1
|
Chung J, Kim S, Won JH, Park H. Integrating Multimodal Neuroimaging and Genetics: A Structurally-Linked Sparse Canonical Correlation Analysis Approach. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE 2024; 12:659-667. [PMID: 39464624 PMCID: PMC11505868 DOI: 10.1109/jtehm.2024.3463720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/16/2024] [Accepted: 09/14/2024] [Indexed: 10/29/2024]
Abstract
Neuroimaging genetics represents a multivariate approach aimed at elucidating the intricate relationships between high-dimensional genetic variations and neuroimaging data. Predominantly, existing methodologies revolve around Sparse Canonical Correlation Analysis (SCCA), a framework we expand to 1) encompass multiple imaging modalities and 2) promote the simultaneous identification of structurally linked features across imaging modalities. The structurally linked brain regions were assessed using diffusion tensor imaging, which quantifies the presence of neuronal fibers, thereby grounding our approach in biologically well-founded prior knowledge within the SCCA model. In our proposed structurally linked SCCA framework, we leverage T1-weighted MRI and functional MRI (fMRI) time series data to delineate both the structural and functional characteristics of the brain. Genetic variations, specifically single nucleotide polymorphisms (SNPs), are also incorporated as a genetic modality. Validation of our methodology was conducted using a simulated dataset and large-scale normative data from the Human Connectome Project (HCP). Our approach demonstrated superior performance compared to existing methods on simulated data and revealed interpretable gene-imaging associations in the real dataset. Thus, our methodology lays the groundwork for elucidating the genetic underpinnings of brain structure and function, thereby providing novel insights into the field of neuroscience. Our code is available at https://github.com/mungegg.
Collapse
Affiliation(s)
- Jiwon Chung
- Department of Electrical and Computer EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
| | - Sunghun Kim
- Department of Electrical and Computer EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
| | - Ji Hye Won
- Department of Computer Engineering and Artificial IntelligencePukyong National UniversityBusan48513Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
- Center for Neuroscience Imaging ResearchInstitute for Basic ScienceSuwon16419Republic of Korea
| |
Collapse
|
2
|
Bi XA, Wang Y, Luo S, Chen K, Xing Z, Xu L. Hypergraph Structural Information Aggregation Generative Adversarial Networks for Diagnosis and Pathogenetic Factors Identification of Alzheimer's Disease With Imaging Genetic Data. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2024; 35:7420-7434. [PMID: 36264725 DOI: 10.1109/tnnls.2022.3212700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with profound pathogenetic causes. Imaging genetic data analysis can provide comprehensive insights into its causes. To fully utilize the multi-level information in the data, this article proposes a hypergraph structural information aggregation model, and constructs a novel deep learning method named hypergraph structural information aggregation generative adversarial networks (HSIA-GANs) for the automatic sample classification and accurate feature extraction. Specifically, HSIA-GAN is composed of generator and discriminator. The generator has three main functions. First, vertex graph and edge graph are constructed based on the input hypergraph to present the low-order relations. Second, the low-order structural information of hypergraph is extracted by the designed vertex convolution layers and edge convolution layers. Finally, the synthetic hypergraph is generated as the input of the discriminator. The discriminator can extract the high-order structural information directly from hypergraph through vertex-edge convolution, fuse the high and low-order structural information, and finalize the results through the full connection (FC) layers. Based on the data acquired from AD neuroimaging initiative, HSIA-GAN shows significant advantages in three classification tasks, and extracts discriminant features conducive to better disease classification.
Collapse
|
3
|
Rashid MM, Selvarajoo K. Advancing drug-response prediction using multi-modal and -omics machine learning integration (MOMLIN): a case study on breast cancer clinical data. Brief Bioinform 2024; 25:bbae300. [PMID: 38904542 PMCID: PMC11190965 DOI: 10.1093/bib/bbae300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
The inherent heterogeneity of cancer contributes to highly variable responses to any anticancer treatments. This underscores the need to first identify precise biomarkers through complex multi-omics datasets that are now available. Although much research has focused on this aspect, identifying biomarkers associated with distinct drug responders still remains a major challenge. Here, we develop MOMLIN, a multi-modal and -omics machine learning integration framework, to enhance drug-response prediction. MOMLIN jointly utilizes sparse correlation algorithms and class-specific feature selection algorithms, which identifies multi-modal and -omics-associated interpretable components. MOMLIN was applied to 147 patients' breast cancer datasets (clinical, mutation, gene expression, tumor microenvironment cells and molecular pathways) to analyze drug-response class predictions for non-responders and variable responders. Notably, MOMLIN achieves an average AUC of 0.989, which is at least 10% greater when compared with current state-of-the-art (data integration analysis for biomarker discovery using latent components, multi-omics factor analysis, sparse canonical correlation analysis). Moreover, MOMLIN not only detects known individual biomarkers such as genes at mutation/expression level, most importantly, it correlates multi-modal and -omics network biomarkers for each response class. For example, an interaction between ER-negative-HMCN1-COL5A1 mutations-FBXO2-CSF3R expression-CD8 emerge as a multimodal biomarker for responders, potentially affecting antimicrobial peptides and FLT3 signaling pathways. In contrast, for resistance cases, a distinct combination of lymph node-TP53 mutation-PON3-ENSG00000261116 lncRNA expression-HLA-E-T-cell exclusions emerged as multimodal biomarkers, possibly impacting neurotransmitter release cycle pathway. MOMLIN, therefore, is expected advance precision medicine, such as to detect context-specific multi-omics network biomarkers and better predict drug-response classifications.
Collapse
Affiliation(s)
- Md Mamunur Rashid
- Biomolecular Sequence to Function Division, BII, (ASTAR), Singapore 138671, Republic of Singapore
| | - Kumar Selvarajoo
- Biomolecular Sequence to Function Division, BII, (ASTAR), Singapore 138671, Republic of Singapore
- Synthetic Biology Translational Research Program, Yong Loo Lin School of Medicine, NUS, Singapore 117456, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University (NTU), Singapore 639798, Republic of Singapore
| |
Collapse
|
4
|
Cruciani F, Aparo A, Brusini L, Combi C, Storti SF, Giugno R, Menegaz G, Boscolo Galazzo I. Identifying the joint signature of brain atrophy and gene variant scores in Alzheimer's Disease. J Biomed Inform 2024; 149:104569. [PMID: 38104851 DOI: 10.1016/j.jbi.2023.104569] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The joint modeling of genetic data and brain imaging information allows for determining the pathophysiological pathways of neurodegenerative diseases such as Alzheimer's disease (AD). This task has typically been approached using mass-univariate methods that rely on a complete set of Single Nucleotide Polymorphisms (SNPs) to assess their association with selected image-derived phenotypes (IDPs). However, such methods are prone to multiple comparisons bias and, most importantly, fail to account for potential cross-feature interactions, resulting in insufficient detection of significant associations. Ways to overcome these limitations while reducing the number of traits aim at conveying genetic information at the gene level and capturing the integrated genetic effects of a set of genetic variants, rather than looking at each SNP individually. Their associations with brain IDPs are still largely unexplored in the current literature, though they can uncover new potential genetic determinants for brain modulations in the AD continuum. In this work, we explored an explainable multivariate model to analyze the genetic basis of the grey matter modulations, relying on the AD Neuroimaging Initiative (ADNI) phase 3 dataset. Cortical thicknesses and subcortical volumes derived from T1-weighted Magnetic Resonance were considered to describe the imaging phenotypes. At the same time the genetic counterpart was represented by gene variant scores extracted by the Sequence Kernel Association Test (SKAT) filtering model. Moreover, transcriptomic analysis was carried on to assess the expression of the resulting genes in the main brain structures as a form of validation. Results highlighted meaningful genotype-phenotype interactionsas defined by three latent components showing a significant difference in the projection scores between patients and controls. Among the significant associations, the model highlighted EPHX1 and BCAS1 gene variant scores involved in neurodegenerative and myelination processes, hence relevant for AD. In particular, the first was associated with decreased subcortical volumes and the second with decreasedtemporal lobe thickness. Noteworthy, BCAS1 is particularly expressed in the dentate gyrus. Overall, the proposed approach allowed capturing genotype-phenotype interactions in a restricted study cohort that was confirmed by transcriptomic analysis, offering insights into the underlying mechanisms of neurodegeneration in AD in line with previous findings and suggesting new potential disease biomarkers.
Collapse
Affiliation(s)
- Federica Cruciani
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
| | - Antonino Aparo
- Department of Computer Science, University of Verona, Verona, Italy
| | - Lorenza Brusini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Carlo Combi
- Department of Computer Science, University of Verona, Verona, Italy
| | - Silvia F Storti
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Verona, Italy
| | - Gloria Menegaz
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | | |
Collapse
|
5
|
Wang M, Shao W, Huang S, Zhang D. Hypergraph-regularized multimodal learning by graph diffusion for imaging genetics based Alzheimer's Disease diagnosis. Med Image Anal 2023; 89:102883. [PMID: 37467641 DOI: 10.1016/j.media.2023.102883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/06/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023]
Abstract
Recent studies show that multi-modal data fusion techniques combining information from diverse sources are helpful to diagnose and predict complex brain disorders. However, most existing diagnosis methods have only simply employed a feature combination strategy for multiple imaging and genetic data, ignoring the imaging phenotypes associated with the risk gene information. To this end, we present a hypergraph-regularized multimodal learning by graph diffusion (HMGD) for joint association learning and outcome prediction. Specifically, we first present a graph diffusion method for enhancing similarity measures among subjects given from multi-modality phenotypes, which fully uses multiple input similarity graphs and integrates them into a unified graph with valuable geometric structures among different imaging phenotypes. Then, we employ the unified graph to represent the high-order similarity relationships among subjects, and enforce a hypergraph-regularized term to incorporate both inter- and cross-modality information for selecting the imaging phenotypes associated with the risk single nucleotide polymorphism (SNP). Finally, a multi-kernel support vector machine (MK-SVM) is adopted to fuse such phenotypic features selected from different modalities for the final diagnosis and prediction. The proposed approach is experimentally explored on brain imaging genetic data of the Alzheimer's Disease Neuroimaging Initiative (ADNI) datasets. Relevant results present that the proposed approach is superior to several competing algorithms, and realizes strong associations and discovers significant consistent and robust ROIs across different imaging phenotypes associated with the genetic risk biomarkers to guide disease interpretation and prediction.
Collapse
Affiliation(s)
- Meiling Wang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China; MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China; Key Laboratory of Brain-Machine Intelligence Technology, Ministry of Education, Nanjing 211106, China
| | - Wei Shao
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China; MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China; Key Laboratory of Brain-Machine Intelligence Technology, Ministry of Education, Nanjing 211106, China
| | - Shuo Huang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China; MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China; Key Laboratory of Brain-Machine Intelligence Technology, Ministry of Education, Nanjing 211106, China
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China; MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China; Key Laboratory of Brain-Machine Intelligence Technology, Ministry of Education, Nanjing 211106, China.
| |
Collapse
|
6
|
Zhang X, Hao Y, Zhang J, Ji Y, Zou S, Zhao S, Xie S, Du L. A multi-task SCCA method for brain imaging genetics and its application in neurodegenerative diseases. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 232:107450. [PMID: 36905750 DOI: 10.1016/j.cmpb.2023.107450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND AND OBJECTIVES In brain imaging genetics, multi-task sparse canonical correlation analysis (MTSCCA) is effective to study the bi-multivariate associations between genetic variations such as single nucleotide polymorphisms (SNPs) and multi-modal imaging quantitative traits (QTs). However, most existing MTSCCA methods are neither supervised nor capable of distinguishing the shared patterns of multi-modal imaging QTs from the specific patterns. METHODS A new diagnosis-guided MTSCCA (DDG-MTSCCA) with parameter decomposition and graph-guided pairwise group lasso penalty was proposed. Specifically, the multi-tasking modeling paradigm enables us to comprehensively identify risk genetic loci by jointly incorporating multi-modal imaging QTs. The regression sub-task was raised to guide the selection of diagnosis-related imaging QTs. To reveal the diverse genetic mechanisms, the parameter decomposition and different constraints were utilized to facilitate the identification of modality-consistent and -specific genotypic variations. Besides, a network constraint was added to find out meaningful brain networks. The proposed method was applied to synthetic data and two real neuroimaging data sets respectively from Alzheimer's disease neuroimaging initiative (ADNI) and Parkinson's progression marker initiative (PPMI) databases. RESULTS Compared with the competitive methods, the proposed method exhibited higher or comparable canonical correlation coefficients (CCCs) and better feature selection results. In particular, in the simulation study, DDG-MTSCCA showed the best anti-noise ability and achieved the highest average hit rate, about 25% higher than MTSCCA. On the real data of Alzheimer's disease (AD) and Parkinson's disease (PD), our method obtained the highest average testing CCCs, about 40% ∼ 50% higher than MTSCCA. Especially, our method could select more comprehensive feature subsets, and the top five SNPs and imaging QTs were all disease-related. The ablation experimental results also demonstrated the significance of each component in the model, i.e., the diagnosis guidance, parameter decomposition, and network constraint. CONCLUSIONS These results on simulated data, ADNI and PPMI cohorts suggested the effectiveness and generalizability of our method in identifying meaningful disease-related markers. DDG-MTSCCA could be a powerful tool in brain imaging genetics, worthy of in-depth study.
Collapse
Affiliation(s)
- Xin Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China
| | - Yipeng Hao
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China
| | - Jin Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China
| | - Yanuo Ji
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China
| | - Shihong Zou
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China
| | - Shijie Zhao
- School of Automation, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China
| | - Songyun Xie
- School of Electronics and Information, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China
| | - Lei Du
- School of Automation, Northwestern Polytechnical University, Xi'an, Shannxi 710072, China.
| |
Collapse
|
7
|
Wang X, Yu G, Yan Z, Wan L, Wang W, Cui L. Lung Cancer Subtype Diagnosis by Fusing Image-Genomics Data and Hybrid Deep Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:512-523. [PMID: 34855599 DOI: 10.1109/tcbb.2021.3132292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Accurate diagnosis of cancer subtypes is crucial for precise treatment, because different cancer subtypes are involved with different pathology and require different therapies. Although deep learning techniques have made great success in computer vision and other fields, they do not work well on Lung cancer subtype diagnosis, due to the distinction of slide images between different cancer subtypes is ambiguous. Furthermore, they often over-fit to high-dimensional genomics data with limited samples, and do not fuse the image and genomics data in a sensible way. In this paper, we propose a hybrid deep network based approach LungDIG for Lung cancer subtype Diagnosis by fusing Image-Genomics data. LungDIG first tiles the tissue slide image into small patches and extracts the patch-level features by fine-tuning an Inception-V3 model. Since the patches may contain some false positives in non-diagnostic regions, it further designs a patch-level feature combination strategy to integrate the extracted patch features and maintain the diversity between different cancer subtypes. At the same time, it extracts the genomics features from Copy Number Variation data by an attention based nonlinear extractor. Next, it fuses the image and genomics features by an attention based multilayer perceptron (MLP) to diagnose cancer subtype. Experiments on TCGA lung cancer data show that LungDIG can not only achieve higher accuracy for cancer subtype diagnosis than state-of-the-art methods, but also have a high authenticity and good interpretability.
Collapse
|
8
|
Bi XA, Mao Y, Luo S, Wu H, Zhang L, Luo X, Xu L. A novel generation adversarial network framework with characteristics aggregation and diffusion for brain disease classification and feature selection. Brief Bioinform 2022; 23:6762742. [PMID: 36259367 DOI: 10.1093/bib/bbac454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/01/2022] [Accepted: 09/23/2022] [Indexed: 12/14/2022] Open
Abstract
Imaging genetics provides unique insights into the pathological studies of complex brain diseases by integrating the characteristics of multi-level medical data. However, most current imaging genetics research performs incomplete data fusion. Also, there is a lack of effective deep learning methods to analyze neuroimaging and genetic data jointly. Therefore, this paper first constructs the brain region-gene networks to intuitively represent the association pattern of pathogenetic factors. Second, a novel feature information aggregation model is constructed to accurately describe the information aggregation process among brain region nodes and gene nodes. Finally, a deep learning method called feature information aggregation and diffusion generative adversarial network (FIAD-GAN) is proposed to efficiently classify samples and select features. We focus on improving the generator with the proposed convolution and deconvolution operations, with which the interpretability of the deep learning framework has been dramatically improved. The experimental results indicate that FIAD-GAN can not only achieve superior results in various disease classification tasks but also extract brain regions and genes closely related to AD. This work provides a novel method for intelligent clinical decisions. The relevant biomedical discoveries provide a reliable reference and technical basis for the clinical diagnosis, treatment and pathological analysis of disease.
Collapse
Affiliation(s)
- Xia-An Bi
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, and College of Information Science and Engineering in Hunan Normal University, Changsha, P.R. China
| | - Yuhua Mao
- Department of Computing, School of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Sheng Luo
- Department of Computing, School of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Hao Wu
- Department of Computing, School of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Lixia Zhang
- School of Information Science and Engineering, Hunan Normal University, Changsha, P.R. China
| | - Xun Luo
- College of Information Science and Engineering in Hunan Normal University, Changsha, P.R. China
| | - Luyun Xu
- College of Business in Hunan Normal University, Changsha, P.R. China
| |
Collapse
|
9
|
Zeng L, Li H, Xiao T, Shen F, Zhong Z. Graph convolutional network with sample and feature weights for Alzheimer’s disease diagnosis. Inf Process Manag 2022. [DOI: 10.1016/j.ipm.2022.102952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
10
|
Song P, Wang Y, Yuan X, Wang S, Song X. Exploring Brain Structural and Functional Biomarkers in Schizophrenia via Brain-Network-Constrained Multi-View SCCA. Front Neurosci 2022; 16:879703. [PMID: 35794950 PMCID: PMC9252525 DOI: 10.3389/fnins.2022.879703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Recent studies have proved that dynamic regional measures extracted from the resting-state functional magnetic resonance imaging, such as the dynamic fractional amplitude of low-frequency fluctuation (d-fALFF), could provide a great insight into brain dynamic characteristics of the schizophrenia. However, the unimodal feature is limited for delineating the complex patterns of brain deficits. Thus, functional and structural imaging data are usually analyzed together for uncovering the neural mechanism of schizophrenia. Investigation of neural function-structure coupling enables to find the potential biomarkers and further helps to understand the biological basis of schizophrenia. Here, a brain-network-constrained multi-view sparse canonical correlation analysis (BN-MSCCA) was proposed to explore the intrinsic associations between brain structure and dynamic brain function. Specifically, the d-fALFF was first acquired based on the sliding window method, whereas the gray matter map was computed based on voxel-based morphometry analysis. Then, the region-of-interest (ROI)-based features were extracted and further selected by performing the multi-view sparse canonical correlation analysis jointly with the diagnosis information. Moreover, the brain-network-based structural constraint was introduced to prompt the detected biomarkers more interpretable. The experiments were conducted on 191 patients with schizophrenia and 191 matched healthy controls. Results showed that the BN-MSCCA could identify the critical ROIs with more sparse canonical weight patterns, which are corresponding to the specific brain networks. These are biologically meaningful findings and could be treated as the potential biomarkers. The proposed method also obtained a higher canonical correlation coefficient for the testing data, which is more consistent with the results on training data, demonstrating its promising capability for the association identification. To demonstrate the effectiveness of the potential clinical applications, the detected biomarkers were further analyzed on a schizophrenia-control classification task and a correlation analysis task. The experimental results showed that our method had a superior performance with a 5-8% increment in accuracy and 6-10% improvement in area under the curve. Furthermore, two of the top-ranked biomarkers were significantly negatively correlated with the positive symptom score of Positive and Negative Syndrome Scale (PANSS). Overall, the proposed method could find the association between brain structure and dynamic brain function, and also help to identify the biological meaningful biomarkers of schizophrenia. The findings enable our further understanding of this disease.
Collapse
Affiliation(s)
- Peilun Song
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Yaping Wang
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Xiuxia Yuan
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China
| | - Shuying Wang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biological Psychiatry International Joint Laboratory of Henan/Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Wang S, Chen H, Kong W, Ke F, Wei K. Identify Biomarkers of Alzheimer's Disease Based on Multi-task Canonical Correlation Analysis and Regression Model. J Mol Neurosci 2022; 72:1749-1763. [PMID: 35698015 DOI: 10.1007/s12031-022-02031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/21/2022] [Indexed: 11/29/2022]
Abstract
Imaging genetics using imaging technology is regarded as a neuroanatomical phenotype to evaluate gene single nucleotide polymorphisms and their effects on the structure and function of different brain regions. It plays a vital role in bridging the initial understanding of the genetic basis of brain structure and dysfunction. Sparse canonical correlation analysis (SCCA) has become a widespread technique in this field because of its powerful ability to identify bivariate relationships and feature selection. Since most traditional SCCA algorithms assume that the input features are independent, this method obviously cannot be used to analyze genetic image data. The MT-SCCA model is unsupervised and cannot identify the genotype-phenotype associations for diagnostic guidance. Meanwhile, a single biological clinical index cannot fully reflect the physiological process of a comprehensive disease. Therefore, it is necessary to find biomarkers that can reflect Alzheimer's disease and physiological functions that can more comprehensively reflect the development of the disease. This article uses a multi-task sparse canonical correlation analysis and regression (MT-SCCAR) model to combine the annual depression level total score (GDSCALE), clinical dementia assessment scale (GLOBAL CDR), functional activity questionnaire (FAQ), and neuropsychiatric Symptom Questionnaire (NPI-Q) in this paper. These four clinical data are used as compensation information and embedded in the algorithm in a linear regression manner. It also reflects its superiority and robustness compared to traditional correlation analysis methods on actual and simulated data. Meanwhile, compared with MT-SCCA, the model utilized in this paper obtains a higher gene-ROI weight and identifies clearer biomarkers, which provides a practical basis for the study of complex human disease pathology.
Collapse
Affiliation(s)
- Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai, 201306, People's Republic of China.
| | - Huiqiu Chen
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai, 201306, People's Republic of China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai, 201306, People's Republic of China
| | - Fengchun Ke
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai, 201306, People's Republic of China
| | - Kai Wei
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai, 201306, People's Republic of China
| |
Collapse
|
12
|
Yu D, Wang L, Kong D, Zhu H. Mapping the Genetic-Imaging-Clinical Pathway with Applications to Alzheimer’s Disease. J Am Stat Assoc 2022; 117:1656-1668. [PMID: 37009529 PMCID: PMC10062702 DOI: 10.1080/01621459.2022.2087658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease is a progressive form of dementia that results in problems with memory, thinking, and behavior. It often starts with abnormal aggregation and deposition of β amyloid and tau, followed by neuronal damage such as atrophy of the hippocampi, leading to Alzheimers Disease (AD). The aim of this paper is to map the genetic-imaging-clinical pathway for AD in order to delineate the genetically-regulated brain changes that drive disease progression based on the Alzheimers Disease Neuroimaging Initiative (ADNI) dataset. We develop a novel two-step approach to delineate the association between high-dimensional 2D hippocampal surface exposures and the Alzheimers Disease Assessment Scale (ADAS) cognitive score, while taking into account the ultra-high dimensional clinical and genetic covariates at baseline. Analysis results suggest that the radial distance of each pixel of both hippocampi is negatively associated with the severity of behavioral deficits conditional on observed clinical and genetic covariates. These associations are stronger in Cornu Ammonis region 1 (CA1) and subiculum subregions compared to Cornu Ammonis region 2 (CA2) and Cornu Ammonis region 3 (CA3) subregions. Supplementary materials for this article, including a standardized description of the materials available for reproducing the work, are available as an online supplement.
Collapse
Affiliation(s)
- Dengdeng Yu
- Department of Mathematics, University of Texas at Arlington
| | - Linbo Wang
- Department of Statistical Sciences, University of Toronto
| | - Dehan Kong
- Department of Statistical Sciences, University of Toronto
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina, Chapel Hill for the Alzheimer’s Disease Neuroimaging Initiative*
| |
Collapse
|
13
|
Wang Y, Fu Y, Luo X. Identification of Pathogenetic Brain Regions via Neuroimaging Data for Diagnosis of Autism Spectrum Disorders. Front Neurosci 2022; 16:900330. [PMID: 35655751 PMCID: PMC9152096 DOI: 10.3389/fnins.2022.900330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a kind of neurodevelopmental disorder that often occurs in children and has a hidden onset. Patients usually have lagged development of communication ability and social behavior and thus suffer an unhealthy physical and mental state. Evidence has indicated that diseases related to ASD have commonalities in brain imaging characteristics. This study aims to study the pathogenesis of ASD based on brain imaging data to locate the ASD-related brain regions. Specifically, we collected the functional magnetic resonance image data of 479 patients with ASD and 478 normal subjects matched in age and gender and used a machine-learning framework named random support vector machine cluster to extract distinctive brain regions from the preprocessed data. According to the experimental results, compared with other existing approaches, the method used in this study can more accurately distinguish patients from normal individuals based on brain imaging data. At the same time, this study found that the development of ASD was highly correlated with certain brain regions, e.g., lingual gyrus, superior frontal gyrus, medial gyrus, insular lobe, and olfactory cortex. This study explores the effectiveness of a novel machine-learning approach in the study of ASD brain imaging and provides a reference brain area for the medical research and clinical treatment of ASD.
Collapse
Affiliation(s)
- Yu Wang
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, China
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
- Hunan Xiangjiang Artificial Intelligence Academy, Changsha, China
| | - Yu Fu
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, China
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
- Hunan Xiangjiang Artificial Intelligence Academy, Changsha, China
- *Correspondence: Yu Fu
| | - Xun Luo
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, China
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
- Hunan Xiangjiang Artificial Intelligence Academy, Changsha, China
| |
Collapse
|
14
|
Xin Y, Sheng J, Miao M, Wang L, Yang Z, Huang H. A review ofimaging genetics in Alzheimer's disease. J Clin Neurosci 2022; 100:155-163. [PMID: 35487021 DOI: 10.1016/j.jocn.2022.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 03/01/2022] [Accepted: 04/15/2022] [Indexed: 01/18/2023]
Abstract
Determining the association between genetic variation and phenotype is a key step to study the mechanism of Alzheimer's disease (AD), laying the foundation for studying drug therapies and biomarkers. AD is the most common type of dementia in the aged population. At present, three early-onset AD genes (APP, PSEN1, PSEN2) and one late-onset AD susceptibility gene apolipoprotein E (APOE) have been determined. However, the pathogenesis of AD remains unknown. Imaging genetics, an emerging interdisciplinary field, is able to reveal the complex mechanisms from the genetic level to human cognition and mental disorders via macroscopic intermediates. This paper reviews methods of establishing genotype-phenotype to explore correlations, including sparse canonical correlation analysis, sparse reduced rank regression, sparse partial least squares and so on. We found that most research work did poorly in supervised learning and exploring the nonlinear relationship between SNP-QT.
Collapse
Affiliation(s)
- Yu Xin
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China
| | - Jinhua Sheng
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China.
| | - Miao Miao
- Beijing Hospital, Beijing 100730, China; National Center of Gerontology, Beijing 100730, China; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Luyun Wang
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China; Hangzhou Vocational & Technical College, Hangzhou, Zhejiang 310018, China
| | - Ze Yang
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China
| | - He Huang
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
15
|
Bi XA, Zhou W, Luo S, Mao Y, Hu X, Zeng B, Xu L. Feature aggregation graph convolutional network based on imaging genetic data for diagnosis and pathogeny identification of Alzheimer's disease. Brief Bioinform 2022; 23:6572662. [PMID: 35453149 DOI: 10.1093/bib/bbac137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 12/30/2022] Open
Abstract
The roles of brain regions activities and gene expressions in the development of Alzheimer's disease (AD) remain unclear. Existing imaging genetic studies usually has the problem of inefficiency and inadequate fusion of data. This study proposes a novel deep learning method to efficiently capture the development pattern of AD. First, we model the interaction between brain regions and genes as node-to-node feature aggregation in a brain region-gene network. Second, we propose a feature aggregation graph convolutional network (FAGCN) to transmit and update the node feature. Compared with the trivial graph convolutional procedure, we replace the input from the adjacency matrix with a weight matrix based on correlation analysis and consider common neighbor similarity to discover broader associations of nodes. Finally, we use a full-gradient saliency graph mechanism to score and extract the pathogenetic brain regions and risk genes. According to the results, FAGCN achieved the best performance among both traditional and cutting-edge methods and extracted AD-related brain regions and genes, providing theoretical and methodological support for the research of related diseases.
Collapse
Affiliation(s)
- Xia-An Bi
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, and the College of Information Science and Engineering in Hunan Normal University, P.R. China
| | - Wenyan Zhou
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Sheng Luo
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Yuhua Mao
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Xi Hu
- College of Information Science and Engineering, Hunan Normal University, Changsha, China
| | - Bin Zeng
- Hunan Youdao Information Technology Co., Ltd, P.R. China
| | - Luyun Xu
- College of Business in Hunan Normal University, P.R. China
| |
Collapse
|
16
|
Wang M, Shao W, Hao X, Huang S, Zhang D. Identify connectome between genotypes and brain network phenotypes via deep self-reconstruction sparse canonical correlation analysis. Bioinformatics 2022; 38:2323-2332. [PMID: 35143604 DOI: 10.1093/bioinformatics/btac074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/21/2022] [Accepted: 02/02/2022] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION As a rising research topic, brain imaging genetics aims to investigate the potential genetic architecture of both brain structure and function. It should be noted that in the brain, not all variations are deservedly caused by genetic effect, and it is generally unknown which imaging phenotypes are promising for genetic analysis. RESULTS In this work, genetic variants (i.e. the single nucleotide polymorphism, SNP) can be correlated with brain networks (i.e. quantitative trait, QT), so that the connectome (including the brain regions and connectivity features) of functional brain networks from the functional magnetic resonance imaging data is identified. Specifically, a connection matrix is firstly constructed, whose upper triangle elements are selected to be connectivity features. Then, the PageRank algorithm is exploited for estimating the importance of different brain regions as the brain region features. Finally, a deep self-reconstruction sparse canonical correlation analysis (DS-SCCA) method is developed for the identification of genetic associations with functional connectivity phenotypic markers. This approach is a regularized, deep extension, scalable multi-SNP-multi-QT method, which is well-suited for applying imaging genetic association analysis to the Alzheimer's Disease Neuroimaging Initiative datasets. It is further optimized by adopting a parametric approach, augmented Lagrange and stochastic gradient descent. Extensive experiments are provided to validate that the DS-SCCA approach realizes strong associations and discovers functional connectivity and brain region phenotypic biomarkers to guide disease interpretation. AVAILABILITY AND IMPLEMENTATION The Matlab code is available at https://github.com/meimeiling/DS-SCCA/tree/main. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Meiling Wang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| | - Wei Shao
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| | - Xiaoke Hao
- School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Shuo Huang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China.,MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing 211106, China
| |
Collapse
|
17
|
Zhang J, Wang H, Zhao Y, Guo L, Du L. Identification of multimodal brain imaging association via a parameter decomposition based sparse multi-view canonical correlation analysis method. BMC Bioinformatics 2022; 23:128. [PMID: 35413798 PMCID: PMC9006414 DOI: 10.1186/s12859-022-04669-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND With the development of noninvasive imaging technology, collecting different imaging measurements of the same brain has become more and more easy. These multimodal imaging data carry complementary information of the same brain, with both specific and shared information being intertwined. Within these multimodal data, it is essential to discriminate the specific information from the shared information since it is of benefit to comprehensively characterize brain diseases. While most existing methods are unqualified, in this paper, we propose a parameter decomposition based sparse multi-view canonical correlation analysis (PDSMCCA) method. PDSMCCA could identify both modality-shared and -specific information of multimodal data, leading to an in-depth understanding of complex pathology of brain disease. RESULTS Compared with the SMCCA method, our method obtains higher correlation coefficients and better canonical weights on both synthetic data and real neuroimaging data. This indicates that, coupled with modality-shared and -specific feature selection, PDSMCCA improves the multi-view association identification and shows meaningful feature selection capability with desirable interpretation. CONCLUSIONS The novel PDSMCCA confirms that the parameter decomposition is a suitable strategy to identify both modality-shared and -specific imaging features. The multimodal association and the diverse information of multimodal imaging data enable us to better understand the brain disease such as Alzheimer's disease.
Collapse
Affiliation(s)
- Jin Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Huiai Wang
- School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Ying Zhao
- School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Lei Guo
- School of Automation, Northwestern Polytechnical University, Xi'an, China
| | - Lei Du
- School of Automation, Northwestern Polytechnical University, Xi'an, China.
| | | |
Collapse
|
18
|
Wang W, Kong W, Wang S, Wei K. Detecting Biomarkers of Alzheimer's Disease Based on Multi-constrained Uncertainty-Aware Adaptive Sparse Multi-view Canonical Correlation Analysis. J Mol Neurosci 2022; 72:841-865. [PMID: 35080765 DOI: 10.1007/s12031-021-01963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/29/2021] [Indexed: 12/01/2022]
Abstract
Image genetics mainly explores the pathogenesis of Alzheimer's disease (AD) by studying the relationship between genetic data (such as SNP, gene expression data, and DNA methylation) and imaging data (such as structural MRI (sMRI), fMRI, and PET). Most of the existing research on brain imaging genomics uses two-way or three-way bi-multivariate methods to explore the correlation analysis between genes and brain imaging. However, many of these methods are still affected by the gradient domination or cannot take into account the effect of feature redundancy on the results, so that the typical correlation coefficient and program running speed are not significantly improved. In order to solve the above problems, this paper proposes a multi-constrained uncertainty-aware adaptive sparse multi-view canonical correlation analysis method (MC-unAdaSMCCA) to explore associations among SNPs, gene expression data, and sMRI; that is, based on traditional unAdaSMCCA, orthogonal constraints are imposed on the weights of the three data features through linear programming, which can reduce the redundancy of feature weights to improve the correlation between the data and reduce the complexity of the algorithm to significantly speed up the running speed of the program. Three adaptive sparse multi-view canonical correlation analysis methods are used as benchmarks to evaluate the difference between real neuroimaging data and synthetic data. Compared with the other three methods, our proposed method has obtained better or comparable typical correlation coefficients and typical weights. Moreover, the following experimental results show that the MC-unAdaSMCCA method cannot only identify biomarkers related to AD and mild cognitive impairment (MCI), but also has a strong ability to resist noise and process high-dimensional data. Therefore, our proposed method provides a reliable approach to multi-modal imaging genetic researches.
Collapse
Affiliation(s)
- Wenbo Wang
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, People's Republic of China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, People's Republic of China.
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, People's Republic of China
| | - Kai Wei
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, People's Republic of China
| |
Collapse
|
19
|
Won JH, Youn J, Park H. Enhanced neuroimaging genetics using multi-view non-negative matrix factorization with sparsity and prior knowledge. Med Image Anal 2022; 77:102378. [DOI: 10.1016/j.media.2022.102378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/29/2021] [Accepted: 01/26/2022] [Indexed: 11/28/2022]
|
20
|
Associating brain imaging phenotypes and genetic in Alzheimer's disease via JSCCA approach with autocorrelation constraints. Med Biol Eng Comput 2021; 60:95-108. [PMID: 34714488 DOI: 10.1007/s11517-021-02439-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Imaging genetics research can explore the potential correlation between imaging and genomics. Most association analysis methods cannot effectively use the prior knowledge of the original data. In this respect, we add the prior knowledge of each original data to mine more effective biomarkers. The study of imaging genetics based on the sparse canonical correlation analysis (SCCA) is helpful to mine the potential biomarkers of neurological diseases. To improve the performance and interpretability of SCCA, we proposed a penalty method based on the autocorrelation matrix for discovering the possible biological mechanism between single nucleotide polymorphisms (SNP) variations and brain regions changes of Alzheimer's disease (AD). The addition of the penalty allows the proposed algorithm to analyze the correlation between different modal features. The proposed algorithm obtains more biologically interpretable ROIs and SNPs that are significantly related to AD, which has better anti-noise performance. Compared with other SCCA-based algorithms (JCB-SCCA, JSNMNMF), the proposed algorithm can still maintain a stronger correlation with ground truth even when the noise is larger. Then, we put the regions of interest (ROI) selected by the three algorithms into the SVM classifier. The proposed algorithm has higher classification accuracy. Also, we use ridge regression with SNPs selected by three algorithms and four AD risk ROIs. The proposed algorithm has a smaller root mean square error (RMSE). It shows that proposed algorithm has a good ability in association recognition and feature selection. Furthermore, it selects important features more stably, improving the clinical diagnosis of new potential biomarkers.
Collapse
|
21
|
Sheng J, Wang L, Cheng H, Zhang Q, Zhou R, Shi Y. Strategies for multivariate analyses of imaging genetics study in Alzheimer's disease. Neurosci Lett 2021; 762:136147. [PMID: 34332030 DOI: 10.1016/j.neulet.2021.136147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 03/27/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease primarily affecting the elderly population. Early diagnosis of AD is critical for the management of this disease. Imaging genetics examines the influence of genetic variants (i.e., single nucleotide polymorphisms (SNPs)) on brain structure and function and many novel approaches of imaging genetics are proposed for studying AD. We review and synthesize the Alzheimer's Disease Neuroimaging Initiative (ADNI) genetic associations with quantitative disease endophenotypes including structural and functional neuroimaging, diffusion tensor imaging (DTI), positron emission tomography (PET), and fluid biomarker assays. In this review, we survey recent publications using neuroimaging and genetic data of AD, with a focus on methods capturing multivariate effects accommodating the large number variables from both imaging data and genetic data. We review methods focused on bridging the imaging and genetic data by establishing genotype-phenotype association, including sparse canonical correlation analysis, parallel independent component analysis, sparse reduced rank regression, sparse partial least squares, genome-wide association study, and so on. The broad availability and wide scope of ADNI genetic and phenotypic data has advanced our understanding of the genetic basis of AD and has nominated novel targets for future pharmaceutical therapy and biomarker development.
Collapse
Affiliation(s)
- Jinhua Sheng
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China.
| | - Luyun Wang
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China; College of Information Engineering, Hangzhou Vocational & Technical College, Hangzhou, Zhejiang 310018, China
| | - Hu Cheng
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | | | - Rougang Zhou
- Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China; School of Mechanical Engineering, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Mstar Technologies Inc., Hangzhou, Zhejiang 310018, China
| | - Yuchen Shi
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
22
|
Ke F, Kong W, Wang S. Identifying Imaging Genetics Biomarkers of Alzheimer's Disease by Multi-Task Sparse Canonical Correlation Analysis and Regression. Front Genet 2021; 12:706986. [PMID: 34422007 PMCID: PMC8375409 DOI: 10.3389/fgene.2021.706986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/19/2021] [Indexed: 11/29/2022] Open
Abstract
Imaging genetics combines neuroimaging and genetics to assess the relationships between genetic variants and changes in brain structure and metabolism. Sparse canonical correlation analysis (SCCA) models are well-known tools for identifying meaningful biomarkers in imaging genetics. However, most SCCA models incorporate only diagnostic status information, which poses challenges for finding disease-specific biomarkers. In this study, we proposed a multi-task sparse canonical correlation analysis and regression (MT-SCCAR) model to reveal disease-specific associations between single nucleotide polymorphisms and quantitative traits derived from multi-modal neuroimaging data in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort. MT-SCCAR uses complementary information carried by multiple-perspective cognitive scores and encourages group sparsity on genetic variants. In contrast with two other multi-modal SCCA models, MT-SCCAR embedded more accurate neuropsychological assessment information through linear regression and enhanced the correlation coefficients, leading to increased identification of high-risk brain regions. Furthermore, MT-SCCAR identified primary genetic risk factors for Alzheimer’s disease (AD), including rs429358, and found some association patterns between genetic variants and brain regions. Thus, MT-SCCAR contributes to deciphering genetic risk factors of brain structural and metabolic changes by identifying potential risk biomarkers.
Collapse
Affiliation(s)
- Fengchun Ke
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| |
Collapse
|
23
|
Wang M, Shao W, Hao X, Shen L, Zhang D. Identify Consistent Cross-Modality Imaging Genetic Patterns via Discriminant Sparse Canonical Correlation Analysis. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1549-1561. [PMID: 31581090 DOI: 10.1109/tcbb.2019.2944825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Sparse canonical correlation analysis (SCCA) is a bi-multivariate technique used in imaging genetics to identify complex multi-SNP-multi-QT associations. However, the traditional SCCA algorithm has been designed to seek a linear correlation between the SNP genotype and brain imaging phenotype, ignoring the discriminant similarity information between within-class subjects in brain imaging genetics association analysis. In addition, multi-modality brain imaging phenotypes are extracted from different perspectives and imaging markers from the same region consistently showing up in multimodalities may provide more insights for the mechanistic understanding of diseases. In this paper, a novel multi-modality discriminant SCCA algorithm (MD-SCCA) is proposed to overcome these limitations as well as to improve learning results by incorporating valuable discriminant similarity information into the SCCA algorithm. Specifically, we first extract the discriminant similarity information between within-class subjects by the sparse representation. Second, the discriminant similarity information is enforced within SCCA to construct a discriminant SCCA algorithm (D-SCCA). At last, the MD-SCCA algorithm is adopted to fully explore the relationships among different modalities of different subjects. In experiments, both synthetic dataset and real data from the Alzheimer's Disease Neuroimaging Initiative database are used to test the performance of our algorithm. The empirical results have demonstrated that the proposed algorithm not only produces improved cross-validation performances but also identifies consistent cross-modality imaging genetic biomarkers.
Collapse
|
24
|
Wang M, Shao W, Hao X, Zhang D. Identify Complex Imaging Genetic Patterns via Fusion Self-Expressive Network Analysis. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:1673-1686. [PMID: 33661732 DOI: 10.1109/tmi.2021.3063785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In the brain imaging genetic studies, it is a challenging task to estimate the association between quantitative traits (QTs) extracted from neuroimaging data and genetic markers such as single-nucleotide polymorphisms (SNPs). Most of the existing association studies are based on the extensions of sparse canonical correlation analysis (SCCA) for the identification of complex bi-multivariate associations, which can take the specific structure and group information into consideration. However, they often take the original data as input without considering its underlying complex multi-subspace structure, which will deteriorate the performance of the following integrative analysis. Accordingly, in this paper, the self-expressive property is exploited for the reconstruction of the original data before the association analysis, which can well describe the similarity structure. Specifically, we first apply the within-class similarity information to construct self-expressive networks by sparse representation. Then, we use the fusion method to iteratively fuse the self-expressive networks from multi-modality brain phenotypes into one network. Finally, we calculate the imaging genetic association based on the fused self-expressive network. We conduct the experiments on both single-modality and multi-modality phenotype data. Related experimental results validate that our method can not only better estimate the potential association between genetic markers and quantitative traits but also identify consistent multi-modality imaging genetic biomarkers to guide the interpretation of Alzheimer's disease.
Collapse
|
25
|
Wang L, Kong W, Wang S. Detecting genetic associations with brain imaging phenotypes in Alzheimer's disease via a novel structured KCCA approach. J Bioinform Comput Biol 2021; 19:2150012. [PMID: 33950804 DOI: 10.1142/s0219720021500128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Neuroimaging genetics has become an important research topic since it can reveal complex associations between genetic variants (i.e. single nucleotide polymorphisms (SNPs) and the structures or functions of the human brain. However, existing kernel mapping is difficult to directly use the sparse representation method in the kernel feature space, which makes it difficult for most existing sparse canonical correlation analysis (SCCA) methods to be directly promoted in the kernel feature space. To bridge this gap, we adopt a novel alternating projected gradient approach, gradient KCCA (gradKCCA) model to develop a powerful model for exploring the intrinsic associations among genetic markers, imaging quantitative traits (QTs) of interest. Specifically, this model solves kernel canonical correlation (KCCA) with an additional constraint that projection directions have pre-images in the original data space, a sparsity-inducing variant of the model is achieved through controlling the [Formula: see text]-norm of the preimages of the projection directions. We evaluate this model using Alzheimer's disease Neuroimaging Initiative (ADNI) cohort to discover the relationships among SNPs from Alzheimer's disease (AD) risk gene APOE, imaging QTs extracted from structural magnetic resonance imaging (MRI) scans. Our results show that the algorithm not only outperforms the traditional KCCA method in terms of Root Mean Square Error (RMSE) and Correlation Coefficient (CC) but also identify the meaningful and relevant biomarkers of SNPs (e.g. rs157594 and rs405697), which are positively related to right Postcentral and right SupraMarginal brain regions in this study. Empirical results indicate its promising capability in revealing biologically meaningful neuroimaging genetics associations and improving the disease-related mechanistic understanding of AD.
Collapse
Affiliation(s)
- Lei Wang
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai 201306, P. R. China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai 201306, P. R. China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave, Shanghai 201306, P. R. China
| |
Collapse
|
26
|
Li Y, Yu C, Zhao Y, Yao W, Aseltine RH, Chen K. Pursuing sources of heterogeneity in modeling clustered population. Biometrics 2021; 78:716-729. [PMID: 33527347 DOI: 10.1111/biom.13434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/22/2020] [Accepted: 01/13/2021] [Indexed: 11/28/2022]
Abstract
Researchers often have to deal with heterogeneous population with mixed regression relationships, increasingly so in the era of data explosion. In such problems, when there are many candidate predictors, it is not only of interest to identify the predictors that are associated with the outcome, but also to distinguish the true sources of heterogeneity, that is, to identify the predictors that have different effects among the clusters and thus are the true contributors to the formation of the clusters. We clarify the concepts of the source of heterogeneity that account for potential scale differences of the clusters and propose a regularized finite mixture effects regression to achieve heterogeneity pursuit and feature selection simultaneously. We develop an efficient algorithm and show that our approach can achieve both estimation and selection consistency. Simulation studies further demonstrate the effectiveness of our method under various practical scenarios. Three applications are presented, namely, an imaging genetics study for linking genetic factors and brain neuroimaging traits in Alzheimer's disease, a public health study for exploring the association between suicide risk among adolescents and their school district characteristics, and a sport analytics study for understanding how the salary levels of baseball players are associated with their performance and contractual status.
Collapse
Affiliation(s)
- Yan Li
- Department of Statistics, University of Connecticut, Storrs, Connecticut
| | - Chun Yu
- School of Statistics, Jiangxi University of Finance and Economics, Nanchang, China
| | - Yize Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Weixin Yao
- Department of Statistics, University of California, Riverside, California
| | - Robert H Aseltine
- Center for Population Health, University of Connecticut Health Center, Farmington, Connecticut
| | - Kun Chen
- Department of Statistics, University of Connecticut, Storrs, Connecticut.,Center for Population Health, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
27
|
Du L, Liu F, Liu K, Yao X, Risacher SL, Han J, Saykin AJ, Shen L. Associating Multi-Modal Brain Imaging Phenotypes and Genetic Risk Factors via a Dirty Multi-Task Learning Method. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:3416-3428. [PMID: 32746095 PMCID: PMC7705646 DOI: 10.1109/tmi.2020.2995510] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Brain imaging genetics becomes more and more important in brain science, which integrates genetic variations and brain structures or functions to study the genetic basis of brain disorders. The multi-modal imaging data collected by different technologies, measuring the same brain distinctly, might carry complementary information. Unfortunately, we do not know the extent to which the phenotypic variance is shared among multiple imaging modalities, which further might trace back to the complex genetic mechanism. In this paper, we propose a novel dirty multi-task sparse canonical correlation analysis (SCCA) to study imaging genetic problems with multi-modal brain imaging quantitative traits (QTs) involved. The proposed method takes advantages of the multi-task learning and parameter decomposition. It can not only identify the shared imaging QTs and genetic loci across multiple modalities, but also identify the modality-specific imaging QTs and genetic loci, exhibiting a flexible capability of identifying complex multi-SNP-multi-QT associations. Using the state-of-the-art multi-view SCCA and multi-task SCCA, the proposed method shows better or comparable canonical correlation coefficients and canonical weights on both synthetic and real neuroimaging genetic data. In addition, the identified modality-consistent biomarkers, as well as the modality-specific biomarkers, provide meaningful and interesting information, demonstrating the dirty multi-task SCCA could be a powerful alternative method in multi-modal brain imaging genetics.
Collapse
Affiliation(s)
- Lei Du
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China
| | - Fang Liu
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China
| | - Kefei Liu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Xiaohui Yao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Junwei Han
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Zhang L, Wang M, Liu M, Zhang D. A Survey on Deep Learning for Neuroimaging-Based Brain Disorder Analysis. Front Neurosci 2020; 14:779. [PMID: 33117114 PMCID: PMC7578242 DOI: 10.3389/fnins.2020.00779] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Deep learning has recently been used for the analysis of neuroimages, such as structural magnetic resonance imaging (MRI), functional MRI, and positron emission tomography (PET), and it has achieved significant performance improvements over traditional machine learning in computer-aided diagnosis of brain disorders. This paper reviews the applications of deep learning methods for neuroimaging-based brain disorder analysis. We first provide a comprehensive overview of deep learning techniques and popular network architectures by introducing various types of deep neural networks and recent developments. We then review deep learning methods for computer-aided analysis of four typical brain disorders, including Alzheimer's disease, Parkinson's disease, Autism spectrum disorder, and Schizophrenia, where the first two diseases are neurodegenerative disorders and the last two are neurodevelopmental and psychiatric disorders, respectively. More importantly, we discuss the limitations of existing studies and present possible future directions.
Collapse
Affiliation(s)
- Li Zhang
- College of Computer Science and Technology, Nanjing Forestry University, Nanjing, China
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Mingliang Wang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Mingxia Liu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| |
Collapse
|
29
|
Zhuang X, Yang Z, Cordes D. A technical review of canonical correlation analysis for neuroscience applications. Hum Brain Mapp 2020; 41:3807-3833. [PMID: 32592530 PMCID: PMC7416047 DOI: 10.1002/hbm.25090] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/23/2020] [Indexed: 12/11/2022] Open
Abstract
Collecting comprehensive data sets of the same subject has become a standard in neuroscience research and uncovering multivariate relationships among collected data sets have gained significant attentions in recent years. Canonical correlation analysis (CCA) is one of the powerful multivariate tools to jointly investigate relationships among multiple data sets, which can uncover disease or environmental effects in various modalities simultaneously and characterize changes during development, aging, and disease progressions comprehensively. In the past 10 years, despite an increasing number of studies have utilized CCA in multivariate analysis, simple conventional CCA dominates these applications. Multiple CCA-variant techniques have been proposed to improve the model performance; however, the complicated multivariate formulations and not well-known capabilities have delayed their wide applications. Therefore, in this study, a comprehensive review of CCA and its variant techniques is provided. Detailed technical formulation with analytical and numerical solutions, current applications in neuroscience research, and advantages and limitations of each CCA-related technique are discussed. Finally, a general guideline in how to select the most appropriate CCA-related technique based on the properties of available data sets and particularly targeted neuroscience questions is provided.
Collapse
Affiliation(s)
- Xiaowei Zhuang
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
| | - Zhengshi Yang
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
- University of ColoradoBoulderColoradoUSA
- Department of Brain HealthUniversity of NevadaLas VegasNevadaUSA
| |
Collapse
|
30
|
Lee H, Park BY, Byeon K, Won JH, Kim M, Kim SH, Park H. Multivariate association between brain function and eating disorders using sparse canonical correlation analysis. PLoS One 2020; 15:e0237511. [PMID: 32785278 PMCID: PMC7423138 DOI: 10.1371/journal.pone.0237511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 07/28/2020] [Indexed: 12/26/2022] Open
Abstract
Eating disorder is highly associated with obesity and it is related to brain dysfunction as well. Still, the functional substrates of the brain associated with behavioral traits of eating disorder are underexplored. Existing neuroimaging studies have explored the association between eating disorder and brain function without using all the information provided by the eating disorder related questionnaire but by adopting summary factors. Here, we aimed to investigate the multivariate association between brain function and eating disorder at fine-grained question-level information. Our study is a retrospective secondary analysis that re-analyzed resting-state functional magnetic resonance imaging of 284 participants from the enhanced Nathan Kline Institute-Rockland Sample database. Leveraging sparse canonical correlation analysis, we associated the functional connectivity of all brain regions and all questions in the eating disorder questionnaires. We found that executive- and inhibitory control-related frontoparietal networks showed positive associations with questions of restraint eating, while brain regions involved in the reward system showed negative associations. Notably, inhibitory control-related brain regions showed a positive association with the degree of obesity. Findings were well replicated in the independent validation dataset (n = 34). The results of this study might contribute to a better understanding of brain function with respect to eating disorder.
Collapse
Affiliation(s)
- Hyebin Lee
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea
| | - Bo-yong Park
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Kyoungseob Byeon
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea
| | - Ji Hye Won
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea
| | - Mansu Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Se-Hong Kim
- Department of Family Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Hyunjin Park
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Korea
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea
- * E-mail:
| |
Collapse
|
31
|
Won JH, Kim M, Youn J, Park H. Prediction of age at onset in Parkinson's disease using objective specific neuroimaging genetics based on a sparse canonical correlation analysis. Sci Rep 2020; 10:11662. [PMID: 32669683 PMCID: PMC7363828 DOI: 10.1038/s41598-020-68301-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/22/2020] [Indexed: 01/19/2023] Open
Abstract
The age at onset (AAO) is an important determinant in Parkinson’s disease (PD). Neuroimaging genetics is suitable for studying AAO in PD as it jointly analyzes imaging and genetics. We aimed to identify features associated with AAO in PD by applying the objective-specific neuroimaging genetics approach and constructing an AAO prediction model. Our objective-specific neuroimaging genetics extended the sparse canonical correlation analysis by an additional data type related to the target task to investigate possible associations of the imaging–genetic, genetic–target, and imaging–target pairs simultaneously. The identified imaging, genetic, and combined features were used to construct analytical models to predict the AAO in a nested five-fold cross-validation. We compared our approach with those from two feature selection approaches where only associations of imaging–target and genetic–target were explored. Using only imaging features, AAO prediction was accurate in all methods. Using only genetic features, the results from other methods were worse or unstable compared to our model. Using both imaging and genetic features, our proposed model predicted the AAO well (r = 0.5486). Our findings could have significant impacts on the characterization of prodromal PD and contribute to diagnosing PD early because genetic features could be measured accurately from birth.
Collapse
Affiliation(s)
- Ji Hye Won
- Department of Electronic and Computer Engineering, Sungkyunkwan University, Suwon, Korea.,Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Mansu Kim
- Department of Electronic and Computer Engineering, Sungkyunkwan University, Suwon, Korea.,Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Jinyoung Youn
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Irwon-ro 81, Gangnam-gu, Seoul, 06351, Korea. .,Neuroscience Center, Samsung Medical Center, Seoul, Korea.
| | - Hyunjin Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea. .,School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
32
|
Future avenues for Alzheimer's disease detection and therapy: liquid biopsy, intracellular signaling modulation, systems pharmacology drug discovery. Neuropharmacology 2020; 185:108081. [PMID: 32407924 DOI: 10.1016/j.neuropharm.2020.108081] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/01/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022]
Abstract
When Alzheimer's disease (AD) disease-modifying therapies will be available, global healthcare systems will be challenged by a large-scale demand for clinical and biological screening. Validation and qualification of globally accessible, minimally-invasive, and time-, cost-saving blood-based biomarkers need to be advanced. Novel pathophysiological mechanisms (and related candidate biomarkers) - including neuroinflammation pathways (TREM2 and YKL-40), axonal degeneration (neurofilament light chain protein), synaptic dysfunction (neurogranin, synaptotagmin, α-synuclein, and SNAP-25) - may be integrated into an expanding pathophysiological and biomarker matrix and, ultimately, integrated into a comprehensive blood-based liquid biopsy, aligned with the evolving ATN + classification system and the precision medicine paradigm. Liquid biopsy-based diagnostic and therapeutic algorithms are increasingly employed in Oncology disease-modifying therapies and medical practice, showing an enormous potential for AD and other brain diseases as well. For AD and other neurodegenerative diseases, newly identified aberrant molecular pathways have been identified as suitable therapeutic targets and are currently investigated by academia/industry-led R&D programs, including the nerve-growth factor pathway in basal forebrain cholinergic neurons, the sigma1 receptor, and the GTPases of the Rho family. Evidence for a clinical long-term effect on cognitive function and brain health span of cholinergic compounds, drug candidates for repositioning programs, and non-pharmacological multidomain interventions (nutrition, cognitive training, and physical activity) is developing as well. Ultimately, novel pharmacological paradigms, such as quantitative systems pharmacology-based integrative/explorative approaches, are gaining momentum to optimize drug discovery and accomplish effective pathway-based strategies for precision medicine. This article is part of the special issue on 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
|
33
|
Kim M, Won JH, Youn J, Park H. Joint-Connectivity-Based Sparse Canonical Correlation Analysis of Imaging Genetics for Detecting Biomarkers of Parkinson's Disease. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:23-34. [PMID: 31144631 DOI: 10.1109/tmi.2019.2918839] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Imaging genetics is a method used to detect associations between imaging and genetic variables. Some researchers have used sparse canonical correlation analysis (SCCA) for imaging genetics. This study was conducted to improve the efficiency and interpretability of SCCA. We propose a connectivity-based penalty for incorporating biological prior information. Our proposed approach, named joint connectivity-based SCCA (JCB-SCCA), includes the proposed penalty and can handle multi-modal neuroimaging datasets. Different neuroimaging techniques provide distinct information on the brain and have been used to investigate various neurological disorders, including Parkinson's disease (PD). We applied our algorithm to simulated and real imaging genetics datasets for performance evaluation. Our algorithm was able to select important features in a more robust manner compared with other multivariate methods. The algorithm revealed promising features of single-nucleotide polymorphisms and brain regions related to PD by using a real imaging genetic dataset. The proposed imaging genetics model can be used to improve clinical diagnosis in the form of novel potential biomarkers. We hope to apply our algorithm to cohorts such as Alzheimer's patients or healthy subjects to determine the generalizability of our algorithm.
Collapse
|
34
|
Shen L, Thompson PM. Brain Imaging Genomics: Integrated Analysis and Machine Learning. PROCEEDINGS OF THE IEEE. INSTITUTE OF ELECTRICAL AND ELECTRONICS ENGINEERS 2020; 108:125-162. [PMID: 31902950 PMCID: PMC6941751 DOI: 10.1109/jproc.2019.2947272] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Brain imaging genomics is an emerging data science field, where integrated analysis of brain imaging and genomics data, often combined with other biomarker, clinical and environmental data, is performed to gain new insights into the phenotypic, genetic and molecular characteristics of the brain as well as their impact on normal and disordered brain function and behavior. It has enormous potential to contribute significantly to biomedical discoveries in brain science. Given the increasingly important role of statistical and machine learning in biomedicine and rapidly growing literature in brain imaging genomics, we provide an up-to-date and comprehensive review of statistical and machine learning methods for brain imaging genomics, as well as a practical discussion on method selection for various biomedical applications.
Collapse
Affiliation(s)
- Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90232, USA
| |
Collapse
|
35
|
Vilor-Tejedor N, Alemany S, Cáceres A, Bustamante M, Pujol J, Sunyer J, González JR. Strategies for integrated analysis in imaging genetics studies. Neurosci Biobehav Rev 2018; 93:57-70. [PMID: 29944960 DOI: 10.1016/j.neubiorev.2018.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/30/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023]
Abstract
Imaging Genetics (IG) integrates neuroimaging and genomic data from the same individual, deepening our knowledge of the biological mechanisms behind neurodevelopmental domains and neurological disorders. Although the literature on IG has exponentially grown over the past years, the majority of studies have mainly analyzed associations between candidate brain regions and individual genetic variants. However, this strategy is not designed to deal with the complexity of neurobiological mechanisms underlying behavioral and neurodevelopmental domains. Moreover, larger sample sizes and increased multidimensionality of this type of data represents a challenge for standardizing modeling procedures in IG research. This review provides a systematic update of the methods and strategies currently used in IG studies, and serves as an analytical framework for researchers working in this field. To complement the functionalities of the Neuroconductor framework, we also describe existing R packages that implement these methodologies. In addition, we present an overview of how these methodological approaches are applied in integrating neuroimaging and genetic data.
Collapse
Affiliation(s)
- Natàlia Vilor-Tejedor
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Barcelona Beta Brain Research Center (BBRC) - Pasqual Maragall Foundation, Barcelona, Spain.
| | - Silvia Alemany
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Alejandro Cáceres
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Mariona Bustamante
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jesús Pujol
- MRI Research Unit, Hospital del Mar, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM G21, Barcelona, Spain
| | - Jordi Sunyer
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Juan R González
- Barcelona Research Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.
| |
Collapse
|