; 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38313197

ABSTRACT

Machine learning has been increasingly used to obtain individualized neuroimaging signatures for disease diagnosis, prognosis, and response to treatment in neuropsychiatric and neurodegenerative disorders. Therefore, it has contributed to a better understanding of disease heterogeneity by identifying disease subtypes that present significant differences in various brain phenotypic measures. In this review, we first present a systematic literature overview of studies using machine learning and multimodal MRI to unravel disease heterogeneity in various neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, schizophrenia, major depressive disorder, autism spectrum disorder, multiple sclerosis, as well as their potential in transdiagnostic settings. Subsequently, we summarize relevant machine learning methodologies and discuss an emerging paradigm which we call dimensional neuroimaging endophenotype (DNE). DNE dissects the neurobiological heterogeneity of neuropsychiatric and neurodegenerative disorders into a low-dimensional yet informative, quantitative brain phenotypic representation, serving as a robust intermediate phenotype (i.e., endophenotype) largely reflecting underlying genetics and etiology. Finally, we discuss the potential clinical implications of the current findings and envision future research avenues.

15.
Nat Commun ; 15(1): 354, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191573

ABSTRACT

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


Subject(s)
Alzheimer Disease , Neuroimaging , Humans , Endophenotypes , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Brain/diagnostic imaging , Cluster Analysis
16.
medRxiv ; 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-37398441

ABSTRACT

Understanding the genetic basis of biological aging in multi-organ systems is vital for elucidating age-related disease mechanisms and identifying therapeutic interventions. This study characterized the genetic architecture of the biological age gap (BAG) across nine human organ systems in 377,028 individuals of European ancestry from the UK Biobank. We discovered 393 genomic loci-BAG pairs (P-value<5×10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary, and renal systems. We observed BAG-organ specificity and inter-organ connections. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system while exerting pleiotropic effects on traits linked to multiple organ systems. A gene-drug-disease network confirmed the involvement of the metabolic BAG-associated genes in drugs targeting various metabolic disorders. Genetic correlation analyses supported Cheverud's Conjecture1 - the genetic correlation between BAGs mirrors their phenotypic correlation. A causal network revealed potential causal effects linking chronic diseases (e.g., Alzheimer's disease), body weight, and sleep duration to the BAG of multiple organ systems. Our findings shed light on promising therapeutic interventions to enhance human organ health within a complex multi-organ network, including lifestyle modifications and potential drug repositioning strategies for treating chronic diseases. All results are publicly available at https://labs-laboratory.com/medicine.

17.
Article in English | MEDLINE | ID: mdl-37837441

ABSTRACT

BACKGROUND: Poor motor function is associated with brain atrophy and cognitive impairment. Less is known about the relationship between motor domains and brain atrophy and whether associations are affected by cerebrovascular burden and/or physical activity. METHODS: We analyzed data from 726 Baltimore Longitudinal Study of Aging participants (mean age 70.6 ±â€…10.1 years, 56% women, 27% Black), 525 of whom had repeat MRI scans over an average of 5.0 ±â€…2.1 years. Two motor domains, manual dexterity and gross motor, were operationalized as latent variables. Associations between the latent variables and cortical and subcortical brain volumes of interest were examined using latent growth curve modeling, adjusted for demographics, white matter hyperintensities, and physical activity. RESULTS: Both higher manual dexterity and gross motor function were cross-sectionally associated with smaller ventricular volume and greater white matter volumes in the frontal, parietal, and temporal lobes (all p < .05). Manual dexterity was also cross-sectionally associated with parietal gray matter (B = 0.14; 95% CI: 0.05, 0.23), hippocampus (B = 0.10; 95% CI: 0.01, 0.20), postcentral gyrus (B = 0.11; 95% CI: 0.01, 0.20), and occipital white matter (B = 0.10; 95% CI: 0.01, 0.21) volumes, and gross motor function with temporal gray matter volume (B = 0.16; 95% CI: 0.05, 0.26). Longitudinally, both higher manual dexterity and gross motor function were associated with less temporal white matter and occipital gray matter atrophy (all p < .05). Manual dexterity was also associated with a slower rate of ventricular enlargement (B = -0.17; 95% CI: -0.29, -0.05) and less atrophy of occipital white matter (B = 0.39; 95% CI: 0.04, 0.71). CONCLUSIONS: Among cognitively normal middle- and older-aged adults, manual dexterity and gross motor function exhibited shared as well as distinct associations with brain atrophy over time.


Subject(s)
Neurodegenerative Diseases , White Matter , Humans , Female , Adult , Middle Aged , Aged , Aged, 80 and over , Male , Brain/pathology , Longitudinal Studies , Gray Matter/diagnostic imaging , Gray Matter/pathology , Aging , White Matter/diagnostic imaging , White Matter/pathology , Magnetic Resonance Imaging , Atrophy
18.
Alzheimers Dement ; 20(3): 1586-1600, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38050662

ABSTRACT

INTRODUCTION: Variability in relationship of tau-based neurofibrillary tangles (T) and neurodegeneration (N) in Alzheimer's disease (AD) arises from non-specific nature of N, modulated by non-AD co-pathologies, age-related changes, and resilience factors. METHODS: We used regional T-N residual patterns to partition 184 patients within the Alzheimer's continuum into data-driven groups. These were compared with groups from 159 non-AD (amyloid "negative") patients partitioned using cortical thickness, and groups in 98 patients with ante mortem MRI and post mortem tissue for measuring N and T, respectively. We applied the initial T-N residual model to classify 71 patients in an independent cohort into predefined groups. RESULTS: AD groups displayed spatial T-N mismatch patterns resembling neurodegeneration patterns in non-AD groups, similarly associated with non-AD factors and diverging cognitive outcomes. In the autopsy cohort, limbic T-N mismatch correlated with TDP-43 co-pathology. DISCUSSION: T-N mismatch may provide a personalized approach for determining non-AD factors associated with resilience/vulnerability in AD.


Subject(s)
Alzheimer Disease , Resilience, Psychological , Humans , Alzheimer Disease/pathology , tau Proteins , Neurofibrillary Tangles/pathology , Magnetic Resonance Imaging , Amyloid beta-Peptides
19.
Invest Radiol ; 59(5): 359-371, 2024 May 01.
Article in English | MEDLINE | ID: mdl-37812483

ABSTRACT

OBJECTIVE: Given the limited repeatability and reproducibility of radiomic features derived from weighted magnetic resonance imaging (MRI), there may be significant advantages to using radiomics in conjunction with quantitative MRI. This study introduces a novel physics-informed discretization (PID) method for reproducible radiomic feature extraction and evaluates its performance using quantitative MRI sequences including magnetic resonance fingerprinting (MRF) and apparent diffusion coefficient (ADC) mapping. MATERIALS AND METHODS: A multiscanner, scan-rescan dataset comprising whole-brain 3D quantitative (MRF T1, MRF T2, and ADC) and weighted MRI (T1w MPRAGE, T2w SPACE, and T2w FLAIR) from 5 healthy subjects was prospectively acquired. Subjects underwent 2 repeated acquisitions on 3 distinct 3 T scanners each, for a total of 6 scans per subject (30 total scans). First-order statistical (n = 23) and second-order texture (n = 74) radiomic features were extracted from 56 brain tissue regions of interest using the proposed PID method (for quantitative MRI) and conventional fixed bin number (FBN) discretization (for quantitative MRI and weighted MRI). Interscanner radiomic feature reproducibility was measured using the intraclass correlation coefficient (ICC), and the effect of image sequence (eg, MRF T1 vs T1w MPRAGE), as well as image discretization method (ie, PID vs FBN), on radiomic feature reproducibility was assessed using repeated measures analysis of variance. The robustness of PID and FBN discretization to segmentation error was evaluated by simulating segmentation differences in brainstem regions of interest. Radiomic features with ICCs greater than 0.75 following simulated segmentation were determined to be robust to segmentation. RESULTS: First-order features demonstrated higher reproducibility in quantitative MRI than weighted MRI sequences, with 30% (n = 7/23) features being more reproducible in MRF T1 and MRF T2 than weighted MRI. Gray level co-occurrence matrix (GLCM) texture features extracted from MRF T1 and MRF T2 were significantly more reproducible using PID compared with FBN discretization; for all quantitative MRI sequences, PID yielded the highest number of texture features with excellent reproducibility (ICC > 0.9). Comparing texture reproducibility of quantitative and weighted MRI, a greater proportion of MRF T1 (n = 225/370, 61%) and MRF T2 (n = 150/370, 41%) texture features had excellent reproducibility (ICC > 0.9) compared with T1w MPRAGE (n = 148/370, 40%), ADC (n = 115/370, 32%), T2w SPACE (n = 98/370, 27%), and FLAIR (n = 102/370, 28%). Physics-informed discretization was also more robust than FBN discretization to segmentation error, as 46% (n = 103/222, 46%) of texture features extracted from quantitative MRI using PID were robust to simulated 6 mm segmentation shift compared with 19% (n = 42/222, 19%) of weighted MRI texture features extracted using FBN discretization. CONCLUSIONS: The proposed PID method yields radiomic features extracted from quantitative MRI sequences that are more reproducible and robust than radiomic features extracted from weighted MRI using conventional (FBN) discretization approaches. Quantitative MRI sequences also demonstrated greater scan-rescan robustness and first-order feature reproducibility than weighted MRI.


Subject(s)
Magnetic Resonance Imaging , Radiomics , Humans , Reproducibility of Results , Magnetic Resonance Imaging/methods , Diffusion Magnetic Resonance Imaging , Brain/diagnostic imaging , Image Processing, Computer-Assisted/methods
20.
Ann Neurol ; 95(2): 260-273, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37801487

ABSTRACT

OBJECTIVE: Few studies have comprehensively examined how health and disease risk influence Alzheimer's disease (AD) biomarkers. The present study examined the association of 14 protein-based health indicators with plasma and neuroimaging biomarkers of AD and neurodegeneration. METHODS: In 706 cognitively normal adults, we examined whether 14 protein-based health indices (ie, SomaSignal® tests) were associated with concurrently measured plasma-based biomarkers of AD pathology (amyloid-ß [Aß]42/40 , tau phosphorylated at threonine-181 [pTau-181]), neuronal injury (neurofilament light chain [NfL]), and reactive astrogliosis (glial fibrillary acidic protein [GFAP]), brain volume, and cortical Aß and tau. In a separate cohort (n = 11,285), we examined whether protein-based health indicators associated with neurodegeneration also predict 25-year dementia risk. RESULTS: Greater protein-based risk for cardiovascular disease, heart failure mortality, and kidney disease was associated with lower Aß42/40 and higher pTau-181, NfL, and GFAP levels, even in individuals without cardiovascular or kidney disease. Proteomic indicators of body fat percentage, lean body mass, and visceral fat were associated with pTau-181, NfL, and GFAP, whereas resting energy rate was negatively associated with NfL and GFAP. Together, these health indicators predicted 12, 31, 50, and 33% of plasma Aß42/40 , pTau-181, NfL, and GFAP levels, respectively. Only protein-based measures of cardiovascular risk were associated with reduced regional brain volumes; these measures predicted 25-year dementia risk, even among those without clinically defined cardiovascular disease. INTERPRETATION: Subclinical peripheral health may influence AD and neurodegenerative disease processes and relevant biomarker levels, particularly NfL. Cardiovascular health, even in the absence of clinically defined disease, plays a central role in brain aging and dementia. ANN NEUROL 2024;95:260-273.


Subject(s)
Alzheimer Disease , Cardiovascular Diseases , Kidney Diseases , Neurodegenerative Diseases , Adult , Humans , Alzheimer Disease/diagnostic imaging , Proteomics , Amyloid beta-Peptides , Biomarkers , tau Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...