616 research outputs found

    A powerful and efficient multivariate approach for voxel-level connectome-wide association studies

    Get PDF
    We describe an approach to multivariate analysis, termed structured kernel principal component regression (sKPCR), to identify associations in voxel-level connectomes using resting-state functional magnetic resonance imaging (rsfMRI) data. This powerful and computationally efficient multivariate method can identify voxel-phenotype associations based on the whole-brain connectivity pattern of voxels, and it can detect linear and non-linear signals in both volume-based and surface-based rsfMRI data. For each voxel, sKPCR first extracts low-dimensional signals from the spatially smoothed connectivities by structured kernel principal component analysis, and then tests the voxel-phenotype associations by an adaptive regression model. The method's power is derived from appropriately modelling the spatial structure of the data when performing dimension reduction, and then adaptively choosing an optimal dimension for association testing using the adaptive regression strategy. Simulations based on real connectome data have shown that sKPCR can accurately control the false-positive rate and that it is more powerful than many state-of-the-art approaches, such as the connectivity-wise generalized linear model (GLM) approach, multivariate distance matrix regression (MDMR), adaptive sum of powered score (aSPU) test, and least-square kernel machine (LSKM). Moreover, since sKPCR can reduce the computational cost of non-parametric permutation tests, its computation speed is much faster. To demonstrate the utility of sKPCR for real data analysis, we have also compared sKPCR with the above methods based on the identification of voxel-wise differences between schizophrenic patients and healthy controls in four independent rsfMRI datasets. The results showed that sKPCR had better between-sites reproducibility and a larger proportion of overlap with existing schizophrenia meta-analysis findings. Code for our approach can be downloaded from https://github.com/weikanggong/sKPCR. [Abstract copyright: Copyright © 2018 Elsevier Inc. All rights reserved.

    Learning and comparing functional connectomes across subjects

    Get PDF
    Functional connectomes capture brain interactions via synchronized fluctuations in the functional magnetic resonance imaging signal. If measured during rest, they map the intrinsic functional architecture of the brain. With task-driven experiments they represent integration mechanisms between specialized brain areas. Analyzing their variability across subjects and conditions can reveal markers of brain pathologies and mechanisms underlying cognition. Methods of estimating functional connectomes from the imaging signal have undergone rapid developments and the literature is full of diverse strategies for comparing them. This review aims to clarify links across functional-connectivity methods as well as to expose different steps to perform a group study of functional connectomes

    Multimodal population brain imaging in the UK Biobank prospective epidemiological study

    Get PDF
    Medical imaging has enormous potential for early disease prediction, but is impeded by the difficulty and expense of acquiring data sets before symptom onset. UK Biobank aims to address this problem directly by acquiring high-quality, consistently acquired imaging data from 100,000 predominantly healthy participants, with health outcomes being tracked over the coming decades. The brain imaging includes structural, diffusion and functional modalities. Along with body and cardiac imaging, genetics, lifestyle measures, biological phenotyping and health records, this imaging is expected to enable discovery of imaging markers of a broad range of diseases at their earliest stages, as well as provide unique insight into disease mechanisms. We describe UK Biobank brain imaging and present results derived from the first 5,000 participants' data release. Although this covers just 5% of the ultimate cohort, it has already yielded a rich range of associations between brain imaging and other measures collected by UK Biobank

    Graph analysis of functional brain networks: practical issues in translational neuroscience

    Full text link
    The brain can be regarded as a network: a connected system where nodes, or units, represent different specialized regions and links, or connections, represent communication pathways. From a functional perspective communication is coded by temporal dependence between the activities of different brain areas. In the last decade, the abstract representation of the brain as a graph has allowed to visualize functional brain networks and describe their non-trivial topological properties in a compact and objective way. Nowadays, the use of graph analysis in translational neuroscience has become essential to quantify brain dysfunctions in terms of aberrant reconfiguration of functional brain networks. Despite its evident impact, graph analysis of functional brain networks is not a simple toolbox that can be blindly applied to brain signals. On the one hand, it requires a know-how of all the methodological steps of the processing pipeline that manipulates the input brain signals and extract the functional network properties. On the other hand, a knowledge of the neural phenomenon under study is required to perform physiological-relevant analysis. The aim of this review is to provide practical indications to make sense of brain network analysis and contrast counterproductive attitudes

    Morphometricity as a measure of the neuroanatomical signature of a trait

    Get PDF
    Complex physiological and behavioral traits, including neurological and psychiatric disorders, often associate with distributed anatomical variation. This paper introduces a global metric, called morphometricity, as a measure of the anatomical signature of different traits. Morphometricity is defined as the proportion of phenotypic variation that can be explained by macroscopic brain morphology. We estimate morphometricity via a linear mixed-effects model that uses an anatomical similarity matrix computed based on measurements derived from structural brain MRI scans. We examined over 3,800 unique MRI scans from nine large-scale studies to estimate the morphometricity of a range of phenotypes, including clinical diagnoses such as Alzheimer’s disease, and nonclinical traits such as measures of cognition. Our results demonstrate that morphometricity can provide novel insights about the neuroanatomical correlates of a diverse set of traits, revealing associations that might not be detectable through traditional statistical techniques.National Institute for Biomedical Imaging and Bioengineering (U.S.) (R01EB006758)National Institute for Biomedical Imaging and Bioengineering (U.S.) (P41EB015896)National Institute for Biomedical Imaging and Bioengineering (U.S.) (R21EB018907)National Institute for Biomedical Imaging and Bioengineering (U.S.) (R01EB019956)National Institute on Aging (5R01AG008122)National Institute on Aging (R01AG016495)National Institute of Neurological Diseases and Stroke (U.S.) (R01NS0525851)National Institute of Neurological Diseases and Stroke (U.S.) (R21NS072652)National Institute of Neurological Diseases and Stroke (U.S.) (R01NS070963)National Institute of Neurological Diseases and Stroke (U.S.) (R01NS083534)National Institute of Neurological Diseases and Stroke (U.S.) (5U01NS086625)United States. National Institutes of Health (5U01-MH093765)United States. National Institutes of Health (R01NS083534)United States. National Institutes of Health (R01NS070963)United States. National Institutes of Health (R41AG052246)United States. National Institutes of Health (1K25EB013649-01

    NEW STATISTICAL METHODS FOR HIGH-DIMENSIONAL DATA WITH COMPLEX STRUCTURES

    Get PDF
    The overwhelming advances in biomedical technology facilitate the availability of high-dimensional biomedical data with complex and organized structures. However, due to the obscured true signals by substantial false-positive noises and the high dimensionality, the statistical inference is challenging with the critical issue of research reproducibility and replicability. Hence, motivated by these urgent needs, this dissertation is devoted to statistical approaches in understanding the latent structures among biomedical objects, as well as improving statistical power and reducing false-positive errors in statistical inference. The first objective of this dissertation is motivated by the group-level brain connectome analysis in neuropsychiatric research with the goal of exhibiting the connectivity abnormality between clinical groups. In Chapter 2, we develop a likelihood-based adaptive dense subgraph discovery (ADSD) procedure to identify connectomic subnetworks (subgraphs) that are systematically associated with brain disorders. We propose the statistical inference procedure leveraging graph properties and combinatorics. We validate the proposed method by a brain fMRI study for schizophrenia research and synthetic data under various settings. In Chapter 3, we are interested in assessing the genetic effects on brain structural imaging with spatial specificity. In contrast to the inference on individual SNP-voxel pairs, we focus on the systematic associations between genetic and imaging measurements, which assists the understanding of a polygenic and pleiotropic association structure. Based on voxel-wise genome-wide association analysis (vGWAS), we characterize the polygenic and pleiotropic SNP-voxel association structure using imaging-genetics dense bi-cliques (IGDBs). We develop the estimation procedure and statistical inference framework on the IGDBs with computationally efficient algorithms. We demonstrate the performance of the proposed approach using imaging-genetics data from the human connectome project (HCP). Chapter 4 carries the analysis of gene co-expression network (GCN) in examining the gene-gene interactions and learning the underlying complex yet highly organized gene regulatory mechanisms. We propose the interconnected community network (ICN) structure that allows the interactions between genes from different communities, which relaxes the constraint of most existing GCN analysis approaches. We develop a computational package to detect the ICN structure based on graph norm shrinkage. The application of ICN detection is illustrated using an RNA-seq data from The Cancer Genome Atlas (TCGA) Acute Myeloid Leukemia (AML) study
    corecore