170 research outputs found

    Systems modeling of white matter microstructural abnormalities in Alzheimer's disease

    Get PDF
    INTRODUCTION: Microstructural abnormalities in white matter (WM) are often reported in Alzheimer's disease (AD). However, it is unclear which brain regions have the strongest WM changes in presymptomatic AD and what biological processes underlie WM abnormality during disease progression. METHODS: We developed a systems biology framework to integrate matched diffusion tensor imaging (DTI), genetic and transcriptomic data to investigate regional vulnerability to AD and identify genetic risk factors and gene subnetworks underlying WM abnormality in AD. RESULTS: We quantified regional WM abnormality and identified most vulnerable brain regions. A SNP rs2203712 in CELF1 was most significantly associated with several DTI-derived features in the hippocampus, the top ranked brain region. An immune response gene subnetwork in the blood was most correlated with DTI features across all the brain regions. DISCUSSION: Incorporation of image analysis with gene network analysis enhances our understanding of disease progression and facilitates identification of novel therapeutic strategies for AD

    A Knowledge-based Integrative Modeling Approach for <em>In-Silico</em> Identification of Mechanistic Targets in Neurodegeneration with Focus on Alzheimer’s Disease

    Get PDF
    Dementia is the progressive decline in cognitive function due to damage or disease in the body beyond what might be expected from normal aging. Based on neuropathological and clinical criteria, dementia includes a spectrum of diseases, namely Alzheimer's dementia, Parkinson's dementia, Lewy Body disease, Alzheimer's dementia with Parkinson's, Pick's disease, Semantic dementia, and large and small vessel disease. It is thought that these disorders result from a combination of genetic and environmental risk factors. Despite accumulating knowledge that has been gained about pathophysiological and clinical characteristics of the disease, no coherent and integrative picture of molecular mechanisms underlying neurodegeneration in Alzheimer’s disease is available. Existing drugs only offer symptomatic relief to the patients and lack any efficient disease-modifying effects. The present research proposes a knowledge-based rationale towards integrative modeling of disease mechanism for identifying potential candidate targets and biomarkers in Alzheimer’s disease. Integrative disease modeling is an emerging knowledge-based paradigm in translational research that exploits the power of computational methods to collect, store, integrate, model and interpret accumulated disease information across different biological scales from molecules to phenotypes. It prepares the ground for transitioning from ‘descriptive’ to “mechanistic” representation of disease processes. The proposed approach was used to introduce an integrative framework, which integrates, on one hand, extracted knowledge from the literature using semantically supported text-mining technologies and, on the other hand, primary experimental data such as gene/protein expression or imaging readouts. The aim of such a hybrid integrative modeling approach was not only to provide a consolidated systems view on the disease mechanism as a whole but also to increase specificity and sensitivity of the mechanistic model by providing disease-specific context. This approach was successfully used for correlating clinical manifestations of the disease to their corresponding molecular events and led to the identification and modeling of three important mechanistic components underlying Alzheimer’s dementia, namely the CNS, the immune system and the endocrine components. These models were validated using a novel in-silico validation method, namely biomarker-guided pathway analysis and a pathway-based target identification approach was introduced, which resulted in the identification of the MAPK signaling pathway as a potential candidate target at the crossroad of the triad components underlying disease mechanism in Alzheimer’s dementia

    MildInt: Deep Learning-Based Multimodal Longitudinal Data Integration Framework

    Get PDF
    As large amounts of heterogeneous biomedical data become available, numerous methods for integrating such datasets have been developed to extract complementary knowledge from multiple domains of sources. Recently, a deep learning approach has shown promising results in a variety of research areas. However, applying the deep learning approach requires expertise for constructing a deep architecture that can take multimodal longitudinal data. Thus, in this paper, a deep learning-based python package for data integration is developed. The python package deep learning-based multimodal longitudinal data integration framework (MildInt) provides the preconstructed deep learning architecture for a classification task. MildInt contains two learning phases: learning feature representation from each modality of data and training a classifier for the final decision. Adopting deep architecture in the first phase leads to learning more task-relevant feature representation than a linear model. In the second phase, linear regression classifier is used for detecting and investigating biomarkers from multimodal data. Thus, by combining the linear model and the deep learning model, higher accuracy and better interpretability can be achieved. We validated the performance of our package using simulation data and real data. For the real data, as a pilot study, we used clinical and multimodal neuroimaging datasets in Alzheimer's disease to predict the disease progression. MildInt is capable of integrating multiple forms of numerical data including time series and non-time series data for extracting complementary features from the multimodal dataset

    Multimodal and multiscale brain networks : understanding aging, Alzheimer’s disease, and other neurodegenerative disorders

    Get PDF
    The human brain can be modeled as a complex network, often referred to as the connectome, where structural and functional connections govern its organization. Several neuroimaging studies have focused on understanding the architecture of healthy brain networks and have shed light on how these networks evolve with age and in the presence of neurodegenerative disorders. Many studies have explored the brain networks in Alzheimer’s disease (AD), the most common type of dementia, using various neuroimaging modalities independently. However, most of these studies ignored the complex and multifactorial nature of AD. The aim of this thesis was to investigate and analyze the brain’s multimodal and multiscale network organization in aging and in AD by using different multilayer brain network analyses and different types of data. Additionally, this research extended its scope to incorporate other dementias, such as Lewy body dementias, allowing for a comparison of these disorders with AD and normal aging. These comparisons were made possible through the application of protein co-expression networks. In Study I, we investigated sex differences in healthy individuals using multimodal brain networks. To do this we used resting-state functional magnetic resonance imaging (rs-fMRI) and diffusion-weighted imaging (DWI) data from the Human Connectome Project (HCP) to perform multilayer and deep learning analyses. These analyses identified differences between men's and women's underlying brain network organization, showing that the deep-learning analysis with multilayer network metrics (area under the curve, AUC, of 0.81) outperforms the classification using single-layer network measures (AUC of 0.72 for functional networks and 0.70 for anatomical networks). Furthermore, we integrated the multilayer brain networks methodology and neural network models into a software package that is easy to use by researchers with different backgrounds and is also easily expandable for researchers with different levels of programming experience. Then, we used the multilayer brain networks methodology to study the interaction between sex and age on the functional network topology using a large group of people from the UK Biobank (Study II). By incorporating multilayer brain network analyses, we analyzed both positive and negative connections derived from functional correlations, and we obtained important insights into how cognitive abilities, physical health, and even genetic factors differ between men and women as they age. Age and sex were strongly associated with multiplex and multilayer measures such as the multiplex participation coefficient, multilayer clustering, and multilayer global efficiency, accounting for up to 89.1%, 79.9%, and 79.5% of the variance related to age, respectively. These results indicate that incorporating separate layers for positive and negative connections within a complex network framework reveals sensitive insights into age- and sex-related variations that are not detected by traditional metrics. Furthermore, our functional metrics exhibited associations with genes that have previously been linked to processes related to aging. In Study III, we assessed whether multilayer connectome analyses could offer new perspectives on the relationship between amyloid pathology and gray matter atrophy across the AD continuum. Subjects from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) were divided into four groups based on cerebrospinal fluid (CSF) amyloid-β (Aβ) biomarker levels and clinical diagnosis. We compared the different groups using weighted and binary multilayer measures that assess the strength of the connections, the modularity, as well as the multiplex segregation and integration of the brain connectomes. Across Aβ-positive (Aβ+) groups, we found widespread increases in the overlapping connectivity strength and decreases in the number of identical connections in both layers. Moreover, the brain modules were reorganized in the mild cognitive impairment (MCI) Aβ+ group and an imbalance in the quantity of couplings between the two layers was found in patients with MCI Aβ+ and AD Aβ+. Using a subsample from the same database, ADNI, we analyzed rs-fMRI data from individuals at preclinical and clinical stages of AD (Study IV). By dividing the time series into different time windows, we built temporal multilayer networks and studied the modular organization across time. We were able to capture the dynamic changes across different AD stages using this temporal multilayer network approach, obtaining outstanding areas under the curve of 0.90, 0.92 and 0.99 in the distinction of controls from preclinical, prodromal, and clinical AD stages, respectively, on top and beyond common risk factors. Our results not only improved the discrimination between various disease stages but, importantly, they also showed that dynamic multilayer functional measures are associated with memory and global cognition in addition to amyloid and tau load derived from positron emission tomography. These results highlight the potential of dynamic multilayer functional connectivity measures as functional biomarkers of AD progression. In Study V, we used in-depth quantitative proteomics to compare post-mortem brains from three key brain regions (prefrontal cortex, cingulate cortex, and the parietal cortex) directly related to the disease mechanisms of AD, Parkinson’s disease with dementia (PDD), dementia with Lewy bodies (DLB) in prospectively followed patients and older adults without dementia. We used covariance weighted networks to find modules of protein sets to further understand altered pathways in these dementias and their implications for prognostic and diagnostic purposes. In conclusion, this thesis explored the complex world of brain networks and offered insightful information about how age, sex, and AD influence these networks. We have improved our understanding of how the brain is organized in different imaging modalities and different time scales, as well as developing software tools to make this methodology available to more researchers. Additionally, we assessed the connections among various proteins in different areas of the brain in relation to health, Alzheimer's disease, and Lewy body dementias. This work contributes to the collective effort of unraveling the mysteries of the human brain organization and offers a foundation for future research to understand brain networks in health and disease

    Novel Deep Learning Models for Medical Imaging Analysis

    Get PDF
    abstract: Deep learning is a sub-field of machine learning in which models are developed to imitate the workings of the human brain in processing data and creating patterns for decision making. This dissertation is focused on developing deep learning models for medical imaging analysis of different modalities for different tasks including detection, segmentation and classification. Imaging modalities including digital mammography (DM), magnetic resonance imaging (MRI), positron emission tomography (PET) and computed tomography (CT) are studied in the dissertation for various medical applications. The first phase of the research is to develop a novel shallow-deep convolutional neural network (SD-CNN) model for improved breast cancer diagnosis. This model takes one type of medical image as input and synthesizes different modalities for additional feature sources; both original image and synthetic image are used for feature generation. This proposed architecture is validated in the application of breast cancer diagnosis and proved to be outperforming the competing models. Motivated by the success from the first phase, the second phase focuses on improving medical imaging synthesis performance with advanced deep learning architecture. A new architecture named deep residual inception encoder-decoder network (RIED-Net) is proposed. RIED-Net has the advantages of preserving pixel-level information and cross-modality feature transferring. The applicability of RIED-Net is validated in breast cancer diagnosis and Alzheimer’s disease (AD) staging. Recognizing medical imaging research often has multiples inter-related tasks, namely, detection, segmentation and classification, my third phase of the research is to develop a multi-task deep learning model. Specifically, a feature transfer enabled multi-task deep learning model (FT-MTL-Net) is proposed to transfer high-resolution features from segmentation task to low-resolution feature-based classification task. The application of FT-MTL-Net on breast cancer detection, segmentation and classification using DM images is studied. As a continuing effort on exploring the transfer learning in deep models for medical application, the last phase is to develop a deep learning model for both feature transfer and knowledge from pre-training age prediction task to new domain of Mild cognitive impairment (MCI) to AD conversion prediction task. It is validated in the application of predicting MCI patients’ conversion to AD with 3D MRI images.Dissertation/ThesisDoctoral Dissertation Industrial Engineering 201

    Levetiracetam modulates brain metabolic networks and transcriptomic signatures in the 5XFAD mouse model of Alzheimer’s disease

    Get PDF
    IntroductionSubcritical epileptiform activity is associated with impaired cognitive function and is commonly seen in patients with Alzheimer’s disease (AD). The anti-convulsant, levetiracetam (LEV), is currently being evaluated in clinical trials for its ability to reduce epileptiform activity and improve cognitive function in AD. The purpose of the current study was to apply pharmacokinetics (PK), network analysis of medical imaging, gene transcriptomics, and PK/PD modeling to a cohort of amyloidogenic mice to establish how LEV restores or drives alterations in the brain networks of mice in a dose-dependent basis using the rigorous preclinical pipeline of the MODEL-AD Preclinical Testing Core.MethodsChronic LEV was administered to 5XFAD mice of both sexes for 3 months based on allometrically scaled clinical dose levels from PK models. Data collection and analysis consisted of a multi-modal approach utilizing 18F-FDG PET/MRI imaging and analysis, transcriptomic analyses, and PK/PD modeling.ResultsPharmacokinetics of LEV showed a sex and dose dependence in Cmax, CL/F, and AUC0-∞, with simulations used to estimate dose regimens. Chronic dosing at 10, 30, and 56 mg/kg, showed 18F-FDG specific regional differences in brain uptake, and in whole brain covariance measures such as clustering coefficient, degree, network density, and connection strength (i.e., positive and negative). In addition, transcriptomic analysis via nanoString showed dose-dependent changes in gene expression in pathways consistent 18F-FDG uptake and network changes, and PK/PD modeling showed a concentration dependence for key genes, but not for network covariance modeling.DiscussionThis study represents the first report detailing the relationships of metabolic covariance and transcriptomic network changes resulting from LEV administration in 5XFAD mice. Overall, our results highlight non-linear kinetics based on dose and sex, where gene expression analysis demonstrated LEV dose- and concentration-dependent changes, along with cerebral metabolism, and/or cerebral homeostatic mechanisms relevant to human AD, which aligned closely with network covariance analysis of 18F-FDG images. Collectively, this study show cases the value of a multimodal connectomic, transcriptomic, and pharmacokinetic approach to further investigate dose dependent relationships in preclinical studies, with translational value toward informing clinical study design

    Genome-wide association study of language performance in Alzheimer's disease

    Get PDF
    Language impairment is common in prodromal stages of Alzheimer's disease (AD) and progresses over time. However, the genetic architecture underlying language performance is poorly understood. To identify novel genetic variants associated with language performance, we analyzed brain MRI and performed a genome-wide association study (GWAS) using a composite measure of language performance from the Alzheimer's Disease Neuroimaging Initiative (ADNI; n=1560). The language composite score was associated with brain atrophy on MRI in language and semantic areas. GWAS identified GLI3 (GLI family zinc finger 3) as significantly associated with language performance (p<5×10-8). Enrichment of GWAS association was identified in pathways related to nervous system development and glutamate receptor function and trafficking. Our results, which warrant further investigation in independent and larger cohorts, implicate GLI3, a developmental transcription factor involved in patterning brain structures, as a putative gene associated with language dysfunction in AD

    Olfactory deficit: a potential functional marker across the Alzheimer’s disease continuum

    Get PDF
    Alzheimer’s disease (AD) is a prevalent form of dementia that affects an estimated 32 million individuals globally. Identifying early indicators is vital for screening at-risk populations and implementing timely interventions. At present, there is an urgent need for early and sensitive biomarkers to screen individuals at risk of AD. Among all sensory biomarkers, olfaction is currently one of the most promising indicators for AD. Olfactory dysfunction signifies a decline in the ability to detect, identify, or remember odors. Within the spectrum of AD, impairment in olfactory identification precedes detectable cognitive impairments, including mild cognitive impairment (MCI) and even the stage of subjective cognitive decline (SCD), by several years. Olfactory impairment is closely linked to the clinical symptoms and neuropathological biomarkers of AD, accompanied by significant structural and functional abnormalities in the brain. Olfactory behavior examination can subjectively evaluate the abilities of olfactory identification, threshold, and discrimination. Olfactory functional magnetic resonance imaging (fMRI) can provide a relatively objective assessment of olfactory capabilities, with the potential to become a promising tool for exploring the neural mechanisms of olfactory damage in AD. Here, we provide a timely review of recent literature on the characteristics, neuropathology, and examination of olfactory dysfunction in the AD continuum. We focus on the early changes in olfactory indicators detected by behavioral and fMRI assessments and discuss the potential of these techniques in MCI and preclinical AD. Despite the challenges and limitations of existing research, olfactory dysfunction has demonstrated its value in assessing neurodegenerative diseases and may serve as an early indicator of AD in the future

    Data-independent acquisition proteomics of cerebrospinal fluid implicates endoplasmic reticulum and inflammatory mechanisms in amyotrophic lateral sclerosis

    Get PDF
    While unbiased proteomics of human cerebrospinal fluid (CSF) has been used successfully to identify biomarkers of amyotrophic lateral sclerosis (ALS), high-abundance proteins mask the presence of lower abundance proteins that may have diagnostic and prognostic value. However, developments in mass spectrometry (MS) proteomic data acquisition methods offer improved protein depth. In this study, MS with library-free data-independent acquisition (DIA) was used to compare the CSF proteome of people with ALS (n = 40), healthy (n = 15) and disease (n = 8) controls. Quantified protein groups were subsequently correlated with clinical variables. Univariate analysis identified 7 proteins, all significantly upregulated in ALS versus healthy controls, and 9 with altered abundance in ALS versus disease controls (FDR < 0.1). Elevated chitotriosidase-1 (CHIT1) was common to both comparisons and was proportional to ALS disability progression rate (Pearson r = 0.41, FDR-adjusted p = 0.035) but not overall survival. Ubiquitin carboxyl-terminal hydrolase isozyme L1 (UCHL1; upregulated in ALS versus healthy controls) was proportional to disability progression rate (Pearson r = 0.53, FDR-adjusted p = 0.003) and survival (Kaplan Meier log-rank p = 0.013) but not independently in multivariate proportional hazards models. Weighted correlation network analysis was used to identify functionally relevant modules of proteins. One module, enriched for inflammatory functions, was associated with age at symptom onset (Pearson r = 0.58, FDR-adjusted p = 0.005) and survival (Hazard Ratio = 1.78, FDR = 0.065), and a second module, enriched for endoplasmic reticulum proteins, was negatively correlated with disability progression rate (r = −0.42, FDR-adjusted p = 0.109). DIA acquisition methodology therefore strengthened the biomarker candidacy of CHIT1 and UCHL1 in ALS, while additionally highlighted inflammatory and endoplasmic reticulum proteins as novel sources of prognostic biomarkers

    Cortico-cortical and hippocampal-cortical interactions in mouse models of Alzheimer’s disease

    Get PDF
    Alzheimer’s disease (AD) is a neurodegenerative disease which is pathologically characterized by extracellular deposition of amyloid beta (Aβ) plaques, intracellular deposition of neurofibrillary tangles (NFT) caused by hyperphosphorylated tau protein, neuroinflammation, and progressive neuron loss. Brain regions involved in memory processing, such as hippocampus and the neocortex, are affected in the early stages of disease pathology. Using in vivo mesoscale wide-field voltage imaging and local field potential (LFP) recording from CA1 region of the hippocampus in 6- and 12-month-old (1) knock-in (AppNL-G-F) and (2) transgenic (5xFAD) mouse model of AD, this study is aimed at understanding how cortico-cortical and hippocampal-cortical interactions are affected by AD. Aberrant sensory evoked cortical activity and resting state cortical functional connectivity were observed in AD and sharp wave ripples (SWRs), which subserve important aspects of hippocampal-cortical interactions are disrupted in AD. Further, gradual cerebral hypoperfusion exacerbate AD pathology and network dysfunctions
    corecore