1
|
Sui J, Zhi D, Calhoun VD. Data-driven multimodal fusion: approaches and applications in psychiatric research. PSYCHORADIOLOGY 2023; 3:kkad026. [PMID: 38143530 PMCID: PMC10734907 DOI: 10.1093/psyrad/kkad026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 12/26/2023]
Abstract
In the era of big data, where vast amounts of information are being generated and collected at an unprecedented rate, there is a pressing demand for innovative data-driven multi-modal fusion methods. These methods aim to integrate diverse neuroimaging perspectives to extract meaningful insights and attain a more comprehensive understanding of complex psychiatric disorders. However, analyzing each modality separately may only reveal partial insights or miss out on important correlations between different types of data. This is where data-driven multi-modal fusion techniques come into play. By combining information from multiple modalities in a synergistic manner, these methods enable us to uncover hidden patterns and relationships that would otherwise remain unnoticed. In this paper, we present an extensive overview of data-driven multimodal fusion approaches with or without prior information, with specific emphasis on canonical correlation analysis and independent component analysis. The applications of such fusion methods are wide-ranging and allow us to incorporate multiple factors such as genetics, environment, cognition, and treatment outcomes across various brain disorders. After summarizing the diverse neuropsychiatric magnetic resonance imaging fusion applications, we further discuss the emerging neuroimaging analyzing trends in big data, such as N-way multimodal fusion, deep learning approaches, and clinical translation. Overall, multimodal fusion emerges as an imperative approach providing valuable insights into the underlying neural basis of mental disorders, which can uncover subtle abnormalities or potential biomarkers that may benefit targeted treatments and personalized medical interventions.
Collapse
Affiliation(s)
- Jing Sui
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Dongmei Zhi
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Vince D Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia Institute of Technology, Emory University and Georgia State University, Atlanta, GA 30303, United States
| |
Collapse
|
2
|
Wu S, Venkataraman A, Ghosal S. GIRUS-net: A Multimodal Deep Learning Model Identifying Imaging and Genetic Biomarkers Linked to Alzheimer's Disease Severity. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083359 PMCID: PMC11005466 DOI: 10.1109/embc40787.2023.10341000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
We introduce an explainable deep neural architecture that combines brain structure with genetic influence to improve disease severity prediction in Alzheimer's disease. Our framework consists of an encoder, a decoder, and a rank-consistent ordinal regression module. The encoder projects neural imaging and genetics data into a low-dimensional latent space regularized by the decoder. The ordinal regression module guides the feature embedding process to find discriminative patterns representative of disease severity. We also add a learnable dropout layer that learns feature importance and extracts explainable biomarkers from the data. We evaluate our model using structural MRI (sMRI) and Single Nucleotide Polymorphism (SNP) data provided by the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. In 2-class severity classification comparison, our model has a median F-score of 0.86 (baseline median F-score range: 0.57-0.81). In 3-class classification comparison, our model's median F-score is 0.50 (baseline range: 0.17 - 0.41). In 4-class classification comparison, our model's median F-score is 0.40 (baseline range: 0.14 - 0.39). We demonstrate that our model provides improved disease diagnosis alongside sparse and clinically relevant biomarkers.Clinical relevance-This study provides a deep-learning model that can predict Alzheimer's disease severity levels while identifying consistent and clinically relevant biomarkers.
Collapse
Affiliation(s)
- Sarah Wu
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Archana Venkataraman
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, USA
| | - Sayan Ghosal
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
3
|
Wang S, Zheng K, Kong W, Huang R, Liu L, Wen G, Yu Y. Multimodal data fusion based on IGERNNC algorithm for detecting pathogenic brain regions and genes in Alzheimer's disease. Brief Bioinform 2023; 24:6887308. [PMID: 36502428 DOI: 10.1093/bib/bbac515] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/28/2022] [Accepted: 10/30/2022] [Indexed: 12/14/2022] Open
Abstract
At present, the study on the pathogenesis of Alzheimer's disease (AD) by multimodal data fusion analysis has been attracted wide attention. It often has the problems of small sample size and high dimension with the multimodal medical data. In view of the characteristics of multimodal medical data, the existing genetic evolution random neural network cluster (GERNNC) model combine genetic evolution algorithm and neural network for the classification of AD patients and the extraction of pathogenic factors. However, the model does not take into account the non-linear relationship between brain regions and genes and the problem that the genetic evolution algorithm can fall into local optimal solutions, which leads to the overall performance of the model is not satisfactory. In order to solve the above two problems, this paper made some improvements on the construction of fusion features and genetic evolution algorithm in GERNNC model, and proposed an improved genetic evolution random neural network cluster (IGERNNC) model. The IGERNNC model uses mutual information correlation analysis method to combine resting-state functional magnetic resonance imaging data with single nucleotide polymorphism data for the construction of fusion features. Based on the traditional genetic evolution algorithm, elite retention strategy and large variation genetic algorithm are added to avoid the model falling into the local optimal solution. Through multiple independent experimental comparisons, the IGERNNC model can more effectively identify AD patients and extract relevant pathogenic factors, which is expected to become an effective tool in the field of AD research.
Collapse
Affiliation(s)
- Shuaiqun Wang
- School of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Kai Zheng
- School of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Wei Kong
- School of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Ruiwen Huang
- School of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Lulu Liu
- School of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Gen Wen
- School of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Yaling Yu
- School of Information Engineering, Shanghai Maritime University, Shanghai, China
| |
Collapse
|
4
|
Huang W, Tan K, Zhang Z, Hu J, Dong S. A Review of Fusion Methods for Omics and Imaging Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:74-93. [PMID: 35044920 DOI: 10.1109/tcbb.2022.3143900] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The development of omics data and biomedical images has greatly advanced the progress of precision medicine in diagnosis, treatment, and prognosis. The fusion of omics and imaging data, i.e., omics-imaging fusion, offers a new strategy for understanding complex diseases. However, due to a variety of issues such as the limited number of samples, high dimensionality of features, and heterogeneity of different data types, efficiently learning complementary or associated discriminative fusion information from omics and imaging data remains a challenge. Recently, numerous machine learning methods have been proposed to alleviate these problems. In this review, from the perspective of fusion levels and fusion methods, we first provide an overview of preprocessing and feature extraction methods for omics and imaging data, and comprehensively analyze and summarize the basic forms and variations of commonly used and newly emerging fusion methods, along with their advantages, disadvantages and the applicable scope. We then describe public datasets and compare experimental results of various fusion methods on the ADNI and TCGA datasets. Finally, we discuss future prospects and highlight remaining challenges in the field.
Collapse
|
5
|
An Alzheimer’s Disease Patient-Derived Olfactory Stem Cell Model Identifies Gene Expression Changes Associated with Cognition. Cells 2022; 11:cells11203258. [PMID: 36291125 PMCID: PMC9601087 DOI: 10.3390/cells11203258] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/25/2022] Open
Abstract
An early symptom of Alzheimer’s disease (AD) is an impaired sense of smell, for which the molecular basis remains elusive. Here, we generated human olfactory neurosphere-derived (ONS) cells from people with AD and mild cognitive impairment (MCI), and performed global RNA sequencing to determine gene expression changes. ONS cells expressed markers of neuroglial differentiation, providing a unique cellular model to explore changes of early AD-associated pathways. Our transcriptomics data from ONS cells revealed differentially expressed genes (DEGs) associated with cognitive processes in AD cells compared to MCI, or matched healthy controls (HC). A-Kinase Anchoring Protein 6 (AKAP6) was the most significantly altered gene in AD compared to both MCI and HC, and has been linked to cognitive function. The greatest change in gene expression of all DEGs occurred between AD and MCI. Gene pathway analysis revealed defects in multiple cellular processes with aging, intellectual deficiency and alternative splicing being the most significantly dysregulated in AD ONS cells. Our results demonstrate that ONS cells can provide a cellular model for AD that recapitulates disease-associated differences. We have revealed potential novel genes, including AKAP6 that may have a role in AD, particularly MCI to AD transition, and should be further examined.
Collapse
|
6
|
Ko W, Jung W, Jeon E, Suk HI. A Deep Generative-Discriminative Learning for Multimodal Representation in Imaging Genetics. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:2348-2359. [PMID: 35344489 DOI: 10.1109/tmi.2022.3162870] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Imaging genetics, one of the foremost emerging topics in the medical imaging field, analyzes the inherent relations between neuroimaging and genetic data. As deep learning has gained widespread acceptance in many applications, pioneering studies employed deep learning frameworks for imaging genetics. However, existing approaches suffer from some limitations. First, they often adopt a simple strategy for joint learning of phenotypic and genotypic features. Second, their findings have not been extended to biomedical applications, e.g., degenerative brain disease diagnosis and cognitive score prediction. Finally, existing studies perform insufficient and inappropriate analyses from the perspective of data science and neuroscience. In this work, we propose a novel deep learning framework to simultaneously tackle the aforementioned issues. Our proposed framework learns to effectively represent the neuroimaging and the genetic data jointly, and achieves state-of-the-art performance when used for Alzheimer's disease and mild cognitive impairment identification. Furthermore, unlike the existing methods, the framework enables learning the relation between imaging phenotypes and genotypes in a nonlinear way without any prior neuroscientific knowledge. To demonstrate the validity of our proposed framework, we conducted experiments on a publicly available dataset and analyzed the results from diverse perspectives. Based on our experimental results, we believe that the proposed framework has immense potential to provide new insights and perspectives in deep learning-based imaging genetics studies.
Collapse
|
7
|
Ruffini N, Klingenberg S, Heese R, Schweiger S, Gerber S. The Big Picture of Neurodegeneration: A Meta Study to Extract the Essential Evidence on Neurodegenerative Diseases in a Network-Based Approach. Front Aging Neurosci 2022; 14:866886. [PMID: 35832065 PMCID: PMC9271745 DOI: 10.3389/fnagi.2022.866886] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
The common features of all neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), and Huntington's disease, are the accumulation of aggregated and misfolded proteins and the progressive loss of neurons, leading to cognitive decline and locomotive dysfunction. Still, they differ in their ultimate manifestation, the affected brain region, and the kind of proteinopathy. In the last decades, a vast number of processes have been described as associated with neurodegenerative diseases, making it increasingly harder to keep an overview of the big picture forming from all those data. In this meta-study, we analyzed genomic, transcriptomic, proteomic, and epigenomic data of the aforementioned diseases using the data of 234 studies in a network-based approach to study significant general coherences but also specific processes in individual diseases or omics levels. In the analysis part, we focus on only some of the emerging findings, but trust that the meta-study provided here will be a valuable resource for various other researchers focusing on specific processes or genes contributing to the development of neurodegeneration.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research, Leibniz Association, Mainz, Germany
| | - Susanne Klingenberg
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Raoul Heese
- Fraunhofer Institute for Industrial Mathematics (ITWM), Kaiserslautern, Germany
| | - Susann Schweiger
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
8
|
Mirabnahrazam G, Ma D, Lee S, Popuri K, Lee H, Cao J, Wang L, Galvin JE, Beg MF. Machine Learning Based Multimodal Neuroimaging Genomics Dementia Score for Predicting Future Conversion to Alzheimer's Disease. J Alzheimers Dis 2022; 87:1345-1365. [PMID: 35466939 PMCID: PMC9195128 DOI: 10.3233/jad-220021] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND The increasing availability of databases containing both magnetic resonance imaging (MRI) and genetic data allows researchers to utilize multimodal data to better understand the characteristics of dementia of Alzheimer's type (DAT). OBJECTIVE The goal of this study was to develop and analyze novel biomarkers that can help predict the development and progression of DAT. METHODS We used feature selection and ensemble learning classifier to develop an image/genotype-based DAT score that represents a subject's likelihood of developing DAT in the future. Three feature types were used: MRI only, genetic only, and combined multimodal data. We used a novel data stratification method to better represent different stages of DAT. Using a pre-defined 0.5 threshold on DAT scores, we predicted whether a subject would develop DAT in the future. RESULTS Our results on Alzheimer's Disease Neuroimaging Initiative (ADNI) database showed that dementia scores using genetic data could better predict future DAT progression for currently normal control subjects (Accuracy = 0.857) compared to MRI (Accuracy = 0.143), while MRI can better characterize subjects with stable mild cognitive impairment (Accuracy = 0.614) compared to genetics (Accuracy = 0.356). Combining MRI and genetic data showed improved classification performance in the remaining stratified groups. CONCLUSION MRI and genetic data can contribute to DAT prediction in different ways. MRI data reflects anatomical changes in the brain, while genetic data can detect the risk of DAT progression prior to the symptomatic onset. Combining information from multimodal data appropriately can improve prediction performance.
Collapse
Affiliation(s)
| | - Da Ma
- School of Engineering, Simon Fraser University, Burnaby, BC, Canada
- School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Sieun Lee
- School of Engineering, Simon Fraser University, Burnaby, BC, Canada
- Mental Health & Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Karteek Popuri
- School of Engineering, Simon Fraser University, Burnaby, BC, Canada
| | - Hyunwoo Lee
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jiguo Cao
- Department of Statistics and Actuarial Science, Simon Fraser University, Burnaby, BC, Canada
| | - Lei Wang
- Psychiatry and Behavioral Health, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - James E Galvin
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mirza Faisal Beg
- School of Engineering, Simon Fraser University, Burnaby, BC, Canada
| | | |
Collapse
|
9
|
Machine learning approaches for parsing comorbidity/heterogeneity in antisociality and substance use disorders: A primer. PERSONALITY NEUROSCIENCE 2021; 4:e6. [PMID: 34909565 PMCID: PMC8640675 DOI: 10.1017/pen.2021.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
By some accounts, as many as 93% of individuals diagnosed with antisocial personality disorder (ASPD) or psychopathy also meet criteria for some form of substance use disorder (SUD). This high level of comorbidity, combined with an overlapping biopsychosocial profile, and potentially interacting features, has made it difficult to delineate the shared/unique characteristics of each disorder. Moreover, while rarely acknowledged, both SUD and antisociality exist as highly heterogeneous disorders in need of more targeted parcellation. While emerging data-driven nosology for psychiatric disorders (e.g., Research Domain Criteria (RDoC), Hierarchical Taxonomy of Psychopathology (HiTOP)) offers the opportunity for a more systematic delineation of the externalizing spectrum, the interrogation of large, complex neuroimaging-based datasets may require data-driven approaches that are not yet widely employed in psychiatric neuroscience. With this in mind, the proposed article sets out to provide an introduction into machine learning methods for neuroimaging that can help parse comorbid, heterogeneous externalizing samples. The modest machine learning work conducted to date within the externalizing domain demonstrates the potential utility of the approach but remains highly nascent. Within the paper, we make suggestions for how future work can make use of machine learning methods, in combination with emerging psychiatric nosology systems, to further diagnostic and etiological understandings of the externalizing spectrum. Finally, we briefly consider some challenges that will need to be overcome to encourage further progress in the field.
Collapse
|
10
|
Pursuit of precision medicine: Systems biology approaches in Alzheimer's disease mouse models. Neurobiol Dis 2021; 161:105558. [PMID: 34767943 PMCID: PMC10112395 DOI: 10.1016/j.nbd.2021.105558] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a complex disease that is mediated by numerous factors and manifests in various forms. A systems biology approach to studying AD involves analyses of various body systems, biological scales, environmental elements, and clinical outcomes to understand the genotype to phenotype relationship that potentially drives AD development. Currently, there are many research investigations probing how modifiable and nonmodifiable factors impact AD symptom presentation. This review specifically focuses on how imaging modalities can be integrated into systems biology approaches using model mouse populations to link brain level functional and structural changes to disease onset and progression. Combining imaging and omics data promotes the classification of AD into subtypes and paves the way for precision medicine solutions to prevent and treat AD.
Collapse
|
11
|
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
|
12
|
Vilor-Tejedor N, Garrido-Martín D, Rodriguez-Fernandez B, Lamballais S, Guigó R, Gispert JD. Multivariate Analysis and Modelling of multiple Brain endOphenotypes: Let's MAMBO! Comput Struct Biotechnol J 2021; 19:5800-5810. [PMID: 34765095 PMCID: PMC8567328 DOI: 10.1016/j.csbj.2021.10.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/01/2022] Open
Abstract
Imaging genetic studies aim to test how genetic information influences brain structure and function by combining neuroimaging-based brain features and genetic data from the same individual. Most studies focus on individual correlation and association tests between genetic variants and a single measurement of the brain. Despite the great success of univariate approaches, given the capacity of neuroimaging methods to provide a multiplicity of cerebral phenotypes, the development and application of multivariate methods become crucial. In this article, we review novel methods and strategies focused on the analysis of multiple phenotypes and genetic data. We also discuss relevant aspects of multi-trait modelling in the context of neuroimaging data.
Collapse
Affiliation(s)
- Natalia Vilor-Tejedor
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Diego Garrido-Martín
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | | | - Sander Lamballais
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| |
Collapse
|
13
|
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: 0.7] [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
|
14
|
Hampel H, Nisticò R, Seyfried NT, Levey AI, Modeste E, Lemercier P, Baldacci F, Toschi N, Garaci F, Perry G, Emanuele E, Valenzuela PL, Lucia A, Urbani A, Sancesario GM, Mapstone M, Corbo M, Vergallo A, Lista S. Omics sciences for systems biology in Alzheimer's disease: State-of-the-art of the evidence. Ageing Res Rev 2021; 69:101346. [PMID: 33915266 DOI: 10.1016/j.arr.2021.101346] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is characterized by non-linear, genetic-driven pathophysiological dynamics with high heterogeneity in biological alterations and disease spatial-temporal progression. Human in-vivo and post-mortem studies point out a failure of multi-level biological networks underlying AD pathophysiology, including proteostasis (amyloid-β and tau), synaptic homeostasis, inflammatory and immune responses, lipid and energy metabolism, oxidative stress. Therefore, a holistic, systems-level approach is needed to fully capture AD multi-faceted pathophysiology. Omics sciences - genomics, epigenomics, transcriptomics, proteomics, metabolomics, lipidomics - embedded in the systems biology (SB) theoretical and computational framework can generate explainable readouts describing the entire biological continuum of a disease. Such path in Neurology is encouraged by the promising results of omics sciences and SB approaches in Oncology, where stage-driven pathway-based therapies have been developed in line with the precision medicine paradigm. Multi-omics data integrated in SB network approaches will help detect and chart AD upstream pathomechanistic alterations and downstream molecular effects occurring in preclinical stages. Finally, integrating omics and neuroimaging data - i.e., neuroimaging-omics - will identify multi-dimensional biological signatures essential to track the clinical-biological trajectories, at the subpopulation or even individual level.
Collapse
|
15
|
Vogrinc D, Goričar K, Dolžan V. Genetic Variability in Molecular Pathways Implicated in Alzheimer's Disease: A Comprehensive Review. Front Aging Neurosci 2021; 13:646901. [PMID: 33815092 PMCID: PMC8012500 DOI: 10.3389/fnagi.2021.646901] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/16/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease, affecting a significant part of the population. The majority of AD cases occur in the elderly with a typical age of onset of the disease above 65 years. AD presents a major burden for the healthcare system and since population is rapidly aging, the burden of the disease will increase in the future. However, no effective drug treatment for a full-blown disease has been developed to date. The genetic background of AD is extensively studied; numerous genome-wide association studies (GWAS) identified significant genes associated with increased risk of AD development. This review summarizes more than 100 risk loci. Many of them may serve as biomarkers of AD progression, even in the preclinical stage of the disease. Furthermore, we used GWAS data to identify key pathways of AD pathogenesis: cellular processes, metabolic processes, biological regulation, localization, transport, regulation of cellular processes, and neurological system processes. Gene clustering into molecular pathways can provide background for identification of novel molecular targets and may support the development of tailored and personalized treatment of AD.
Collapse
Affiliation(s)
| | | | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
16
|
Ruffini N, Klingenberg S, Schweiger S, Gerber S. Common Factors in Neurodegeneration: A Meta-Study Revealing Shared Patterns on a Multi-Omics Scale. Cells 2020; 9:E2642. [PMID: 33302607 PMCID: PMC7764447 DOI: 10.3390/cells9122642] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are heterogeneous, progressive diseases with frequently overlapping symptoms characterized by a loss of neurons. Studies have suggested relations between neurodegenerative diseases for many years (e.g., regarding the aggregation of toxic proteins or triggering endogenous cell death pathways). We gathered publicly available genomic, transcriptomic, and proteomic data from 177 studies and more than one million patients to detect shared genetic patterns between the neurodegenerative diseases on three analyzed omics-layers. The results show a remarkably high number of shared differentially expressed genes between the transcriptomic and proteomic levels for all conditions, while showing a significant relation between genomic and proteomic data between AD and PD and AD and ALS. We identified a set of 139 genes being differentially expressed in several transcriptomic experiments of all four diseases. These 139 genes showed overrepresented gene ontology (GO) Terms involved in the development of neurodegeneration, such as response to heat and hypoxia, positive regulation of cytokines and angiogenesis, and RNA catabolic process. Furthermore, the four analyzed neurodegenerative diseases (NDDs) were clustered by their mean direction of regulation throughout all transcriptomic studies for this set of 139 genes, with the closest relation regarding this common gene set seen between AD and HD. GO-Term and pathway analysis of the proteomic overlap led to biological processes (BPs), related to protein folding and humoral immune response. Taken together, we could confirm the existence of many relations between Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis on transcriptomic and proteomic levels by analyzing the pathways and GO-Terms arising in these intersections. The significance of the connection and the striking relation of the results to processes leading to neurodegeneration between the transcriptomic and proteomic data for all four analyzed neurodegenerative diseases showed that exploring many studies simultaneously, including multiple omics-layers of different neurodegenerative diseases simultaneously, holds new relevant insights that do not emerge from analyzing these data separately. Furthermore, the results shed light on processes like the humoral immune response that have previously been described only for certain diseases. Our data therefore suggest human patients with neurodegenerative diseases should be addressed as complex biological systems by integrating multiple underlying data sources.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
- Leibniz Institute for Resilience Research, Leibniz Association, Wallstraße 7, 55122 Mainz, Germany
| | - Susanne Klingenberg
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susann Schweiger
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susanne Gerber
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| |
Collapse
|
17
|
Falakshahi H, Vergara VM, Liu J, Mathalon DH, Ford JM, Voyvodic J, Mueller BA, Belger A, McEwen S, Potkin SG, Preda A, Rokham H, Sui J, Turner JA, Plis S, Calhoun VD. Meta-Modal Information Flow: A Method for Capturing Multimodal Modular Disconnectivity in Schizophrenia. IEEE Trans Biomed Eng 2020; 67:2572-2584. [PMID: 31944934 PMCID: PMC7538162 DOI: 10.1109/tbme.2020.2964724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Multimodal measurements of the same phenomena provide complementary information and highlight different perspectives, albeit each with their own limitations. A focus on a single modality may lead to incorrect inferences, which is especially important when a studied phenomenon is a disease. In this paper, we introduce a method that takes advantage of multimodal data in addressing the hypotheses of disconnectivity and dysfunction within schizophrenia (SZ). METHODS We start with estimating and visualizing links within and among extracted multimodal data features using a Gaussian graphical model (GGM). We then propose a modularity-based method that can be applied to the GGM to identify links that are associated with mental illness across a multimodal data set. Through simulation and real data, we show our approach reveals important information about disease-related network disruptions that are missed with a focus on a single modality. We use functional MRI (fMRI), diffusion MRI (dMRI), and structural MRI (sMRI) to compute the fractional amplitude of low frequency fluctuations (fALFF), fractional anisotropy (FA), and gray matter (GM) concentration maps. These three modalities are analyzed using our modularity method. RESULTS Our results show missing links that are only captured by the cross-modal information that may play an important role in disconnectivity between the components. CONCLUSION We identified multimodal (fALFF, FA and GM) disconnectivity in the default mode network area in patients with SZ, which would not have been detectable in a single modality. SIGNIFICANCE The proposed approach provides an important new tool for capturing information that is distributed among multiple imaging modalities.
Collapse
|
18
|
Prasad H, Rao R. Endosomal Acid-Base Homeostasis in Neurodegenerative Diseases. Rev Physiol Biochem Pharmacol 2020; 185:195-231. [PMID: 32737755 PMCID: PMC7614123 DOI: 10.1007/112_2020_25] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neurodegenerative disorders are debilitating and largely untreatable conditions that pose a significant burden to affected individuals and caregivers. Overwhelming evidence supports a crucial preclinical role for endosomal dysfunction as an upstream pathogenic hub and driver in Alzheimer's disease (AD) and related neurodegenerative disorders. We present recent advances on the role of endosomal acid-base homeostasis in neurodegeneration and discuss evidence for converging mechanisms. The strongest genetic risk factor in sporadic AD is the ε4 allele of Apolipoprotein E (ApoE4), which potentiates pre-symptomatic endosomal dysfunction and prominent amyloid beta (Aβ) pathology, although how these pathways are linked mechanistically has remained unclear. There is emerging evidence that the Christianson syndrome protein NHE6 is a prominent ApoE4 effector linking endosomal function to Aβ pathologies. By functioning as a dominant leak pathway for protons, the Na+/H+ exchanger activity of NHE6 limits endosomal acidification and regulates β-secretase (BACE)-mediated Aβ production and LRP1 receptor-mediated Aβ clearance. Pathological endosomal acidification may impact both Aβ generation and clearance mechanisms and emerges as a promising therapeutic target in AD. We also offer our perspective on the complex role of endosomal acid-base homeostasis in the pathogenesis of neurodegeneration and its therapeutic implications for neuronal rescue and repair strategies.
Collapse
Affiliation(s)
- Hari Prasad
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India, Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Zhao K, Ding Y, Han Y, Fan Y, Alexander-Bloch AF, Han T, Jin D, Liu B, Lu J, Song C, Wang P, Wang D, Wang Q, Xu K, Yang H, Yao H, Zheng Y, Yu C, Zhou B, Zhang X, Zhou Y, Jiang T, Zhang X, Liu Y. Independent and reproducible hippocampal radiomic biomarkers for multisite Alzheimer's disease: diagnosis, longitudinal progress and biological basis. Sci Bull (Beijing) 2020; 65:1103-1113. [PMID: 36659162 DOI: 10.1016/j.scib.2020.04.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/31/2020] [Accepted: 03/17/2020] [Indexed: 01/21/2023]
Abstract
Hippocampal morphological change is one of the main hallmarks of Alzheimer's disease (AD). However, whether hippocampal radiomic features are robust as predictors of progression from mild cognitive impairment (MCI) to AD dementia and whether these features provide any neurobiological foundation remains unclear. The primary aim of this study was to verify whether hippocampal radiomic features can serve as robust magnetic resonance imaging (MRI) markers for AD. Multivariate classifier-based support vector machine (SVM) analysis provided individual-level predictions for distinguishing AD patients (n = 261) from normal controls (NCs; n = 231) with an accuracy of 88.21% and intersite cross-validation. Further analyses of a large, independent the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset (n = 1228) reinforced these findings. In MCI groups, a systemic analysis demonstrated that the identified features were significantly associated with clinical features (e.g., apolipoprotein E (APOE) genotype, polygenic risk scores, cerebrospinal fluid (CSF) Aβ, CSF Tau), and longitudinal changes in cognition ability; more importantly, the radiomic features had a consistently altered pattern with changes in the MMSE scores over 5 years of follow-up. These comprehensive results suggest that hippocampal radiomic features can serve as robust biomarkers for clinical application in AD/MCI, and further provide evidence for predicting whether an MCI subject would convert to AD based on the radiomics of the hippocampus. The results of this study are expected to have a substantial impact on the early diagnosis of AD/MCI.
Collapse
Affiliation(s)
- Kun Zhao
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; School of Information Science and Engineering, Shandong Normal University, Ji'nan 250358, China
| | - Yanhui Ding
- School of Information Science and Engineering, Shandong Normal University, Ji'nan 250358, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing 100069, China; Beijing Institute of Geriatrics, Beijing 100053, China; National Clinical Research Center for Geriatric Disorders, Beijing 100053, China
| | - Yong Fan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Dan Jin
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Liu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Chengyuan Song
- Department of Neurology, Qilu Hospital of Shandong University, Ji'nan 250012, China
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin University, Tianjin 300350, China; Department of Neurology, The Secondary Medical Center, National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing 100853, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Ji'nan 250012, China
| | - Qing Wang
- Department of Radiology, Qilu Hospital of Shandong University, Ji'nan 250012, China
| | - Kaibin Xu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Hongwei Yang
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Hongxiang Yao
- Department of Radiology, The Secondary Medical Center, National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing 100853, China
| | - Yuanjie Zheng
- School of Information Science and Engineering, Shandong Normal University, Ji'nan 250358, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Bo Zhou
- Department of Neurology, The Secondary Medical Center, National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing 100853, China
| | - Xinqing Zhang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yuying Zhou
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin University, Tianjin 300350, China
| | - Tianzi Jiang
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xi Zhang
- Department of Neurology, The Secondary Medical Center, National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing 100853, China.
| | - Yong Liu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
20
|
Endocytic regulation of cellular ion homeostasis controls lysosome biogenesis. Nat Cell Biol 2020; 22:815-827. [PMID: 32601373 DOI: 10.1038/s41556-020-0535-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/21/2020] [Indexed: 12/24/2022]
Abstract
Lysosomes serve as cellular degradation and signalling centres that coordinate metabolism in response to intracellular cues and extracellular signals. Lysosomal capacity is adapted to cellular needs by transcription factors, such as TFEB and TFE3, which activate the expression of lysosomal and autophagy genes. Nuclear translocation and activation of TFEB are induced by a variety of conditions such as starvation, lysosome stress and lysosomal storage disorders. How these various cues are integrated remains incompletely understood. Here, we describe a pathway initiated at the plasma membrane that controls lysosome biogenesis via the endocytic regulation of intracellular ion homeostasis. This pathway is based on the exo-endocytosis of NHE7, a Na+/H+ exchanger mutated in X-linked intellectual disability, and serves to control intracellular ion homeostasis and thereby Ca2+/calcineurin-mediated activation of TFEB and downstream lysosome biogenesis in response to osmotic stress to promote the turnover of toxic proteins and cell survival.
Collapse
|
21
|
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: 20.5] [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
|
22
|
Pedersen SF, Counillon L. The SLC9A-C Mammalian Na +/H + Exchanger Family: Molecules, Mechanisms, and Physiology. Physiol Rev 2019; 99:2015-2113. [PMID: 31507243 DOI: 10.1152/physrev.00028.2018] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Na+/H+ exchangers play pivotal roles in the control of cell and tissue pH by mediating the electroneutral exchange of Na+ and H+ across cellular membranes. They belong to an ancient family of highly evolutionarily conserved proteins, and they play essential physiological roles in all phyla. In this review, we focus on the mammalian Na+/H+ exchangers (NHEs), the solute carrier (SLC) 9 family. This family of electroneutral transporters constitutes three branches: SLC9A, -B, and -C. Within these, each isoform exhibits distinct tissue expression profiles, regulation, and physiological roles. Some of these transporters are highly studied, with hundreds of original articles, and some are still only rudimentarily understood. In this review, we present and discuss the pioneering original work as well as the current state-of-the-art research on mammalian NHEs. We aim to provide the reader with a comprehensive view of core knowledge and recent insights into each family member, from gene organization over protein structure and regulation to physiological and pathophysiological roles. Particular attention is given to the integrated physiology of NHEs in the main organ systems. We provide several novel analyses and useful overviews, and we pinpoint main remaining enigmas, which we hope will inspire novel research on these highly versatile proteins.
Collapse
Affiliation(s)
- S F Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; and Université Côte d'Azur, CNRS, Laboratoire de Physiomédecine Moléculaire, LP2M, France, and Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| | - L Counillon
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; and Université Côte d'Azur, CNRS, Laboratoire de Physiomédecine Moléculaire, LP2M, France, and Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| |
Collapse
|
23
|
|
24
|
Distinct structural brain circuits indicate mood and apathy profiles in bipolar disorder. NEUROIMAGE-CLINICAL 2019; 26:101989. [PMID: 31451406 PMCID: PMC7229320 DOI: 10.1016/j.nicl.2019.101989] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/01/2019] [Accepted: 08/16/2019] [Indexed: 11/22/2022]
Abstract
Bipolar disorder (BD) is a severe manic-depressive illness. Patients with BD have been shown to have gray matter (GM) deficits in prefrontal, frontal, parietal, and temporal regions; however, the relationship between structural effects and clinical profiles has proved elusive when considered on a region by region or voxel by voxel basis. In this study, we applied parallel independent component analysis (pICA) to structural neuroimaging measures and the positive and negative syndrome scale (PANSS) in 110 patients (mean age 34.9 ± 11.65) with bipolar disorder, to examine networks of brain regions that relate to symptom profiles. The pICA revealed two distinct symptom profiles and associated GM concentration alteration circuits. The first PANSS pICA profile mainly involved anxiety, depression and guilty feelings, reflecting mood symptoms. Reduced GM concentration in right temporal regions predicted worse mood symptoms in this profile. The second PANSS pICA profile generally covered blunted affect, emotional withdrawal, passive/apathetic social withdrawal, depression and active social avoidance, exhibiting a withdrawal or apathy dominating component. Lower GM concentration in bilateral parietal and frontal regions showed worse symptom severity in this profile. In summary, a pICA decomposition suggested BD patients showed distinct mood and apathy profiles differing from the original PANSS subscales, relating to distinct brain structural networks. Structural relationships with symptoms in bipolar disorder are complex. A parallel ICA analysis of PANSS questions and structural images finds two correlated profiles. The first pair links mood symptoms with right temporal regions. The second pair highlights social withdrawal and apathy symptoms linked to bilateral frontal and parietal regions.
Collapse
|
25
|
Cosín-Tomàs M, Senserrich J, Arumí-Planas M, Alquézar C, Pallàs M, Martín-Requero Á, Suñol C, Kaliman P, Sanfeliu C. Role of Resveratrol and Selenium on Oxidative Stress and Expression of Antioxidant and Anti-Aging Genes in Immortalized Lymphocytes from Alzheimer's Disease Patients. Nutrients 2019; 11:E1764. [PMID: 31370365 PMCID: PMC6723840 DOI: 10.3390/nu11081764] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/24/2019] [Accepted: 07/28/2019] [Indexed: 12/29/2022] Open
Abstract
Oxidative damage is involved in the pathophysiology of age-related ailments, including Alzheimer's disease (AD). Studies have shown that the brain tissue and also lymphocytes from AD patients present increased oxidative stress compared to healthy controls (HCs). Here, we use lymphoblastoid cell lines (LCLs) from AD patients and HCs to investigate the role of resveratrol (RV) and selenium (Se) in the reduction of reactive oxygen species (ROS) generated after an oxidative injury. We also studied whether these compounds elicited expression changes in genes involved in the antioxidant cell response and other aging-related mechanisms. AD LCLs showed higher ROS levels than those from HCs in response to H2O2 and FeSO4 oxidative insults. RV triggered a protective response against ROS under control and oxidizing conditions, whereas Se exerted antioxidant effects only in AD LCLs under oxidizing conditions. RV increased the expression of genes encoding known antioxidants (catalase, copper chaperone for superoxide dismutase 1, glutathione S-transferase zeta 1) and anti-aging factors (sirtuin 1 and sirtuin 3) in both AD and HC LCLs. Our findings support RV as a candidate for inducing resilience and protection against AD, and reinforce the value of LCLs as a feasible peripheral cell model for understanding the protective mechanisms of nutraceuticals against oxidative stress in aging and AD.
Collapse
Affiliation(s)
- Marta Cosín-Tomàs
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
- Department of Human Genetics, Research Institute of the McGill University Health Centre, Montreal, QC H3A 0C7, Canada
| | - Júlia Senserrich
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
| | - Marta Arumí-Planas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
| | - Carolina Alquézar
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, 28040 Madrid, Spain
| | - Mercè Pallàs
- Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, 08028 Barcelona, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Ángeles Martín-Requero
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, 28040 Madrid, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Cristina Suñol
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Perla Kaliman
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain
- Faculty of Health Sciences, Universitat Oberta de Catalunya, 08018 Barcelona, Spain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain.
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28031 Madrid, Spain.
- Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.
| |
Collapse
|
26
|
Sun N, Mormino EC, Chen J, Sabuncu MR, Yeo BTT. Multi-modal latent factor exploration of atrophy, cognitive and tau heterogeneity in Alzheimer's disease. Neuroimage 2019; 201:116043. [PMID: 31344486 DOI: 10.1016/j.neuroimage.2019.116043] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 06/23/2019] [Accepted: 07/21/2019] [Indexed: 12/22/2022] Open
Abstract
Individuals with Alzheimer's disease (AD) dementia exhibit significant heterogeneity across clinical symptoms, atrophy patterns, and spatial distribution of Tau deposition. Most previous studies of AD heterogeneity have focused on atypical clinical subtypes, defined subtypes with a single modality, or restricted their analyses to a priori brain regions and cognitive tests. Here, we considered a data-driven hierarchical Bayesian model to identify latent factors from atrophy patterns and cognitive deficits simultaneously, thus exploiting the rich dimensionality within each modality. Unlike most previous studies, our model allows each factor to be expressed to varying degrees within an individual, in order to reflect potential multiple co-existing pathologies. By applying our model to ADNI-GO/2 AD dementia participants, we found three atrophy-cognitive factors. The first factor was associated with medial temporal lobe atrophy, episodic memory deficits and disorientation to time/place ("MTL-Memory"). The second factor was associated with lateral temporal atrophy and language deficits ("Lateral Temporal-Language"). The third factor was associated with atrophy in posterior bilateral cortex, and visuospatial executive function deficits ("Posterior Cortical-Executive"). While the MTL-Memory and Posterior Cortical-Executive factors were discussed in previous literature, the Lateral Temporal-Language factor is novel and emerged only by considering atrophy and cognition jointly. Several analyses were performed to ensure generalizability, replicability and stability of the estimated factors. First, the factors generalized to new participants within a 10-fold cross-validation of ADNI-GO/2 AD dementia participants. Second, the factors were replicated in an independent ADNI-1 AD dementia cohort. Third, factor loadings of ADNI-GO/2 AD dementia participants were longitudinally stable, suggesting that these factors capture heterogeneity across patients, rather than longitudinal disease progression. Fourth, the model outperformed canonical correlation analysis at capturing associations between atrophy patterns and cognitive deficits. To explore the influence of the factors early in the disease process, factor loadings were estimated in ADNI-GO/2 mild cognitively impaired (MCI) participants. Although the associations between the atrophy patterns and cognitive profiles were weak in MCI compared to AD, we found that factor loadings were associated with inter-individual regional variation in Tau uptake. Taken together, these results suggest that distinct atrophy-cognitive patterns exist in typical Alzheimer's disease, and are associated with distinct patterns of Tau depositions before clinical dementia emerges.
Collapse
Affiliation(s)
- Nanbo Sun
- Department of Electrical and Computer Engineering, NUS Clinical Imaging Research Centre, N.1 Institute for Health and Memory Networks Program, National University of Singapore, Singapore
| | | | - Jianzhong Chen
- Department of Electrical and Computer Engineering, NUS Clinical Imaging Research Centre, N.1 Institute for Health and Memory Networks Program, National University of Singapore, Singapore
| | - Mert R Sabuncu
- School of Electrical and Computer Engineering, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - B T Thomas Yeo
- Department of Electrical and Computer Engineering, NUS Clinical Imaging Research Centre, N.1 Institute for Health and Memory Networks Program, National University of Singapore, Singapore; Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Center for Cognitive Neuroscience, Duke-NUS Graduate Medical School, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.
| |
Collapse
|
27
|
Li Y, Yao Z, Yu Y, Zou Y, Fu Y, Hu B. Brain network alterations in individuals with and without mild cognitive impairment: parallel independent component analysis of AV1451 and AV45 positron emission tomography. BMC Psychiatry 2019; 19:165. [PMID: 31159754 PMCID: PMC6547610 DOI: 10.1186/s12888-019-2149-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/17/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Amyloid β (Aβ) and tau proteins are considered as critical factors that affect Alzheimer's disease (AD) and mild cognitive impairment (MCI). Although many studies have conducted on these two proteins, little study has investigated the relationship between their spatial distributions. This study aims to explore the associations of spatial patterns between Aβ deposition and tau deposition in patients with MCI and normal control (NC). METHODS We used multimodality positron emission tomography (PET) data from a clinically heterogeneous population of patients with MCI and NC. All data were obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database containing information of 65 patients with MCI and 75 NC who both had undergone AV45 (Aβ) and AV1451 (tau) PET. To assess the spatial distribution of Aβ and tau deposition, we employed parallel independent component analysis (pICA), which enabled the joint analysis of multimodal imaging data. pICA was conducted to identify the significant difference and correlation relationship of brain networks between Aβ PET and tau PET in MCI and NC groups. RESULTS Our results revealed the strongly correlated network between Aβ PET and tau PET were colocalized with the default-mode network (DMN). Simultaneously, in comparison of the spatial distribution between Aβ PET and tau PET, it was found that the significant differences between MCI and NC were mainly distributed in DMN, cognitive control network and visual networks. The altered brain networks obtained from pICA analysis are consistent with the abnormalities of brain network in MCI patients. CONCLUSIONS Findings suggested the abnormal spatial distribution regions of tau PET were correlated with the abnormal spatial distribution regions of Aβ PET, and both of which were located in DMN network. This study revealed that combining pICA with multimodal imaging data is an effective approach for distinguishing MCI patients from NC group.
Collapse
Affiliation(s)
- Yuan Li
- grid.410585.dSchool of Information Science and Engineering, Shandong Normal University, Jinan, Shandong Province 250358 People’s Republic of China
| | - Zhijun Yao
- 0000 0000 8571 0482grid.32566.34School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province China
| | - Yue Yu
- 0000 0000 8571 0482grid.32566.34School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province China
| | - Ying Zou
- 0000 0000 8571 0482grid.32566.34School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province China
| | - Yu Fu
- 0000 0000 8571 0482grid.32566.34School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province China
| | - Bin Hu
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong Province, 250358, People's Republic of China. .,School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China.
| | | |
Collapse
|
28
|
Matsuda S, Kakegawa W, Yuzaki M. PhotonSABER: new tool shedding light on endocytosis and learning mechanisms in vivo. Commun Integr Biol 2019; 12:34-37. [PMID: 31143361 PMCID: PMC6527187 DOI: 10.1080/19420889.2019.1586048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 11/01/2022] Open
Abstract
In the central nervous system, activity-dependent endocytosis of postsynaptic AMPA-type glutamate receptors (AMPA receptors) is thought to mediate long-term depression (LTD), which is a synaptic plasticity model in various neuronal circuits. However, whether and how AMPA receptor endocytosis and LTD at specific synapses are causally linked to learning and memory in vivo remains unclear. Recently, we developed a new optogenetic tool, PhotonSABER, which could control AMPA receptor endocytosis in temporal, spatial, and cell-type-specific manners at activated synapses. Using PhotonSABER, we found that AMPA receptor endocytosis and LTD at synapses between parallel fibers and Purkinje cells in the cerebellum mediate oculomotor learning. We also found that PhotonSABER could inhibit endocytosis of epidermal growth factor receptors in HeLa cells upon light stimulation. These results demonstrate that PhotonSABER is a powerful tool for analyzing the physiological functions of endocytosis in non-neuronal cells, as well as the roles of LTD in various brain regions.
Collapse
Affiliation(s)
- Shinji Matsuda
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan.,Brain Science Inspired Life Support Research Center (BLSC), The University of Electro-Communications, Tokyo, Japan
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Muñoz-Braceras S, Tornero-Écija AR, Vincent O, Escalante R. VPS13A is closely associated with mitochondria and is required for efficient lysosomal degradation. Dis Model Mech 2019; 12:dmm036681. [PMID: 30709847 PMCID: PMC6398486 DOI: 10.1242/dmm.036681] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
Members of the VPS13 family are associated with various human diseases. In particular, the loss of function of VPS13A leads to chorea-acanthocytosis (ChAc), a rare neurodegenerative disease without available curative treatments. Autophagy has been considered a promising therapeutic target because the absence of VPS13A causes a defective autophagy flux. However, the mechanistic details of this deficiency are unknown. Here, we identified Rab7A as an interactor of one of the VPS13 family members in Dictyostelium discoideum and showed that this interaction is conserved between the human homologs VPS13A and RAB7A in HeLa cells. As RAB7A is a key player in endosome trafficking, we addressed the possible function of VPS13A in endosome dynamics and lysosome degradation. Our results suggest that the decrease in autophagy observed in the absence of VPS13A may be the result of a more general defect in endocytic trafficking and lysosomal degradation. Unexpectedly, we found that VPS13A is closely localized to mitochondria, suggesting that the role of VPS13A in the endolysosomal pathway might be related to inter-organelle communication. We show that VPS13A localizes at the interface between mitochondria-endosomes and mitochondria-endoplasmic reticulum and that the presence of membrane contact sites is altered in the absence of VPS13A. Based on these findings, we propose that therapeutic strategies aimed at modulating the endolysosomal pathway could be beneficial in the treatment of ChAc.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sandra Muñoz-Braceras
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Alba R Tornero-Écija
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Olivier Vincent
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Ricardo Escalante
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| |
Collapse
|
30
|
de la Monte SM. The Full Spectrum of Alzheimer's Disease Is Rooted in Metabolic Derangements That Drive Type 3 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:45-83. [PMID: 31062325 PMCID: PMC9996398 DOI: 10.1007/978-981-13-3540-2_4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The standard practice in neuropathology is to diagnose Alzheimer's disease (AD) based on the distribution and abundance of neurofibrillary tangles and Aβ deposits. However, other significant abnormalities including neuroinflammation, gliosis, white matter degeneration, non-Aβ microvascular disease, and insulin-related metabolic dysfunction require further study to understand how they could be targeted to more effectively remediate AD. This review addresses non-Aβ and non-pTau AD-associated pathologies, highlighting their major features, roles in neurodegeneration, and etiopathic links to deficits in brain insulin and insulin-like growth factor signaling and cognitive impairment. The discussion delineates why AD with its most characteristic clinical and pathological phenotypic profiles should be regarded as a brain form of diabetes, i.e., type 3 diabetes, and entertains the hypothesis that type 3 diabetes is just one of the categories of insulin resistance diseases that can occur independently or overlap with one or more of the others, including type 2 diabetes, metabolic syndrome, and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Departments of Neurology, Neuropathology, and Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology and Laboratory Medicine, Providence VA Medical Center, Providence, RI, USA.
| |
Collapse
|
31
|
Pan J, Sawyer K, McDonough E, Slotpole L, Gansler D. Cognitive, Neuroanatomical, and Genetic Predictors of Executive Function in Healthy Children and Adolescents. Dev Neuropsychol 2018; 43:535-550. [PMID: 30216102 DOI: 10.1080/87565641.2018.1516770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The Dimensional Change Card Sort (DCCS) is a measure of cognitive flexibility for children, which requires rule-use and shifting. Demographic, cognitive, regional cortical thickness, and genetic variables, including those related to language and executive function, were used to build predictive models of DCCS scores in 556 healthy pediatric participants. Gender, age, frontal, and temporal lobe regions of interest, and measures of sustained attention, inhibition, and word reading were selected as the best predictors of DCCS performance. Results indicated that DCCS performance is related to a broad range of cognitive functions and anatomic regions associated with various levels of cognitive function.
Collapse
Affiliation(s)
- Jessica Pan
- a Department of Psychology , Suffolk University , Boston , MA , USA
| | - Kayle Sawyer
- b Boston VA Healthcare System , Boston , MA , USA.,c Department of Anatomy and Neurobiology , Boston University School of Medicine , Boston , MA , USA.,d Sawyer Scientific, LLC , Boston , MA , USA.,e Athinoula A. Martinos Center for Biomedical Imaging , Massachusetts General Hospital , Boston , MA , USA
| | - EmilyKate McDonough
- d Sawyer Scientific, LLC , Boston , MA , USA.,f Medical Education , Tufts University , Boston , MA , USA
| | - Laura Slotpole
- g Department of Psychology , Dickinson College , Carlisle , PA , USA
| | - David Gansler
- a Department of Psychology , Suffolk University , Boston , MA , USA
| |
Collapse
|
32
|
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: 1.2] [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
|
33
|
Gershon ES, Pearlson G, Keshavan MS, Tamminga C, Clementz B, Buckley PF, Alliey-Rodriguez N, Liu C, Sweeney JA, Keedy S, Meda SA, Tandon N, Shafee R, Bishop JR, Ivleva EI. Genetic analysis of deep phenotyping projects in common disorders. Schizophr Res 2018; 195:51-57. [PMID: 29056493 PMCID: PMC5910299 DOI: 10.1016/j.schres.2017.09.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/19/2017] [Accepted: 09/22/2017] [Indexed: 11/19/2022]
Abstract
Several studies of complex psychotic disorders with large numbers of neurobiological phenotypes are currently under way, in living patients and controls, and on assemblies of brain specimens. Genetic analyses of such data typically present challenges, because of the choice of underlying hypotheses on genetic architecture of the studied disorders and phenotypes, large numbers of phenotypes, the appropriate multiple testing corrections, limited numbers of subjects, imputations required on missing phenotypes and genotypes, and the cross-disciplinary nature of the phenotype measures. Advances in genotype and phenotype imputation, and in genome-wide association (GWAS) methods, are useful in dealing with these challenges. As compared with the more traditional single-trait analyses, deep phenotyping with simultaneous genome-wide analyses serves as a discovery tool for previously unsuspected relationships of phenotypic traits with each other, and with specific molecular involvements.
Collapse
Affiliation(s)
- Elliot S Gershon
- Department of Psychiatry, Department of Human Genetics, University of Chicago, United States.
| | - Godfrey Pearlson
- Yale University Departments of Psychiatry & Neuroscience, Hartford, CT, United States; Olin Neuropsychiatry Research Center, Institute of Living, Hartford, Connecticut, USA
| | | | - Carol Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Brett Clementz
- Department of Psychology, University of Georgia, Athens, GA, United States
| | - Peter F Buckley
- School of Medicine Virginia Commonwealth University (VCU), Richmond, VA, United States
| | - Ney Alliey-Rodriguez
- University of Chicago, Department of Psychiatry and Behavioral Neurosciences, Chicago, IL, United States
| | - Chunyu Liu
- University of Illinois at Chicago, Chicago, IL, United States
| | - John A Sweeney
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States; University of Cincinnati, Department of Psychiatry and Behavioral Neuroscience, Cincinnati, OH, United States
| | - Sarah Keedy
- University of Chicago, Department of Psychiatry and Behavioral Neurosciences, Chicago, IL, United States
| | - Shashwath A Meda
- Yale University Departments of Psychiatry & Neuroscience, Hartford, CT, United States
| | - Neeraj Tandon
- Beth Israel Deaconess Medical Center, Dept of Psychiatry, Harvard Medical School, United States
| | - Rebecca Shafee
- Broad Institute of MIT and Harvard, Cambridge, MA, United States; Department of Genetics, Harvard Medical School, United States
| | - Jeffrey R Bishop
- Department of Clinical and Experimental Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Elena I Ivleva
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
34
|
Abstract
Alzheimer's disease (AD), the main form of dementia in the elderly, is the most common progressive neurodegenerative disease characterized by rapidly progressive cognitive dysfunction and behavior impairment. AD exhibits a considerable heritability and great advances have been made in approaches to searching the genetic etiology of AD. In AD genetic studies, methods have developed from classic linkage-based and candidate-gene-based association studies to genome-wide association studies (GWAS) and next generation sequencing (NGS). The identification of new susceptibility genes has provided deeper insights to understand the mechanisms underlying AD. In addition to searching novel genes associated with AD in large samples, the NGS technologies can also be used to shed light on the 'black matter' discovery even in smaller samples. The shift in AD genetics between traditional studies and individual sequencing will allow biomaterials of each patient as the central unit of genetic studies. This review will cover genetic findings in AD and consequences of AD genetic findings. Firstly, we will discuss the discovery of mutations in APP, PSEN1, PSEN2, APOE, and ADAM10. Then we will summarize and evaluate the information obtained from GWAS of AD. Finally, we will outline the efforts to identify rare variants associated with AD using NGS.
Collapse
|
35
|
Brain Network Alterations in Alzheimer's Disease Identified by Early-Phase PIB-PET. CONTRAST MEDIA & MOLECULAR IMAGING 2018. [PMID: 29531506 PMCID: PMC5817202 DOI: 10.1155/2018/6830105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The aim of this study was to identify the brain networks from early-phase 11C-PIB (perfusion PIB, pPIB) data and to compare the brain networks of patients with differentiating Alzheimer's disease (AD) with cognitively normal subjects (CN) and of mild cognitively impaired patients (MCI) with CN. Forty participants (14 CN, 12 MCI, and 14 AD) underwent 11C-PIB and 18F-FDG PET/CT scans. Parallel independent component analysis (pICA) was used to identify correlated brain networks from the 11C-pPIB and 18F-FDG data, and a two-sample t-test was used to evaluate group differences in the corrected brain networks between AD and CN, and between MCI and CN. Our study identified a brain network of perfusion (early-phase 11C-PIB) that highly correlated with a glucose metabolism (18F-FDG) brain network and colocalized with the default mode network (DMN) in an AD-specific neurodegenerative cohort. Particularly, decreased 18F-FDG uptake correlated with a decreased regional cerebral blood flow in the frontal, parietal, and temporal regions of the DMN. The group comparisons revealed similar spatial patterns of the brain networks derived from the 11C-pPIB and 18F-FDG data. Our findings indicate that 11C-pPIB derived from the early-phase 11C-PIB could provide complementary information for 18F-FDG examination in AD.
Collapse
|
36
|
Szefer E, Lu D, Nathoo F, Beg MF, Graham J. Multivariate association between single-nucleotide polymorphisms in Alzgene linkage regions and structural changes in the brain: discovery, refinement and validation. Stat Appl Genet Mol Biol 2017; 16:349-365. [PMID: 29091582 PMCID: PMC9008768 DOI: 10.1515/sagmb-2016-0077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
AbstractUsing publicly-available data from the Alzheimer’s Disease Neuroimaging Initiative, we investigate the joint association between single-nucleotide polymorphisms (SNPs) in previously established linkage regions for Alzheimer’s disease (AD) and rates of decline in brain structure. In an initial, discovery stage of analysis, we applied a weighted
Collapse
Affiliation(s)
- Elena Szefer
- Department of Statistics and Actuarial Science, Simon Fraser University, 8888 University Dr, Burnaby, BC V5A 1S6, Canada
| | - Donghuan Lu
- School of Engineering Science, Simon Fraser University, 8888 University Dr, Burnaby, BC V5A 1S6, Canada
| | - Farouk Nathoo
- Department of Mathematics and Statistics, University of Victoria, PO Box 1700 STN CSC Victoria, BC V8W 2Y2, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, 8888 University Dr, Burnaby, BC V5A 1S6, Canada
| | - Jinko Graham
- Corresponding author: Jinko Graham, Department of Statistics and Actuarial Science, Simon Fraser University, 8888 University Dr, Burnaby, BC V5A 1S6, Canada,
| | | |
Collapse
|
37
|
Guo X, Qiu W, Garcia-Milian R, Lin X, Zhang Y, Cao Y, Tan Y, Wang Z, Shi J, Wang J, Liu D, Song L, Xu Y, Wang X, Liu N, Sun T, Zheng J, Luo J, Zhang H, Xu J, Kang L, Ma C, Wang K, Luo X. Genome-wide significant, replicated and functional risk variants for Alzheimer's disease. J Neural Transm (Vienna) 2017; 124:1455-1471. [PMID: 28770390 PMCID: PMC5654670 DOI: 10.1007/s00702-017-1773-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023]
Abstract
Genome-wide association studies (GWASs) have reported numerous associations between risk variants and Alzheimer's disease (AD). However, these associations do not necessarily indicate a causal relationship. If the risk variants can be demonstrated to be biologically functional, the possibility of a causal relationship would be increased. In this article, we reviewed all of the published GWASs to extract the genome-wide significant (p < 5×10-8) and replicated associations between risk variants and AD or AD-biomarkers. The regulatory effects of these risk variants on the expression of a novel class of non-coding RNAs (piRNAs) and protein-coding RNAs (mRNAs), the alteration of proteins caused by these variants, the associations between AD and these variants in our own sample, the expression of piRNAs, mRNAs and proteins in human brains targeted by these variants, the expression correlations between the risk genes and APOE, the pathways and networks that the risk genes belonged to, and the possible long non-coding RNAs (LncRNAs) that might regulate the risk genes were analyzed, to investigate the potential biological functions of the risk variants and explore the potential mechanisms underlying the SNP-AD associations. We found replicated and significant associations for AD or AD-biomarkers, surprisingly, only at 17 SNPs located in 11 genes/snRNAs/LncRNAs in eight genomic regions. Most of these 17 SNPs enriched some AD-related pathways or networks, and were potentially functional in regulating piRNAs and mRNAs; some SNPs were associated with AD in our sample, and some SNPs altered protein structures. Most of the protein-coding genes regulated by the risk SNPs were expressed in human brain and correlated with APOE expression. We conclude that these variants were most robust risk markers for AD, and their contributions to AD risk was likely to be causal. As expected, APOE and the lipoprotein metabolism pathway possess the highest weight among these contributions.
Collapse
Affiliation(s)
- Xiaoyun Guo
- Shanghai Mental Health Center, Shanghai 200030, China
- Department of Psychiatry, Yale University School of Medicine, New
Haven, CT 06510, USA
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of
Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences,
School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Rolando Garcia-Milian
- Curriculum & Research Support Department, Cushing/Whitney
Medical Library, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xiandong Lin
- Department of Pathology, Fujian Provincial Cancer Hospital, the
Teaching Hospital of Fujian Medical University, Fuzhou, Fujian 350014, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin 300222, China
| | - Yuping Cao
- Department of Psychiatry, Second Xiangya Hospital, Central South
University, Changsha 410012, China
| | - Yunlong Tan
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| | - Zhiren Wang
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| | - Jing Shi
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| | - Jijun Wang
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Dengtang Liu
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Lisheng Song
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Yifeng Xu
- Shanghai Mental Health Center, Shanghai 200030, China
| | - Xiaoping Wang
- Department of Neurology, Shanghai Tongren Hospital, Shanghai Jiao
Tong University, Shanghai 200080, China
| | - Na Liu
- Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029,
China
| | - Tao Sun
- Huashan Hospital, Fudan University School of Medicine, Shanghai
200040, China
| | - Jianming Zheng
- Huashan Hospital, Fudan University School of Medicine, Shanghai
200040, China
| | - Justine Luo
- Department of Psychiatry, Yale University School of Medicine, New
Haven, CT 06510, USA
| | - Huihao Zhang
- The First Affiliated Hospital, Fujian Medical University, Fuzhou
350001, China
| | - Jianying Xu
- Zhuhai Municipal Maternal and Children’s Health Hospital,
Zhuhai, Guangdong 519000, China
| | - Longli Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention
Research on High Altitude Diseases of Tibet Autonomous Region, Xizang Minzu
University School of Medicine, Xiangyang, Shaanxi 712082, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of
Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences,
School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Kesheng Wang
- Department of Biostatistics and Epidemiology, College of Public
Health, East Tennessee State University, Johnson City, TN 37614, USA
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New
Haven, CT 06510, USA
- Biological Psychiatry Research Center, Beijing Huilongguan Hospital,
Beijing 100096, China
| |
Collapse
|
38
|
Pattern Discovery in Brain Imaging Genetics via SCCA Modeling with a Generic Non-convex Penalty. Sci Rep 2017; 7:14052. [PMID: 29070790 PMCID: PMC5656688 DOI: 10.1038/s41598-017-13930-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/02/2017] [Indexed: 01/21/2023] Open
Abstract
Brain imaging genetics intends to uncover associations between genetic markers and neuroimaging quantitative traits. Sparse canonical correlation analysis (SCCA) can discover bi-multivariate associations and select relevant features, and is becoming popular in imaging genetic studies. The L1-norm function is not only convex, but also singular at the origin, which is a necessary condition for sparsity. Thus most SCCA methods impose [Formula: see text]-norm onto the individual feature or the structure level of features to pursuit corresponding sparsity. However, the [Formula: see text]-norm penalty over-penalizes large coefficients and may incurs estimation bias. A number of non-convex penalties are proposed to reduce the estimation bias in regression tasks. But using them in SCCA remains largely unexplored. In this paper, we design a unified non-convex SCCA model, based on seven non-convex functions, for unbiased estimation and stable feature selection simultaneously. We also propose an efficient optimization algorithm. The proposed method obtains both higher correlation coefficients and better canonical loading patterns. Specifically, these SCCA methods with non-convex penalties discover a strong association between the APOE e4 rs429358 SNP and the hippocampus region of the brain. They both are Alzheimer's disease related biomarkers, indicating the potential and power of the non-convex methods in brain imaging genetics.
Collapse
|
39
|
Qiu W, Guo X, Lin X, Yang Q, Zhang W, Zhang Y, Zuo L, Zhu Y, Li CSR, Ma C, Luo X. Transcriptome-wide piRNA profiling in human brains of Alzheimer's disease. Neurobiol Aging 2017; 57:170-177. [PMID: 28654860 DOI: 10.1016/j.neurobiolaging.2017.05.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/21/2017] [Accepted: 05/26/2017] [Indexed: 01/03/2023]
Abstract
Discovered in the brains of multiple animal species, piRNAs may contribute to the pathogenesis of neuropsychiatric illnesses. The present study aimed to identify brain piRNAs across transcriptome that are associated with Alzheimer's disease (AD). Prefrontal cortical tissues of 6 AD cases and 6 controls were examined for piRNA expression levels using an Arraystar HG19 piRNA array (containing 23,677 piRNAs) and genotyped for 17 genome-wide significant and replicated risk SNPs. We examined whether piRNAs are expressed differently between AD cases and controls and explored the potential regulatory effects of risk SNPs on piRNA expression levels. We identified a total of 9453 piRNAs in human brains, with 103 nominally (p < 0.05) differentially (>1.5 fold) expressed in AD cases versus controls and most of the 103 piRNAs nominally correlated with genome-wide significant risk SNPs. We conclude that piRNAs are abundant in human brains and may represent risk biomarkers of AD.
Collapse
Affiliation(s)
- Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoyun Guo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Shanghai Mental Health Center, Shanghai, China
| | - Xiandong Lin
- Department of Pathology, Fujian Provincial Cancer Hospital, the Teaching Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Qian Yang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wanying Zhang
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yong Zhang
- Tianjin Mental Health Center, Tianjin, China
| | - Lingjun Zuo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yong Zhu
- Department of Environmental Health Sciences, Yale University School of Public Health, New Haven, CT, USA
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Biological Psychiatry Research Center, Beijing Huilongguan Hospital, Beijing, China.
| |
Collapse
|
40
|
Tandon N, Nanda P, Padmanabhan JL, Mathew IT, Eack SM, Narayanan B, Meda SA, Bergen SE, Ruaño G, Windemuth A, Kocherla M, Petryshen TL, Clementz B, Sweeney J, Tamminga C, Pearlson G, Keshavan MS. Novel gene-brain structure relationships in psychotic disorder revealed using parallel independent component analyses. Schizophr Res 2017; 182:74-83. [PMID: 27789186 DOI: 10.1016/j.schres.2016.10.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/14/2016] [Accepted: 10/16/2016] [Indexed: 01/13/2023]
Abstract
BACKGROUND Schizophrenia, schizoaffective disorder, and psychotic bipolar disorder overlap with regard to symptoms, structural and functional brain abnormalities, and genetic risk factors. Neurobiological pathways connecting genes to clinical phenotypes across the spectrum from schizophrenia to psychotic bipolar disorder remain largely unknown. METHODS We examined the relationship between structural brain changes and risk alleles across the psychosis spectrum in the multi-site Bipolar-Schizophrenia Network for Intermediate Phenotypes (B-SNIP) cohort. Regional MRI brain volumes were examined in 389 subjects with a psychotic disorder (139 schizophrenia, 90 schizoaffective disorder, and 160 psychotic bipolar disorder) and 123 healthy controls. 451,701 single-nucleotide polymorphisms were screened and processed using parallel independent component analysis (para-ICA) to assess associations between genes and structural brain abnormalities in probands. RESULTS 482 subjects were included after quality control (364 individuals with psychotic disorder and 118 healthy controls). Para-ICA identified four genetic components including several risk genes already known to contribute to schizophrenia and bipolar disorder and revealed three structural components that showed overlapping relationships with the disease risk genes across the three psychotic disorders. Functional ontologies representing these gene clusters included physiological pathways involved in brain development, synaptic transmission, and ion channel activity. CONCLUSIONS Heritable brain structural findings such as reduced cortical thickness and surface area in probands across the psychosis spectrum were associated with somewhat distinct genes related to putative disease pathways implicated in psychotic disorders. This suggests that brain structural alterations might represent discrete psychosis intermediate phenotypes along common neurobiological pathways underlying disease expression across the psychosis spectrum.
Collapse
Affiliation(s)
- Neeraj Tandon
- Psychiatry, Harvard Medical School, Beth Israel Deaconess Medical Ctr, Boston, MA, USA; Baylor College of Medicine, Texas Medical Center, Houston, TX, USA.
| | - Pranav Nanda
- Psychiatry, Harvard Medical School, Beth Israel Deaconess Medical Ctr, Boston, MA, USA; College of Physicians & Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Jaya L Padmanabhan
- Psychiatry, Harvard Medical School, Beth Israel Deaconess Medical Ctr, Boston, MA, USA
| | - Ian T Mathew
- Psychiatry, Harvard Medical School, Beth Israel Deaconess Medical Ctr, Boston, MA, USA
| | - Shaun M Eack
- School of Social Work, University of Pittsburgh, Pittsburgh, PA, USA
| | - Balaji Narayanan
- Olin Neuropsychiatry Research Center, Hartford, CT, USA; Department of Psychiatry and Neurobiology, Yale University, New Haven, CT, USA
| | - Shashwath A Meda
- Olin Neuropsychiatry Research Center, Hartford, CT, USA; Department of Psychiatry and Neurobiology, Yale University, New Haven, CT, USA
| | - Sarah E Bergen
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | | | | | | | - Tracey L Petryshen
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Brett Clementz
- Department of Psychology, Department of Neuroscience, Bio-Imaging Research Center, University of Georgia, Athens, GA, USA
| | | | | | - Godfrey Pearlson
- Olin Neuropsychiatry Research Center, Hartford, CT, USA; Department of Psychiatry and Neurobiology, Yale University, New Haven, CT, USA
| | - Matcheri S Keshavan
- Psychiatry, Harvard Medical School, Beth Israel Deaconess Medical Ctr, Boston, MA, USA
| |
Collapse
|
41
|
Beydoun R, Hamood MA, Gomez Zubieta DM, Kondapalli KC. Na +/H + Exchanger 9 Regulates Iron Mobilization at the Blood-Brain Barrier in Response to Iron Starvation. J Biol Chem 2017; 292:4293-4301. [PMID: 28130443 DOI: 10.1074/jbc.m116.769240] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/24/2017] [Indexed: 12/21/2022] Open
Abstract
Iron is essential for brain function, with loss of iron homeostasis in the brain linked to neurological diseases ranging from rare syndromes to more common disorders, such as Parkinson's and Alzheimer's diseases. Iron entry into the brain is regulated by the blood-brain barrier (BBB). Molecular mechanisms regulating this transport are poorly understood. Using an in vitro model of the BBB, we identify NHE9, an endosomal cation/proton exchanger, as a novel regulator of this system. Human brain microvascular endothelial cells (hBMVECs) that constitute the BBB receive brain iron status information via paracrine signals from ensheathing astrocytes. In hBMVECs, we show that NHE9 expression is up-regulated very early in a physiological response invoked by paracrine signals from iron-starved astrocytes. Ectopic expression of NHE9 in hBMVECs without external cues induced up-regulation of the transferrin receptor (TfR) and down-regulation of ferritin, leading to an increase in iron uptake. Mechanistically, we demonstrate that NHE9 localizes to recycling endosomes in hBMVECs where it raises the endosomal pH. The ensuing alkalization of the endosomal lumen increased translocation of TfRs to the hBMVEC membrane. TfRs on the membrane were previously shown to facilitate both recycling-dependent and -independent iron uptake. We propose that NHE9 regulates TfR-dependent, recycling-independent iron uptake in hBMVECs by fine-tuning the endosomal pH in response to paracrine signals and is therefore an important regulator in iron mobilization pathway at the BBB.
Collapse
Affiliation(s)
- Rami Beydoun
- From the Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan 48128
| | - Mohamed A Hamood
- From the Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan 48128
| | - Daniela M Gomez Zubieta
- From the Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan 48128
| | - Kalyan C Kondapalli
- From the Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan 48128
| |
Collapse
|
42
|
De M, Oleskie AN, Ayyash M, Dutta S, Mancour L, Abazeed ME, Brace EJ, Skiniotis G, Fuller RS. The Vps13p-Cdc31p complex is directly required for TGN late endosome transport and TGN homotypic fusion. J Cell Biol 2017; 216:425-439. [PMID: 28122955 PMCID: PMC5294781 DOI: 10.1083/jcb.201606078] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/04/2016] [Accepted: 01/11/2017] [Indexed: 01/09/2023] Open
Abstract
VPS13 proteins are widely conserved in eukaryotes and associated with human neurodegenerative and neurodevelopmental diseases. De et al. describe the lipid specificity and structure of yeast Vps13p, providing insight into its role in both TGN late endosome transport and TGN homotypic fusion. Yeast VPS13 is the founding member of a eukaryotic gene family of growing interest in cell biology and medicine. Mutations in three of four human VPS13 genes cause autosomal recessive neurodegenerative or neurodevelopmental disease, making yeast Vps13p an important structural and functional model. Using cell-free reconstitution with purified Vps13p, we show that Vps13p is directly required both for transport from the trans-Golgi network (TGN) to the late endosome/prevacuolar compartment (PVC) and for TGN homotypic fusion. Vps13p must be in complex with the small calcium-binding protein Cdc31p to be active. Single-particle electron microscopic analysis of negatively stained Vps13p indicates that this 358-kD protein is folded into a compact rod-shaped density (20 × 4 nm) with a loop structure at one end with a circular opening ∼6 nm in diameter. Vps13p exhibits ATP-stimulated binding to yeast membranes and specific interactions with phosphatidic acid and phosphorylated forms of phosphatidyl inositol at least in part through the binding affinities of conserved N- and C-terminal domains.
Collapse
Affiliation(s)
- Mithu De
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Austin N Oleskie
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Mariam Ayyash
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Somnath Dutta
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Liliya Mancour
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Mohamed E Abazeed
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Eddy J Brace
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Georgios Skiniotis
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Robert S Fuller
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
43
|
de la Monte SM. Insulin Resistance and Neurodegeneration: Progress Towards the Development of New Therapeutics for Alzheimer's Disease. Drugs 2017; 77:47-65. [PMID: 27988872 PMCID: PMC5575843 DOI: 10.1007/s40265-016-0674-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) should be regarded as a degenerative metabolic disease caused by brain insulin resistance and deficiency, and overlapping with the molecular, biochemical, pathophysiological, and metabolic dysfunctions in diabetes mellitus, non-alcoholic fatty liver disease, and metabolic syndrome. Although most of the diagnostic and therapeutic approaches over the past several decades have focused on amyloid-beta (Aβ42) and aberrantly phosphorylated tau, which could be caused by consequences of brain insulin resistance, the broader array of pathologies including white matter atrophy with loss of myelinated fibrils and leukoaraiosis, non-Aβ42 microvascular disease, dysregulated lipid metabolism, mitochondrial dysfunction, astrocytic gliosis, neuro-inflammation, and loss of synapses vis-à-vis growth of dystrophic neurites, is not readily accounted for by Aβ42 accumulations, but could be explained by dysregulated insulin/IGF-1 signaling with attendant impairments in signal transduction and gene expression. This review covers the diverse range of brain abnormalities in AD and discusses how insulins, incretins, and insulin sensitizers could be utilized to treat at different stages of neurodegeneration.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Pierre Galletti Research Building, 55 Claverick Street, Room 419, Providence, RI, 02903, USA.
- Department of Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Neuropathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
44
|
Vilor-Tejedor N, Cáceres A, Pujol J, Sunyer J, González JR. Imaging genetics in attention-deficit/hyperactivity disorder and related neurodevelopmental domains: state of the art. Brain Imaging Behav 2016; 11:1922-1931. [DOI: 10.1007/s11682-016-9663-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
45
|
Silva RF, Plis SM, Sui J, Pattichis MS, Adalı T, Calhoun VD. Blind Source Separation for Unimodal and Multimodal Brain Networks: A Unifying Framework for Subspace Modeling. IEEE JOURNAL OF SELECTED TOPICS IN SIGNAL PROCESSING 2016; 10:1134-1149. [PMID: 28461840 PMCID: PMC5409135 DOI: 10.1109/jstsp.2016.2594945] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In the past decade, numerous advances in the study of the human brain were fostered by successful applications of blind source separation (BSS) methods to a wide range of imaging modalities. The main focus has been on extracting "networks" represented as the underlying latent sources. While the broad success in learning latent representations from multiple datasets has promoted the wide presence of BSS in modern neuroscience, it also introduced a wide variety of objective functions, underlying graphical structures, and parameter constraints for each method. Such diversity, combined with a host of datatype-specific know-how, can cause a sense of disorder and confusion, hampering a practitioner's judgment and impeding further development. We organize the diverse landscape of BSS models by exposing its key features and combining them to establish a novel unifying view of the area. In the process, we unveil important connections among models according to their properties and subspace structures. Consequently, a high-level descriptive structure is exposed, ultimately helping practitioners select the right model for their applications. Equipped with that knowledge, we review the current state of BSS applications to neuroimaging. The gained insight into model connections elicits a broader sense of generalization, highlighting several directions for model development. In light of that, we discuss emerging multi-dataset multidimensional (MDM) models and summarize their benefits for the study of the healthy brain and disease-related changes.
Collapse
Affiliation(s)
- Rogers F. Silva
- Dept. of ECE at The University of New Mexico, NM USA
- The Mind Research Network, LBERI, Albuquerque, New Mexico USA
| | - Sergey M. Plis
- The Mind Research Network, LBERI, Albuquerque, New Mexico USA
| | - Jing Sui
- Brainnetome Center & NLPR, Institute of Automation, Chinese Academy of Sciences, Beijing China
- The Mind Research Network, LBERI, Albuquerque, New Mexico USA
| | | | - Tülay Adalı
- Dept. of CSEE, University of Maryland Baltimore County, Baltimore, Maryland USA
| | - Vince D. Calhoun
- Dept. of ECE at The University of New Mexico, NM USAThe Mind Research Network, LBERI, Albuquerque, New Mexico USA
| |
Collapse
|
46
|
Safaralizadeh T, Jamshidi J, Esmaili Shandiz E, Movafagh A, Fazeli A, Emamalizadeh B, Manafi N, Taghavi S, Tafakhori A, Darvish H. SIPA1L2 , MIR4697 , GCH1 and VPS13C loci and risk of Parkinson's diseases in Iranian population: A case-control study. J Neurol Sci 2016; 369:1-4. [DOI: 10.1016/j.jns.2016.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/20/2016] [Accepted: 08/01/2016] [Indexed: 12/01/2022]
|
47
|
Chekouo T, Stingo FC, Guindani M, Do KA. A Bayesian predictive model for imaging genetics with application to schizophrenia. Ann Appl Stat 2016. [DOI: 10.1214/16-aoas948] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
48
|
Smith AR, Mill J, Smith RG, Lunnon K. Elucidating novel dysfunctional pathways in Alzheimer's disease by integrating loci identified in genetic and epigenetic studies. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.nepig.2016.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
49
|
Mokhtari M, Narayanan B, Hamm JP, Soh P, Calhoun VD, Ruaño G, Kocherla M, Windemuth A, Clementz BA, Tamminga CA, Sweeney JA, Keshavan MS, Pearlson GD. Multivariate Genetic Correlates of the Auditory Paired Stimuli-Based P2 Event-Related Potential in the Psychosis Dimension From the BSNIP Study. Schizophr Bull 2016; 42:851-62. [PMID: 26462502 PMCID: PMC4838080 DOI: 10.1093/schbul/sbv147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The complex molecular etiology of psychosis in schizophrenia (SZ) and psychotic bipolar disorder (PBP) is not well defined, presumably due to their multifactorial genetic architecture. Neurobiological correlates of psychosis can be identified through genetic associations of intermediate phenotypes such as event-related potential (ERP) from auditory paired stimulus processing (APSP). Various ERP components of APSP are heritable and aberrant in SZ, PBP and their relatives, but their multivariate genetic factors are less explored. METHODS We investigated the multivariate polygenic association of ERP from 64-sensor auditory paired stimulus data in 149 SZ, 209 PBP probands, and 99 healthy individuals from the multisite Bipolar-Schizophrenia Network on Intermediate Phenotypes study. Multivariate association of 64-channel APSP waveforms with a subset of 16 999 single nucleotide polymorphisms (SNPs) (reduced from 1 million SNP array) was examined using parallel independent component analysis (Para-ICA). Biological pathways associated with the genes were assessed using enrichment-based analysis tools. RESULTS Para-ICA identified 2 ERP components, of which one was significantly correlated with a genetic network comprising multiple linearly coupled gene variants that explained ~4% of the ERP phenotype variance. Enrichment analysis revealed epidermal growth factor, endocannabinoid signaling, glutamatergic synapse and maltohexaose transport associated with P2 component of the N1-P2 ERP waveform. This ERP component also showed deficits in SZ and PBP. CONCLUSIONS Aberrant P2 component in psychosis was associated with gene networks regulating several fundamental biologic functions, either general or specific to nervous system development. The pathways and processes underlying the gene clusters play a crucial role in brain function, plausibly implicated in psychosis.
Collapse
Affiliation(s)
- Mohammadreza Mokhtari
- Olin Neuropsychiatry Research Center, Hartford Hospital, Institute of Living, Hartford, CT
| | - Balaji Narayanan
- Olin Neuropsychiatry Research Center, Hartford Hospital, Institute of Living, Hartford, CT;
| | - Jordan P. Hamm
- Department of Psychology, University of Georgia, Athens, GA
| | - Pauline Soh
- Olin Neuropsychiatry Research Center, Hartford Hospital, Institute of Living, Hartford, CT
| | - Vince D. Calhoun
- Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM;,Image Analysis and MR Research Center, The Mind Research Network, Albuquerque, NM
| | - Gualberto Ruaño
- Genetics Research Center, Hartford Hospital, Hartford, CT;,Genomas Inc, Hartford, CT
| | - Mohan Kocherla
- Genetics Research Center, Hartford Hospital, Hartford, CT;,Genomas Inc, Hartford, CT
| | | | | | - Carol A. Tamminga
- Department of Psychiatry, UT Southwestern Medical School, Dallas, TX
| | - John A. Sweeney
- Department of Psychiatry, UT Southwestern Medical School, Dallas, TX
| | - Matcheri S. Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Godfrey D. Pearlson
- Olin Neuropsychiatry Research Center, Hartford Hospital, Institute of Living, Hartford, CT;,Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
50
|
Yang RY, Xue H, Yu L, Velayos-Baeza A, Monaco AP, Liu FT. Identification of VPS13C as a Galectin-12-Binding Protein That Regulates Galectin-12 Protein Stability and Adipogenesis. PLoS One 2016; 11:e0153534. [PMID: 27073999 PMCID: PMC4830523 DOI: 10.1371/journal.pone.0153534] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/30/2016] [Indexed: 01/13/2023] Open
Abstract
Galectin-12, a member of the galectin family of β-galactoside-binding animal lectins, is preferentially expressed in adipocytes and required for adipocyte differentiation in vitro. This protein was recently found to regulate lipolysis, whole body adiposity, and glucose homeostasis in vivo. Here we identify VPS13C, a member of the VPS13 family of vacuolar protein sorting-associated proteins highly conserved throughout eukaryotic evolution, as a major galectin-12-binding protein. VPS13C is upregulated during adipocyte differentiation, and is required for galectin-12 protein stability. Knockdown of Vps13c markedly reduces the steady-state levels of galectin-12 by promoting its degradation through primarily the lysosomal pathway, and impairs adipocyte differentiation. Our studies also suggest that VPS13C may have a broader role in protein quality control. The regulation of galectin-12 stability by VPS13C could potentially be exploited for therapeutic intervention of obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Ri-Yao Yang
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
| | - Huiting Xue
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
- School of Life Sciences, Northeast Normal University, Changchun, 130024, People’s Republic of China
| | - Lan Yu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
| | | | - Anthony P. Monaco
- Wellcome Trust Centre for Human Genetics, OX3 7BN, Oxford, United Kingdom
| | - Fu-Tong Liu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, 95817, United States of America
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| |
Collapse
|