51
|
Mengelkoch S, Gassen J, Lev-Ari S, Alley JC, Schüssler-Fiorenza Rose SM, Snyder MP, Slavich GM. Multi-omics in stress and health research: study designs that will drive the field forward. Stress 2024; 27:2321610. [PMID: 38425100 PMCID: PMC11216062 DOI: 10.1080/10253890.2024.2321610] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
Despite decades of stress research, there still exist substantial gaps in our understanding of how social, environmental, and biological factors interact and combine with developmental stressor exposures, cognitive appraisals of stressors, and psychosocial coping processes to shape individuals' stress reactivity, health, and disease risk. Relatively new biological profiling approaches, called multi-omics, are helping address these issues by enabling researchers to quantify thousands of molecules from a single blood or tissue sample, thus providing a panoramic snapshot of the molecular processes occurring in an organism from a systems perspective. In this review, we summarize two types of research designs for which multi-omics approaches are best suited, and describe how these approaches can help advance our understanding of stress processes and the development, prevention, and treatment of stress-related pathologies. We first discuss incorporating multi-omics approaches into theory-rich, intensive longitudinal study designs to characterize, in high-resolution, the transition to stress-related multisystem dysfunction and disease throughout development. Next, we discuss how multi-omics approaches should be incorporated into intervention research to better understand the transition from stress-related dysfunction back to health, which can help inform novel precision medicine approaches to managing stress and fostering biopsychosocial resilience. Throughout, we provide concrete recommendations for types of studies that will help advance stress research, and translate multi-omics data into better health and health care.
Collapse
Affiliation(s)
- Summer Mengelkoch
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Jeffrey Gassen
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Shahar Lev-Ari
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Health Promotion, Tel Aviv University, Tel Aviv, Israel
| | - Jenna C. Alley
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | | | | | - George M. Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
52
|
Perez AA, Goronzy IN, Blanco MR, Guo JK, Guttman M. ChIP-DIP: A multiplexed method for mapping hundreds of proteins to DNA uncovers diverse regulatory elements controlling gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571730. [PMID: 38187704 PMCID: PMC10769186 DOI: 10.1101/2023.12.14.571730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Gene expression is controlled by the dynamic localization of thousands of distinct regulatory proteins to precise regions of DNA. Understanding this cell-type specific process has been a goal of molecular biology for decades yet remains challenging because most current DNA-protein mapping methods study one protein at a time. To overcome this, we developed ChIP-DIP (ChIP Done In Parallel), a split-pool based method that enables simultaneous, genome-wide mapping of hundreds of diverse regulatory proteins in a single experiment. We demonstrate that ChIP-DIP generates highly accurate maps for all classes of DNA-associated proteins, including histone modifications, chromatin regulators, transcription factors, and RNA Polymerases. Using these data, we explore quantitative combinations of protein localization on genomic DNA to define distinct classes of regulatory elements and their functional activity. Our data demonstrate that ChIP-DIP enables the generation of 'consortium level', context-specific protein localization maps within any molecular biology lab.
Collapse
|
53
|
Maden SK, Kwon SH, Huuki-Myers LA, Collado-Torres L, Hicks SC, Maynard KR. Challenges and opportunities to computationally deconvolve heterogeneous tissue with varying cell sizes using single-cell RNA-sequencing datasets. Genome Biol 2023; 24:288. [PMID: 38098055 PMCID: PMC10722720 DOI: 10.1186/s13059-023-03123-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Deconvolution of cell mixtures in "bulk" transcriptomic samples from homogenate human tissue is important for understanding disease pathologies. However, several experimental and computational challenges impede transcriptomics-based deconvolution approaches using single-cell/nucleus RNA-seq reference atlases. Cells from the brain and blood have substantially different sizes, total mRNA, and transcriptional activities, and existing approaches may quantify total mRNA instead of cell type proportions. Further, standards are lacking for the use of cell reference atlases and integrative analyses of single-cell and spatial transcriptomics data. We discuss how to approach these key challenges with orthogonal "gold standard" datasets for evaluating deconvolution methods.
Collapse
Affiliation(s)
- Sean K Maden
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA.
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA.
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
54
|
Bazinet V, Hansen JY, Misic B. Towards a biologically annotated brain connectome. Nat Rev Neurosci 2023; 24:747-760. [PMID: 37848663 DOI: 10.1038/s41583-023-00752-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
The brain is a network of interleaved neural circuits. In modern connectomics, brain connectivity is typically encoded as a network of nodes and edges, abstracting away the rich biological detail of local neuronal populations. Yet biological annotations for network nodes - such as gene expression, cytoarchitecture, neurotransmitter receptors or intrinsic dynamics - can be readily measured and overlaid on network models. Here we review how connectomes can be represented and analysed as annotated networks. Annotated connectomes allow us to reconceptualize architectural features of networks and to relate the connection patterns of brain regions to their underlying biology. Emerging work demonstrates that annotated connectomes help to make more veridical models of brain network formation, neural dynamics and disease propagation. Finally, annotations can be used to infer entirely new inter-regional relationships and to construct new types of network that complement existing connectome representations. In summary, biologically annotated connectomes offer a compelling way to study neural wiring in concert with local biological features.
Collapse
Affiliation(s)
- Vincent Bazinet
- Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Justine Y Hansen
- Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Bratislav Misic
- Montréal Neurological Institute, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
55
|
Bettencourt C, Skene N, Bandres-Ciga S, Anderson E, Winchester LM, Foote IF, Schwartzentruber J, Botia JA, Nalls M, Singleton A, Schilder BM, Humphrey J, Marzi SJ, Toomey CE, Kleifat AA, Harshfield EL, Garfield V, Sandor C, Keat S, Tamburin S, Frigerio CS, Lourida I, Ranson JM, Llewellyn DJ. Artificial intelligence for dementia genetics and omics. Alzheimers Dement 2023; 19:5905-5921. [PMID: 37606627 PMCID: PMC10841325 DOI: 10.1002/alz.13427] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 08/23/2023]
Abstract
Genetics and omics studies of Alzheimer's disease and other dementia subtypes enhance our understanding of underlying mechanisms and pathways that can be targeted. We identified key remaining challenges: First, can we enhance genetic studies to address missing heritability? Can we identify reproducible omics signatures that differentiate between dementia subtypes? Can high-dimensional omics data identify improved biomarkers? How can genetics inform our understanding of causal status of dementia risk factors? And which biological processes are altered by dementia-related genetic variation? Artificial intelligence (AI) and machine learning approaches give us powerful new tools in helping us to tackle these challenges, and we review possible solutions and examples of best practice. However, their limitations also need to be considered, as well as the need for coordinated multidisciplinary research and diverse deeply phenotyped cohorts. Ultimately AI approaches improve our ability to interrogate genetics and omics data for precision dementia medicine. HIGHLIGHTS: We have identified five key challenges in dementia genetics and omics studies. AI can enable detection of undiscovered patterns in dementia genetics and omics data. Enhanced and more diverse genetics and omics datasets are still needed. Multidisciplinary collaborative efforts using AI can boost dementia research.
Collapse
Affiliation(s)
- Conceicao Bettencourt
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Nathan Skene
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Emma Anderson
- Department of Mental Health of Older People, Division of Psychiatry, University College London, London, UK
| | | | - Isabelle F Foote
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado, USA
| | - Jeremy Schwartzentruber
- Open Targets, Cambridge, UK
- Wellcome Sanger Institute, Cambridge, UK
- Illumina Artificial Intelligence Laboratory, Illumina Inc, Foster City, California, USA
| | - Juan A Botia
- Departamento de Ingeniería de la Información y las Comunicaciones, Universidad de Murcia, Murcia, Spain
| | - Mike Nalls
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Data Tecnica International LLC, Washington, DC, USA
| | - Andrew Singleton
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian M Schilder
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Jack Humphrey
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Sarah J Marzi
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Christina E Toomey
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Ahmad Al Kleifat
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Eric L Harshfield
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Victoria Garfield
- MRC Unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, London, UK
| | - Cynthia Sandor
- UK Dementia Research Institute. School of Medicine, Cardiff University, Cardiff, UK
| | - Samuel Keat
- UK Dementia Research Institute. School of Medicine, Cardiff University, Cardiff, UK
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement Sciences, Neurology Section, University of Verona, Verona, Italy
| | - Carlo Sala Frigerio
- UK Dementia Research Institute, Queen Square Institute of Neurology, University College London, London, UK
| | | | | | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK
- The Alan Turing Institute, London, UK
| |
Collapse
|
56
|
Tüshaus J, Sakhteman A, Lechner S, The M, Mucha E, Krisp C, Schlegel J, Delbridge C, Kuster B. A region-resolved proteomic map of the human brain enabled by high-throughput proteomics. EMBO J 2023; 42:e114665. [PMID: 37916885 PMCID: PMC10690467 DOI: 10.15252/embj.2023114665] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023] Open
Abstract
Substantial efforts are underway to deepen our understanding of human brain morphology, structure, and function using high-resolution imaging as well as high-content molecular profiling technologies. The current work adds to these approaches by providing a comprehensive and quantitative protein expression map of 13 anatomically distinct brain regions covering more than 11,000 proteins. This was enabled by the optimization, characterization, and implementation of a high-sensitivity and high-throughput microflow liquid chromatography timsTOF tandem mass spectrometry system (LC-MS/MS) capable of analyzing more than 2,000 consecutive samples prepared from formalin-fixed paraffin embedded (FFPE) material. Analysis of this proteomic resource highlighted brain region-enriched protein expression patterns and functional protein classes, protein localization differences between brain regions and individual markers for specific areas. To facilitate access to and ease further mining of the data by the scientific community, all data can be explored online in a purpose-built R Shiny app (https://brain-region-atlas.proteomics.ls.tum.de).
Collapse
Affiliation(s)
- Johanna Tüshaus
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life SciencesTechnical University of MunichMunichGermany
| | - Amirhossein Sakhteman
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life SciencesTechnical University of MunichMunichGermany
| | - Severin Lechner
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life SciencesTechnical University of MunichMunichGermany
| | - Matthew The
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life SciencesTechnical University of MunichMunichGermany
| | - Eike Mucha
- Bruker Daltonics GmbH & Co. KGBremenGermany
| | | | - Jürgen Schlegel
- Department of Neuropathology, Klinikum Rechts der ISAR, School of MedicineTechnical University MunichMunichGermany
| | - Claire Delbridge
- Department of Neuropathology, Klinikum Rechts der ISAR, School of MedicineTechnical University MunichMunichGermany
| | - Bernhard Kuster
- Proteomics and Bioanalytics, Department of Molecular Life Sciences, School of Life SciencesTechnical University of MunichMunichGermany
- German Cancer Consortium (DKTK), Munich SiteHeidelbergGermany
| |
Collapse
|
57
|
Zhang J, Zhao H. eQTL studies: from bulk tissues to single cells. J Genet Genomics 2023; 50:925-933. [PMID: 37207929 PMCID: PMC10656365 DOI: 10.1016/j.jgg.2023.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
An expression quantitative trait locus (eQTL) is a chromosomal region where genetic variants are associated with the expression levels of specific genes that can be both nearby or distant. The identifications of eQTLs for different tissues, cell types, and contexts have led to a better understanding of the dynamic regulations of gene expressions and implications of functional genes and variants for complex traits and diseases. Although most eQTL studies have been performed on data collected from bulk tissues, recent studies have demonstrated the importance of cell-type-specific and context-dependent gene regulations in biological processes and disease mechanisms. In this review, we discuss statistical methods that have been developed to enable the detection of cell-type-specific and context-dependent eQTLs from bulk tissues, purified cell types, and single cells. We also discuss the limitations of the current methods and future research opportunities.
Collapse
Affiliation(s)
- Jingfei Zhang
- Information Systems and Operations Management, Emory University, Atlanta, GA 30322, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 208034, USA.
| |
Collapse
|
58
|
Seidlitz J, Mallard TT, Vogel JW, Lee YH, Warrier V, Ball G, Hansson O, Hernandez LM, Mandal AS, Wagstyl K, Lombardo MV, Courchesne E, Glessner JT, Satterthwaite TD, Bethlehem RAI, Bernstock JD, Tasaki S, Ng B, Gaiteri C, Smoller JW, Ge T, Gur RE, Gandal MJ, Alexander-Bloch AF. The molecular genetic landscape of human brain size variation. Cell Rep 2023; 42:113439. [PMID: 37963017 PMCID: PMC11694216 DOI: 10.1016/j.celrep.2023.113439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/13/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
Human brain size changes dynamically through early development, peaks in adolescence, and varies up to 2-fold among adults. However, the molecular genetic underpinnings of interindividual variation in brain size remain unknown. Here, we leveraged postmortem brain RNA sequencing and measurements of brain weight (BW) in 2,531 individuals across three independent datasets to identify 928 genome-wide significant associations with BW. Genes associated with higher or lower BW showed distinct neurodevelopmental trajectories and spatial patterns that mapped onto functional and cellular axes of brain organization. Expression of BW genes was predictive of interspecies differences in brain size, and bioinformatic annotation revealed enrichment for neurogenesis and cell-cell communication. Genome-wide, transcriptome-wide, and phenome-wide association analyses linked BW gene sets to neuroimaging measurements of brain size and brain-related clinical traits. Cumulatively, these results represent a major step toward delineating the molecular pathways underlying human brain size variation in health and disease.
Collapse
Affiliation(s)
- Jakob Seidlitz
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA
| | - Jacob W Vogel
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA; Lifespan Informatics and Neuroimaging Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Younga H Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA
| | - Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; Department of Psychology, University of Cambridge, Cambridge CB2 1TN, UK
| | - Gareth Ball
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Melbourne, VIC 3052, Australia
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö P663+Q9, Sweden; Memory Clinic, Skåne University Hospital, Malmö P663+Q9, Sweden
| | - Leanna M Hernandez
- Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA 90024, USA
| | - Ayan S Mandal
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Konrad Wagstyl
- Wellcome Centre for Human Neuroimaging, University College London, London WC1N 3AR, UK
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Eric Courchesne
- Department of Neuroscience, University of California, San Diego, San Diego, CA 92093, USA; Autism Center of Excellence, University of California, San Diego, San Diego, CA 92093, USA
| | - Joseph T Glessner
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Theodore D Satterthwaite
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA; Lifespan Informatics and Neuroimaging Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | - Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard University, Boston, MA 02115, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard University, Boston, MA 02115, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA; Center for Precision Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA 02142, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02142, USA; Center for Precision Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Raquel E Gur
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Gandal
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aaron F Alexander-Bloch
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
59
|
Jané P, Xu X, Taelman V, Jané E, Gariani K, Dumont RA, Garama Y, Kim F, Del Val Gomez M, Walter MA. The Imageable Genome. Nat Commun 2023; 14:7329. [PMID: 37957176 PMCID: PMC10643363 DOI: 10.1038/s41467-023-43123-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Understanding human disease on a molecular level, and translating this understanding into targeted diagnostics and therapies are central tenets of molecular medicine1. Realizing this doctrine requires an efficient adaptation of molecular discoveries into the clinic. We present an approach to facilitate this process by describing the Imageable Genome, the part of the human genome whose expression can be assessed via molecular imaging. Using a deep learning-based hybrid human-AI pipeline, we bridge individual genes and their relevance in human diseases with specific molecular imaging methods. Cross-referencing the Imageable Genome with RNA-seq data from over 60,000 individuals reveals diagnostic, prognostic and predictive imageable genes for a wide variety of major human diseases. Having both the critical size and focus to be altered in its expression during the development and progression of any human disease, the Imageable Genome will generate new imaging tools that improve the understanding, diagnosis and management of human diseases.
Collapse
Affiliation(s)
- Pablo Jané
- University of Geneva, Geneva, Switzerland
- Nuclear Medicine and Molecular Imaging Division, Geneva University Hospitals, Geneva, Switzerland
| | | | | | - Eduardo Jané
- Departamento de Matemática Aplicada a la Ingeniería Aeroespacial - ETSIAE, Universidad Politécnica de Madrid, 28040, Madrid, Spain
| | - Karim Gariani
- Division of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Geneva, Switzerland
| | | | | | | | - María Del Val Gomez
- Servicio de Medicina Nuclear, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Martin A Walter
- University of Lucerne, Lucerne, Switzerland.
- St. Anna Hospital, University of Lucerne, Lucerne, Switzerland.
| |
Collapse
|
60
|
Liang Q, Jiang Y, Shieh AW, Zhou D, Chen R, Wang F, Xu M, Niu M, Wang X, Pinto D, Wang Y, Cheng L, Vadukapuram R, Zhang C, Grennan K, Giase G, The PsychENCODE Consortium, White KP, Peng J, Li B, Liu C, Chen C, Wang SH. The impact of common variants on gene expression in the human brain: from RNA to protein to schizophrenia risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.04.543603. [PMID: 37873195 PMCID: PMC10592607 DOI: 10.1101/2023.06.04.543603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Background The impact of genetic variants on gene expression has been intensely studied at the transcription level, yielding in valuable insights into the association between genes and the risk of complex disorders, such as schizophrenia (SCZ). However, the downstream impact of these variants and the molecular mechanisms connecting transcription variation to disease risk are not well understood. Results We quantitated ribosome occupancy in prefrontal cortex samples of the BrainGVEX cohort. Together with transcriptomics and proteomics data from the same cohort, we performed cis-Quantitative Trait Locus (QTL) mapping and identified 3,253 expression QTLs (eQTLs), 1,344 ribosome occupancy QTLs (rQTLs), and 657 protein QTLs (pQTLs) out of 7,458 genes quantitated in all three omics types from 185 samples. Of the eQTLs identified, only 34% have their effects propagated to the protein level. Further analysis on the effect size of prefrontal cortex eQTLs identified from an independent dataset showed clear post-transcriptional attenuation of eQTL effects. To investigate the biological relevance of the attenuated eQTLs, we identified 70 expression-specific QTLs (esQTLs), 51 ribosome-occupancy-specific QTLs (rsQTLs), and 107 protein-specific QTLs (psQTLs). Five of these omics-specific QTLs showed strong colocalization with SCZ GWAS signals, three of them are esQTLs. The limited number of GWAS colocalization discoveries from omics-specific QTLs and the apparent prevalence of eQTL attenuation prompted us to take a complementary approach to investigate the functional relevance of attenuated eQTLs. Using S-PrediXcan we identified 74 SCZ risk genes, 34% of which were novel, and 67% of these risk genes were replicated in a MR-Egger test. Notably, 52 out of 74 risk genes were identified using eQTL data and 70% of these SCZ-risk-gene-driving eQTLs show little to no evidence of driving corresponding variations at the protein level. Conclusion The effect of eQTLs on gene expression in the prefrontal cortex is commonly attenuated post-transcriptionally. Many of the attenuated eQTLs still correlate with SCZ GWAS signal. Further investigation is needed to elucidate a mechanistic link between attenuated eQTLs and SCZ disease risk.
Collapse
Affiliation(s)
- Qiuman Liang
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Yi Jiang
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Annie W. Shieh
- Center for Human Genetics, The Brown foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Rui Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Feiran Wang
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Meng Xu
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Mingming Niu
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Xusheng Wang
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dalila Pinto
- Department of Psychiatry, and Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Lijun Cheng
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Ramu Vadukapuram
- Department of Psychiatry, The University of Texas Rio Grande Valley, Harlingen, TX 78550, USA
| | - Chunling Zhang
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Kay Grennan
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Gina Giase
- The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | - Kevin P White
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Junmin Peng
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Bingshan Li
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Chunyu Liu
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- School of Psychology, Shaanxi Normal University, Xi’an, Shaanxi 710062, China
| | - Chao Chen
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- Furong Laboratory, Changsha, Hunan 410000, China
- National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan 410000, China
| | - Sidney H. Wang
- Center for Human Genetics, The Brown foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
61
|
Guo MG, Reynolds DL, Ang CE, Liu Y, Zhao Y, Donohue LKH, Siprashvili Z, Yang X, Yoo Y, Mondal S, Hong A, Kain J, Meservey L, Fabo T, Elfaki I, Kellman LN, Abell NS, Pershad Y, Bayat V, Etminani P, Holodniy M, Geschwind DH, Montgomery SB, Duncan LE, Urban AE, Altman RB, Wernig M, Khavari PA. Integrative analyses highlight functional regulatory variants associated with neuropsychiatric diseases. Nat Genet 2023; 55:1876-1891. [PMID: 37857935 PMCID: PMC10859123 DOI: 10.1038/s41588-023-01533-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/15/2023] [Indexed: 10/21/2023]
Abstract
Noncoding variants of presumed regulatory function contribute to the heritability of neuropsychiatric disease. A total of 2,221 noncoding variants connected to risk for ten neuropsychiatric disorders, including autism spectrum disorder, attention deficit hyperactivity disorder, bipolar disorder, borderline personality disorder, major depression, generalized anxiety disorder, panic disorder, post-traumatic stress disorder, obsessive-compulsive disorder and schizophrenia, were studied in developing human neural cells. Integrating epigenomic and transcriptomic data with massively parallel reporter assays identified differentially-active single-nucleotide variants (daSNVs) in specific neural cell types. Expression-gene mapping, network analyses and chromatin looping nominated candidate disease-relevant target genes modulated by these daSNVs. Follow-up integration of daSNV gene editing with clinical cohort analyses suggested that magnesium transport dysfunction may increase neuropsychiatric disease risk and indicated that common genetic pathomechanisms may mediate specific symptoms that are shared across multiple neuropsychiatric diseases.
Collapse
Affiliation(s)
- Margaret G Guo
- Stanford Program in Biomedical Informatics, Stanford University, Stanford, CA, USA
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - David L Reynolds
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Cheen E Ang
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Yingfei Liu
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA, USA
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yang Zhao
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Laura K H Donohue
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Xue Yang
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
- Stanford Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Yongjin Yoo
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Smarajit Mondal
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Audrey Hong
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
| | - Jessica Kain
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Tania Fabo
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ibtihal Elfaki
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Laura N Kellman
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA
- Stanford Program in Cancer Biology, Stanford University, Stanford, CA, USA
| | - Nathan S Abell
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Yash Pershad
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | | | - Mark Holodniy
- Public Health Surveillance and Research, Department of Veterans Affairs, Washington, DC, USA
- Division of Infectious Disease & Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel H Geschwind
- Program in Neurobehavioral Genetics, Semel Institute, UCLA, Los Angeles, CA, USA
| | - Stephen B Montgomery
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Laramie E Duncan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Alexander E Urban
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Russ B Altman
- Stanford Program in Biomedical Informatics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Marius Wernig
- Department of Pathology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University, Stanford, CA, USA.
- Stanford Program in Cancer Biology, Stanford University, Stanford, CA, USA.
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA.
| |
Collapse
|
62
|
Chiou KL, Huang X, Bohlen MO, Tremblay S, DeCasien AR, O’Day DR, Spurrell CH, Gogate AA, Zintel TM, Cayo Biobank Research Unit, Andrews MG, Martínez MI, Starita LM, Montague MJ, Platt ML, Shendure J, Snyder-Mackler N. A single-cell multi-omic atlas spanning the adult rhesus macaque brain. SCIENCE ADVANCES 2023; 9:eadh1914. [PMID: 37824616 PMCID: PMC10569716 DOI: 10.1126/sciadv.adh1914] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Cataloging the diverse cellular architecture of the primate brain is crucial for understanding cognition, behavior, and disease in humans. Here, we generated a brain-wide single-cell multimodal molecular atlas of the rhesus macaque brain. Together, we profiled 2.58 M transcriptomes and 1.59 M epigenomes from single nuclei sampled from 30 regions across the adult brain. Cell composition differed extensively across the brain, revealing cellular signatures of region-specific functions. We also identified 1.19 M candidate regulatory elements, many previously unidentified, allowing us to explore the landscape of cis-regulatory grammar and neurological disease risk in a cell type-specific manner. Altogether, this multi-omic atlas provides an open resource for investigating the evolution of the human brain and identifying novel targets for disease interventions.
Collapse
Affiliation(s)
- Kenneth L. Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Xingfan Huang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Martin O. Bohlen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sébastien Tremblay
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex R. DeCasien
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Diana R. O’Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Cailyn H. Spurrell
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Aishwarya A. Gogate
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Trisha M. Zintel
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Cayo Biobank Research Unit
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Caribbean Primate Research Center, University of Puerto Rico, San Juan, PR, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Madeline G. Andrews
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Melween I. Martínez
- Caribbean Primate Research Center, University of Puerto Rico, San Juan, PR, USA
| | - Lea M. Starita
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Michael J. Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael L. Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
63
|
Martínez Carrasco A, Real R, Lawton M, Hertfelder Reynolds R, Tan M, Wu L, Williams N, Carroll C, Corvol JC, Hu M, Grosset D, Hardy J, Ryten M, Ben-Shlomo Y, Shoai M, Morris HR. Genome-wide Analysis of Motor Progression in Parkinson Disease. Neurol Genet 2023; 9:e200092. [PMID: 37560120 PMCID: PMC10409573 DOI: 10.1212/nxg.0000000000200092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/08/2023] [Indexed: 08/11/2023]
Abstract
Background and Objectives The genetic basis of Parkinson disease (PD) motor progression is largely unknown. Previous studies of the genetics of PD progression have included small cohorts and shown a limited overlap with genetic PD risk factors from case-control studies. Here, we have studied genomic variation associated with PD motor severity and early-stage progression in large longitudinal cohorts to help to define the biology of PD progression and potential new drug targets. Methods We performed a GWAS meta-analysis of early PD motor severity and progression up to 3 years from study entry. We used linear mixed-effect models with additive effects, corrected for age at diagnosis, sex, and the first 5 genetic principal components to assess variability in axial, limb, and total Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) III scores. Results We included 3,572 unrelated European ancestry patients with PD from 5 observational cohorts and 1 drug trial. The average AAO was 62.6 years (SD = 9.83), and 63% of participants were male. We found an average increase in the total MDS-UPDRS III score of 2.3 points/year. We identified an association between PD axial motor progression and variation at the GJA5 locus at 1q12 (β = -0.25, SE = 0.04, p = 3.4e-10). Exploration of the regulation of gene expression in the region (cis-expression quantitative trait loci [eQTL] analysis) showed that the lead variant was associated with expression of ACP6, a lysophosphatidic acid phosphatase that regulates mitochondrial lipid biosynthesis (cis-eQTL p-values in blood and brain RNA expression data sets: <10-14 in eQTLGen and 10-7 in PsychEncode). Discussion Our study highlights the potential role of mitochondrial lipid homeostasis in the progression of PD, which may be important in establishing new drug targets that might modify disease progression.
Collapse
Affiliation(s)
- Alejandro Martínez Carrasco
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Raquel Real
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Michael Lawton
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Regina Hertfelder Reynolds
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Manuela Tan
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Lesley Wu
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Nigel Williams
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Camille Carroll
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Jean-Christophe Corvol
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Michele Hu
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Donald Grosset
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - John Hardy
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Mina Ryten
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Yoav Ben-Shlomo
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Maryam Shoai
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| | - Huw R Morris
- From the Department of Clinical and Movement Neurosciences (A.M.C., R.R., L.W., H.R.M.), UCL Queen Square Institute of Neurology; UCL Movement Disorders Centre (A.M.C., R.R., L.W., H.R.M.), University College London, United Kingdom; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network (A.M.C., R.R., R.H.R. L.W., M.R., M.S. J.H., H.R.M.), Chevy Chase, MD; Population Health Sciences (M.L., Y.B.-S.), Bristol Medical School, University of Bristol; Genetics and Genomic Medicine (R.H.R., M.R.), UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Neurology (M.T.), Oslo University Hospital, Norway; Institute of Psychological Medicine and Clinical Neurosciences (N.W.), MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University; Faculty of Health (C.C.), University of Plymouth, United Kingdom; Sorbonne Université (J.-C.C.), Institut du Cerveau - Paris Brain Institute - ICM, INSERM, CNRS; Assistance Publique Hôpitaux de Paris (J.-C.C.), Department of Neurology, Hôpital Pitié-Salpêtrière, France; Division of Clinical Neurology (M.H.), Nuffield Department of Clinical Neurosciences; Oxford Parkinson's Disease Centre (M.H.), University of Oxford; School of Neuroscience and Psychology (D.G.), University of Glasgow; Department of Neurodegenerative Diseases (J.H., M.S.), UCL Queen Square Institute of Neurology; UK Dementia Research Institute (J.H., M.S.), University College London; Reta Lila Weston Institute (J.H., M.S.), UCL Queen Square Institute of Neurology; National Institute for Health Research (NIHR), University College London Hospitals Biomedical Research Centre (J.H.); Institute for Advanced Study (J.H.), The Hong Kong University of Science and Technology, Hong Kong SAR, China; and NIHR Great Ormond Street Hospital Biomedical Research Centre (M.R.), University College London, United Kingdom
| |
Collapse
|
64
|
Xiong X, James BT, Boix CA, Park YP, Galani K, Victor MB, Sun N, Hou L, Ho LL, Mantero J, Scannail AN, Dileep V, Dong W, Mathys H, Bennett DA, Tsai LH, Kellis M. Epigenomic dissection of Alzheimer's disease pinpoints causal variants and reveals epigenome erosion. Cell 2023; 186:4422-4437.e21. [PMID: 37774680 PMCID: PMC10782612 DOI: 10.1016/j.cell.2023.08.040] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/04/2023] [Accepted: 08/29/2023] [Indexed: 10/01/2023]
Abstract
Recent work has identified dozens of non-coding loci for Alzheimer's disease (AD) risk, but their mechanisms and AD transcriptional regulatory circuitry are poorly understood. Here, we profile epigenomic and transcriptomic landscapes of 850,000 nuclei from prefrontal cortexes of 92 individuals with and without AD to build a map of the brain regulome, including epigenomic profiles, transcriptional regulators, co-accessibility modules, and peak-to-gene links in a cell-type-specific manner. We develop methods for multimodal integration and detecting regulatory modules using peak-to-gene linking. We show AD risk loci are enriched in microglial enhancers and for specific TFs including SPI1, ELF2, and RUNX1. We detect 9,628 cell-type-specific ATAC-QTL loci, which we integrate alongside peak-to-gene links to prioritize AD variant regulatory circuits. We report differential accessibility of regulatory modules in late AD in glia and in early AD in neurons. Strikingly, late-stage AD brains show global epigenome dysregulation indicative of epigenome erosion and cell identity loss.
Collapse
Affiliation(s)
- Xushen Xiong
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Benjamin T James
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Carles A Boix
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Yongjin P Park
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA; Department of Pathology and Laboratory Medicine, Department of Statistics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kyriaki Galani
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Matheus B Victor
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Na Sun
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Lei Hou
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Li-Lun Ho
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Julio Mantero
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Aine Ni Scannail
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vishnu Dileep
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Weixiu Dong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hansruedi Mathys
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Li-Huei Tsai
- The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA; Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA 02139, USA; The Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA.
| |
Collapse
|
65
|
Warrier V, Stauffer EM, Huang QQ, Wigdor EM, Slob EAW, Seidlitz J, Ronan L, Valk SL, Mallard TT, Grotzinger AD, Romero-Garcia R, Baron-Cohen S, Geschwind DH, Lancaster MA, Murray GK, Gandal MJ, Alexander-Bloch A, Won H, Martin HC, Bullmore ET, Bethlehem RAI. Genetic insights into human cortical organization and development through genome-wide analyses of 2,347 neuroimaging phenotypes. Nat Genet 2023; 55:1483-1493. [PMID: 37592024 PMCID: PMC10600728 DOI: 10.1038/s41588-023-01475-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Our understanding of the genetics of the human cerebral cortex is limited both in terms of the diversity and the anatomical granularity of brain structural phenotypes. Here we conducted a genome-wide association meta-analysis of 13 structural and diffusion magnetic resonance imaging-derived cortical phenotypes, measured globally and at 180 bilaterally averaged regions in 36,663 individuals and identified 4,349 experiment-wide significant loci. These phenotypes include cortical thickness, surface area, gray matter volume, measures of folding, neurite density and water diffusion. We identified four genetic latent structures and causal relationships between surface area and some measures of cortical folding. These latent structures partly relate to different underlying gene expression trajectories during development and are enriched for different cell types. We also identified differential enrichment for neurodevelopmental and constrained genes and demonstrate that common genetic variants associated with cortical expansion are associated with cephalic disorders. Finally, we identified complex interphenotype and inter-regional genetic relationships among the 13 phenotypes, reflecting the developmental differences among them. Together, these analyses identify distinct genetic organizational principles of the cortex and their correlates with neurodevelopment.
Collapse
Affiliation(s)
- Varun Warrier
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
- Department of Psychology, University of Cambridge, Cambridge, UK.
| | | | | | | | - Eric A W Slob
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, Rotterdam, the Netherlands
- Erasmus University Rotterdam Institute for Behavior and Biology, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Jakob Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa Ronan
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Sofie L Valk
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, FZ Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Andrew D Grotzinger
- Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA
- Institute for Behavioral Genetics, University of Colorado at Boulder, Boulder, CO, USA
| | - Rafael Romero-Garcia
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Instituto de Biomedicina de Sevilla (IBiS) HUVR/CSIC/Universidad de Sevilla/CIBERSAM, ISCIII, Dpto. de Fisiología Médica y Biofísica, Seville, Spain
| | - Simon Baron-Cohen
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Daniel H Geschwind
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, University of California, Los Angeles, CA, USA
- Center for Autism Research and Treatment, Jane and TerrySemel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Institute of Precision Health, University of California, Los Angeles, CA, USA
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, UK
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Trust, Cambridge, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Michael J Gandal
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
| | - Aaron Alexander-Bloch
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Hyejung Won
- Department of Genetics and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Trust, Cambridge, UK
| | - Richard A I Bethlehem
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
66
|
Han S, DiBlasi E, Monson ET, Shabalin A, Ferris E, Chen D, Fraser A, Yu Z, Staley M, Callor WB, Christensen ED, Crockett DK, Li QS, Willour V, Bakian AV, Keeshin B, Docherty AR, Eilbeck K, Coon H. Whole-genome sequencing analysis of suicide deaths integrating brain-regulatory eQTLs data to identify risk loci and genes. Mol Psychiatry 2023; 28:3909-3919. [PMID: 37794117 PMCID: PMC10730410 DOI: 10.1038/s41380-023-02282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023]
Abstract
Recent large-scale genome-wide association studies (GWAS) have started to identify potential genetic risk loci associated with risk of suicide; however, a large portion of suicide-associated genetic factors affecting gene expression remain elusive. Dysregulated gene expression, not assessed by GWAS, may play a significant role in increasing the risk of suicide death. We performed the first comprehensive genomic association analysis prioritizing brain expression quantitative trait loci (eQTLs) within regulatory regions in suicide deaths from the Utah Suicide Genetic Risk Study (USGRS). 440,324 brain-regulatory eQTLs were obtained by integrating brain eQTLs, histone modification ChIP-seq, ATAC-seq, DNase-seq, and Hi-C results from publicly available data. Subsequent genomic analyses were conducted in whole-genome sequencing (WGS) data from 986 suicide deaths of non-Finnish European (NFE) ancestry and 415 ancestrally matched controls. Additional independent USGRS suicide deaths with genotyping array data (n = 4657) and controls from the Genome Aggregation Database were explored for WGS result replication. One significant eQTL locus, rs926308 (p = 3.24e-06), was identified. The rs926308-T is associated with lower expression of RFPL3S, a gene important for neocortex development and implicated in arousal. Gene-based analyses performed using Sherlock Bayesian statistical integrative analysis also detected 20 genes with expression changes that may contribute to suicide risk. From analyzing publicly available transcriptomic data, ten of these genes have previous evidence of differential expression in suicide death or in psychiatric disorders that may be associated with suicide, including schizophrenia and autism (ZNF501, ZNF502, CNN3, IGF1R, KLHL36, NBL1, PDCD6IP, SNX19, BCAP29, and ARSA). Electronic health records (EHR) data was further merged to evaluate if there were clinically relevant subsets of suicide deaths associated with genetic variants. In summary, our study identified one risk locus and ten genes associated with suicide risk via gene expression, providing new insight into possible genetic and molecular mechanisms leading to suicide.
Collapse
Affiliation(s)
- Seonggyun Han
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA.
| | - Emily DiBlasi
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Eric T Monson
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Andrey Shabalin
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Elliott Ferris
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Danli Chen
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Alison Fraser
- Pedigree & Population Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Zhe Yu
- Pedigree & Population Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael Staley
- Office of the Medical Examiner, Utah Department of Health and Human Services, Salt Lake City, UT, USA
| | - W Brandon Callor
- Office of the Medical Examiner, Utah Department of Health and Human Services, Salt Lake City, UT, USA
| | - Erik D Christensen
- Office of the Medical Examiner, Utah Department of Health and Human Services, Salt Lake City, UT, USA
| | - David K Crockett
- Clinical Analytics, Intermountain Health, Salt Lake City, UT, USA
| | - Qingqin S Li
- Neuroscience Therapeutic Area, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Virginia Willour
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Amanda V Bakian
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Brooks Keeshin
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Anna R Docherty
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Karen Eilbeck
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hilary Coon
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
67
|
Zhang W, Zhang M, Xu Z, Yan H, Wang H, Jiang J, Wan J, Tang B, Liu C, Chen C, Meng Q. Human forebrain organoid-based multi-omics analyses of PCCB as a schizophrenia associated gene linked to GABAergic pathways. Nat Commun 2023; 14:5176. [PMID: 37620341 PMCID: PMC10449845 DOI: 10.1038/s41467-023-40861-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Identifying genes whose expression is associated with schizophrenia (SCZ) risk by transcriptome-wide association studies (TWAS) facilitates downstream experimental studies. Here, we integrated multiple published datasets of TWAS, gene coexpression, and differential gene expression analysis to prioritize SCZ candidate genes for functional study. Convergent evidence prioritized Propionyl-CoA Carboxylase Subunit Beta (PCCB), a nuclear-encoded mitochondrial gene, as an SCZ risk gene. However, the PCCB's contribution to SCZ risk has not been investigated before. Using dual luciferase reporter assay, we identified that SCZ-associated SNPs rs6791142 and rs35874192, two eQTL SNPs for PCCB, showed differential allelic effects on transcriptional activities. PCCB knockdown in human forebrain organoids (hFOs) followed by RNA sequencing analysis revealed dysregulation of genes enriched with multiple neuronal functions including gamma-aminobutyric acid (GABA)-ergic synapse. The metabolomic and mitochondrial function analyses confirmed the decreased GABA levels resulted from inhibited tricarboxylic acid cycle in PCCB knockdown hFOs. Multielectrode array recording analysis showed that PCCB knockdown in hFOs resulted into SCZ-related phenotypes including hyper-neuroactivities and decreased synchronization of neural network. In summary, this study utilized hFOs-based multi-omics analyses and revealed that PCCB downregulation may contribute to SCZ risk through regulating GABAergic pathways, highlighting the mitochondrial function in SCZ.
Collapse
Affiliation(s)
- Wendiao Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
| | - Ming Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhenhong Xu
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
| | - Hongye Yan
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
| | - Huimin Wang
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
| | - Jiamei Jiang
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
| | - Juan Wan
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
| | - Beisha Tang
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Chao Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Molecular Precision Medicine, Central South University, Changsha, Hunan, 410008, China.
| | - Qingtuan Meng
- The First Affiliated Hospital, Multi-Omics Research Center for Brain Disorders, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China.
- The First Affiliated Hospital, Clinical Research Center for Immune-Related Encephalopathy of Hunan Province, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China.
- The First Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, 421000, Hengyang, Hunan, China.
- MOE Key Lab of Rare Pediatric Diseases & School of Life Sciences, University of South China, 421001, Hengyang, Hunan, China.
| |
Collapse
|
68
|
Ollà I, Pardiñas AF, Parras A, Hernández IH, Santos-Galindo M, Picó S, Callado LF, Elorza A, Rodríguez-López C, Fernández-Miranda G, Belloc E, Walters JTR, O'Donovan MC, Méndez R, Toma C, Meana JJ, Owen MJ, Lucas JJ. Pathogenic Mis-splicing of CPEB4 in Schizophrenia. Biol Psychiatry 2023; 94:341-351. [PMID: 36958377 DOI: 10.1016/j.biopsych.2023.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/16/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Schizophrenia (SCZ) is caused by an interplay of polygenic risk and environmental factors, which may alter regulators of gene expression leading to pathogenic misexpression of SCZ risk genes. The CPEB family of RNA-binding proteins (CPEB1-4) regulates translation of target RNAs (approximately 40% of overall genes). We previously identified CPEB4 as a key dysregulated translational regulator in autism spectrum disorder (ASD) because its neuronal-specific microexon (exon 4) is mis-spliced in ASD brains, causing underexpression of numerous ASD risk genes. The genetic factors and pathogenic mechanisms shared between SCZ and ASD led us to hypothesize CPEB4 mis-splicing in SCZ leading to underexpression of multiple SCZ-related genes. METHODS We performed MAGMA-enrichment analysis on Psychiatric Genomics Consortium genome-wide association study data and analyzed RNA sequencing data from the PsychENCODE Consortium. Reverse transcriptase polymerase chain reaction and Western blot were performed on postmortem brain tissue, and the presence/absence of antipsychotics was assessed through toxicological analysis. Finally, mice with mild overexpression of exon 4-lacking CPEB4 (CPEB4Δ4) were generated and analyzed biochemically and behaviorally. RESULTS First, we found enrichment of SCZ-associated genes for CPEB4-binder transcripts. We also found decreased usage of CPEB4 microexon in SCZ probands, which was correlated with decreased protein levels of CPEB4-target SCZ-associated genes only in antipsychotic-free individuals. Interestingly, differentially expressed genes fit those reported for SCZ, specifically in the SCZ probands with decreased CPEB4-microexon inclusion. Finally, we demonstrated that mice with mild overexpression of CPEB4Δ4 showed decreased protein levels of CPEB4-target SCZ genes and SCZ-linked behaviors. CONCLUSIONS We identified aberrant CPEB4 splicing and downstream misexpression of SCZ risk genes as a novel etiological mechanism in SCZ.
Collapse
Affiliation(s)
- Ivana Ollà
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio F Pardiñas
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Alberto Parras
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Ivó H Hernández
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - María Santos-Galindo
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Picó
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Biocruces Bizkaia Health Research Institute and Networking Research Center on Mental Health (Centro de investigación Biomédica en Red | Salud Mental), Leioa, Bizkaia, Spain
| | - Ainara Elorza
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Claudia Rodríguez-López
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo Fernández-Miranda
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Eulàlia Belloc
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - James T R Walters
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Raúl Méndez
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain; Institució Catalana de RIcerca i Estudis Avançats, Barcelona, Spain
| | - Claudio Toma
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Biocruces Bizkaia Health Research Institute and Networking Research Center on Mental Health (Centro de investigación Biomédica en Red | Salud Mental), Leioa, Bizkaia, Spain
| | - Michael J Owen
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - José J Lucas
- Center for Molecular Biology "Severo Ochoa," Spanish National Research Council/Autonomous University of Madrid, Madrid, Spain; Networking Research Center on Neurodegenerative Diseases (Centro de Investigación Biomédica en Red|Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
69
|
Sznajder L, Khan M, Tadross M, Ciesiołka A, Nutter C, Taylor K, Pearson C, Sobczak K, Lewis M, Swanson M, Yuen R. Autistic traits in myotonic dystrophy type 1 due to MBNL inhibition and RNA mis-splicing. RESEARCH SQUARE 2023:rs.3.rs-3221704. [PMID: 37645891 PMCID: PMC10462192 DOI: 10.21203/rs.3.rs-3221704/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Tandem repeat expansions are enriched in autism spectrum disorder, including CTG expansion in the DMPK gene that underlines myotonic muscular dystrophy type 1. Although the clinical connection of autism to myotonic dystrophy is corroborated, the molecular links remained unknown. Here, we show a mechanistic path of autism via repeat expansion in myotonic dystrophy. We found that inhibition of muscleblind-like (MBNL) splicing factors by expanded CUG RNAs alerts the splicing of autism-risk genes during brain development especially a class of autism-relevant microexons. To provide in vivo evidence that the CTG expansion and MBNL inhibition axis leads to the presentation of autistic traits, we demonstrate that CTG expansion and MBNL-null mouse models recapitulate autism-relevant mis-splicing profiles and demonstrate social deficits. Our findings indicate that DMPK CTG expansion-associated autism arises from developmental mis-splicing. Understanding this pathomechanistic connection provides an opportunity for greater in-depth investigations of mechanistic threads in autism.
Collapse
|
70
|
Woodward DJ, Thorp JG, Akosile W, Ong JS, Gamazon ER, Derks EM, Gerring ZF. Identification of drug repurposing candidates for the treatment of anxiety: A genetic approach. Psychiatry Res 2023; 326:115343. [PMID: 37473490 PMCID: PMC10493169 DOI: 10.1016/j.psychres.2023.115343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/03/2023] [Accepted: 07/10/2023] [Indexed: 07/22/2023]
Abstract
Anxiety disorders are a group of prevalent and heritable neuropsychiatric diseases. We previously conducted a genome-wide association study (GWAS) which identified genomic loci associated with anxiety; however, the biological consequences underlying the genetic associations are largely unknown. Integrating GWAS and functional genomic data may improve our understanding of the genetic effects on intermediate molecular phenotypes such as gene expression. This can provide an opportunity for the discovery of drug targets for anxiety via drug repurposing. We used the GWAS summary statistics to determine putative causal genes for anxiety using MAGMA and colocalization analyses. A transcriptome-wide association study was conducted to identify genes with differential genetically regulated levels of gene expression in human brain tissue. The genes were integrated with a large drug-gene expression database (Connectivity Map), discovering compounds that are predicted to "normalise" anxiety-associated expression changes. The study identified 64 putative causal genes associated with anxiety (35 genes upregulated; 29 genes downregulated). Drug mechanisms adrenergic receptor agonists, sigma receptor agonists, and glutamate receptor agonists gene targets were enriched in anxiety-associated genetic signal and exhibited an opposing effect on the anxiety-associated gene expression signature. The significance of the project demonstrated genetic links for novel drug candidates to potentially advance anxiety therapeutics.
Collapse
Affiliation(s)
- Damian J Woodward
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; School of Biomedical Science, Queensland University of Technology, Kelvin Grove, QLD, Australia.
| | - Jackson G Thorp
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Wole Akosile
- School of Medicine, University of Queensland, St Lucia, QLD, Australia
| | - Jue-Sheng Ong
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Centre, Nashville, TN, USA; Clare Hall, University of Cambridge, Cambridge, UK
| | - Eske M Derks
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Zachary F Gerring
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.
| |
Collapse
|
71
|
Abstract
OBJECTIVE Due to the phenotypic heterogeneity and etiological complexity of bipolar disorder (BD), many patients do not respond well to the current medications, and developing novel effective treatment is necessary. Whether any BD genome-wide association study (GWAS) risk genes were targets of existing drugs or novel drugs that can be repurposed in the clinical treatment of BD is a hot topic in the GWAS era of BD. METHODS A list of 425 protein-coding BD risk genes was distilled through the BD GWAS, and 4479 protein-coding druggable targets were retrieved from the druggable genome. The overlapped genes/targets were subjected to further analyses in DrugBank, Pharos, and DGIdb datasets in terms of their FDA status, mechanism of action and primary indication, to identify their potential for repurposing. RESULTS We identified 58 BD GWAS risk genes grouped as the druggable targets, and several genes were given higher priority. These BD risk genes were targets of antipsychotics, antidepressants, antiepileptics, calcium channel antagonists, as well as anxiolytics and analgesics, either existing clinically-approved drugs for BD or the drugs than can be repurposed for treatment of BD in the future. Those genes were also likely relevant to BD pathophysiology, as many of them encode ion channel, ion transporter or neurotransmitter receptor, or the mice manipulating those genes are likely to mimic the phenotypes manifest in BD patients. CONCLUSIONS This study identifies several targets that may facilitate the discovery of novel treatments in BD, and implies the value of conducting GWAS into clinical translation.
Collapse
Affiliation(s)
- Hao-Xiang Qi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
72
|
Antunes AS, Martins-de-Souza D. Single-Cell RNA Sequencing and Its Applications in the Study of Psychiatric Disorders. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:329-339. [PMID: 37519459 PMCID: PMC10382703 DOI: 10.1016/j.bpsgos.2022.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022] Open
Abstract
Neuroscience is currently one of the most challenging research fields owing to the enormous complexity of the mammalian nervous system. We are yet to understand precise transcriptional programs that govern cell fate during neurodevelopment, resolve the connectome of the mammalian brain, and determine the etiology of various neurodegenerative and psychiatric disorders. Technological advances in the past decade, notably single-cell RNA sequencing, have enabled huge progress in our understanding of such features. Our current knowledge of the transcriptome is largely derived from bulk RNA sequencing, which reveals only the average gene expression of millions of cells, potentially missing out on minor transcriptome differences between cells detectable only at single-cell resolution. Since 2009, several single-cell RNA sequencing techniques have emerged that enable the accurate classification of neuronal and glial cell subtypes beyond classical molecular markers and electrophysiological features and allow the identification of previously unknown cell types. Furthermore, it enables the interrogation of molecular and disease-relevant mechanisms and offers further possibilities for the discovery of new drug targets and disease biomarkers. This review intends to familiarize the reader with the main single-cell RNA sequencing techniques developed throughout the past decade and discusses their application in the fields of brain cell taxonomy, neurodevelopment, and psychiatric disorders.
Collapse
Affiliation(s)
- André S.L.M. Antunes
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster, University of Campinas, Campinas, Brazil
- D'Or Institute for Research and Education, Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria, Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
| |
Collapse
|
73
|
Wu Y, Song J, Liu M, Ma H, Zhang J. Integrating GWAS and proteome data to identify novel drug targets for MU. Sci Rep 2023; 13:10437. [PMID: 37369724 DOI: 10.1038/s41598-023-37177-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Mouth ulcers have been associated with numerous loci in genome wide association studies (GWAS). Nonetheless, it remains unclear what mechanisms are involved in the pathogenesis of mouth ulcers at these loci, as well as what the most effective ulcer drugs are. Thus, we aimed to screen hub genes responsible for mouth ulcer pathogenesis. We conducted an imputed/in-silico proteome-wide association study to discover candidate genes that impact the development of mouth ulcers and affect the expression and concentration of associated proteins in the bloodstream. The integrative analysis revealed that 35 genes play a significant role in the development of mouth ulcers, both in terms of their protein and transcriptional levels. Following this analysis, the researchers identified 6 key genes, namely BTN3A3, IL12B, BPI, FAM213A, PLXNB2, and IL22RA2, which were related to the onset of mouth ulcers. By combining with multidimensional data, six genes were found to correlate with mouth ulcer pathogenesis, which can be useful for further biological and therapeutic research.
Collapse
Affiliation(s)
- Yadong Wu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China.
| | - Manyi Liu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Hong Ma
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China.
| | - Junmei Zhang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, 550002, China.
| |
Collapse
|
74
|
Boltz T, Schwarz T, Bot M, Hou K, Caggiano C, Lapinska S, Duan C, Boks MP, Kahn RS, Zaitlen N, Pasaniuc B, Ophoff R. Cell type deconvolution of bulk blood RNA-Seq to reveal biological insights of neuropsychiatric disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542156. [PMID: 37293101 PMCID: PMC10245943 DOI: 10.1101/2023.05.24.542156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Genome-wide association studies (GWAS) have uncovered susceptibility loci associated with psychiatric disorders like bipolar disorder (BP) and schizophrenia (SCZ). However, most of these loci are in non-coding regions of the genome with unknown causal mechanisms of the link between genetic variation and disease risk. Expression quantitative trait loci (eQTL) analysis of bulk tissue is a common approach to decipher underlying mechanisms, though this can obscure cell-type specific signals thus masking trait-relevant mechanisms. While single-cell sequencing can be prohibitively expensive in large cohorts, computationally inferred cell type proportions and cell type gene expression estimates have the potential to overcome these problems and advance mechanistic studies. Using bulk RNA-Seq from 1,730 samples derived from whole blood in a cohort ascertained for individuals with BP and SCZ this study estimated cell type proportions and their relation with disease status and medication. We found between 2,875 and 4,629 eGenes for each cell type, including 1,211 eGenes that are not found using bulk expression alone. We performed a colocalization test between cell type eQTLs and various traits and identified hundreds of associations between cell type eQTLs and GWAS loci that are not detected in bulk eQTLs. Finally, we investigated the effects of lithium use on cell type expression regulation and found examples of genes that are differentially regulated dependent on lithium use. Our study suggests that computational methods can be applied to large bulk RNA-Seq datasets of non-brain tissue to identify disease-relevant, cell type specific biology of psychiatric disorders and psychiatric medication.
Collapse
Affiliation(s)
- Toni Boltz
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Merel Bot
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Kangcheng Hou
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Christa Caggiano
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Sandra Lapinska
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Chenda Duan
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, USA
| | - Marco P Boks
- Department of Psychiatry, Brain Center University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Rene S Kahn
- Department of Psychiatry, Brain Center University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
- Department of Psychiatry, Icahn School of Medicine, Mount Sinai, NY, USA
| | - Noah Zaitlen
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Bogdan Pasaniuc
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Roel Ophoff
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
75
|
Maden SK, Kwon SH, Huuki-Myers LA, Collado-Torres L, Hicks SC, Maynard KR. Challenges and opportunities to computationally deconvolve heterogeneous tissue with varying cell sizes using single cell RNA-sequencing datasets. ARXIV 2023:arXiv:2305.06501v1. [PMID: 37214135 PMCID: PMC10197733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Deconvolution of cell mixtures in "bulk" transcriptomic samples from homogenate human tissue is important for understanding the pathologies of diseases. However, several experimental and computational challenges remain in developing and implementing transcriptomics-based deconvolution approaches, especially those using a single cell/nuclei RNA-seq reference atlas, which are becoming rapidly available across many tissues. Notably, deconvolution algorithms are frequently developed using samples from tissues with similar cell sizes. However, brain tissue or immune cell populations have cell types with substantially different cell sizes, total mRNA expression, and transcriptional activity. When existing deconvolution approaches are applied to these tissues, these systematic differences in cell sizes and transcriptomic activity confound accurate cell proportion estimates and instead may quantify total mRNA content. Furthermore, there is a lack of standard reference atlases and computational approaches to facilitate integrative analyses, including not only bulk and single cell/nuclei RNA-seq data, but also new data modalities from spatial -omic or imaging approaches. New multi-assay datasets need to be collected with orthogonal data types generated from the same tissue block and the same individual, to serve as a "gold standard" for evaluating new and existing deconvolution methods. Below, we discuss these key challenges and how they can be addressed with the acquisition of new datasets and approaches to analysis.
Collapse
Affiliation(s)
- Sean K Maden
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | | | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
76
|
Guo Q, Wu S, Geschwind DH. Characterization of Gene Regulatory Elements in Human Fetal Cortical Development: Enhancing Our Understanding of Neurodevelopmental Disorders and Evolution. Dev Neurosci 2023; 46:69-83. [PMID: 37231806 DOI: 10.1159/000530929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
The neocortex is the region that most distinguishes human brain from other mammals and primates [Annu Rev Genet. 2021 Nov;55(1):555-81]. Studying the development of human cortex is important in understanding the evolutionary changes occurring in humans relative to other primates, as well as in elucidating mechanisms underlying neurodevelopmental disorders. Cortical development is a highly regulated process, spatially and temporally coordinated by expression of essential transcriptional factors in response to signaling pathways [Neuron. 2019 Sep;103(6):980-1004]. Enhancers are the most well-understood cis-acting, non-protein-coding regulatory elements that regulate gene expression [Nat Rev Genet. 2014 Apr;15(4):272-86]. Importantly, given the conservation of both DNA sequence and molecular function of the majority of proteins across mammals [Genome Res. 2003 Dec;13(12):2507-18], enhancers [Science. 2015 Mar;347(6226):1155-9], which are far more divergent at the sequence level, likely account for the phenotypes that distinguish the human brain by changing the regulation of gene expression. In this review, we will revisit the conceptual framework of gene regulation during human brain development, as well as the evolution of technologies to study transcriptional regulation, with recent advances in genome biology that open a window allowing us to systematically characterize cis-regulatory elements in developing human brain [Hum Mol Genet. 2022 Oct;31(R1):R84-96]. We provide an update on work to characterize the suite of all enhancers in the developing human brain and the implications for understanding neuropsychiatric disorders. Finally, we discuss emerging therapeutic ideas that utilize our emerging knowledge of enhancer function.
Collapse
Affiliation(s)
- Qiuyu Guo
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA
- Center for Autism Research and Treatment, Semel Institute, University of California Los Angeles, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Sarah Wu
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA
| | - Daniel H Geschwind
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA
- Center for Autism Research and Treatment, Semel Institute, University of California Los Angeles, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Institute of Precision Health, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
77
|
Paff M, Grieco SF, Xu X. Obtaining brain tissue from living patients for psychiatry research: collaboration with patients with epilepsy and neurosurgeons. Lancet Psychiatry 2023; 10:381. [PMID: 37088087 DOI: 10.1016/s2215-0366(23)00142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023]
Affiliation(s)
- Michelle Paff
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA 92697, USA; Department of Neurosurgery, School of Medicine, University of California, Irvine, CA 92697, USA.
| | - Steven F Grieco
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA 92697, USA; Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Xiangmin Xu
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, CA 92697, USA; Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697, USA
| |
Collapse
|
78
|
Choudhury M, Fu T, Amoah K, Jun HI, Chan TW, Park S, Walker DW, Bahn JH, Xiao X. Widespread RNA hypoediting in schizophrenia and its relevance to mitochondrial function. SCIENCE ADVANCES 2023; 9:eade9997. [PMID: 37027465 PMCID: PMC10081846 DOI: 10.1126/sciadv.ade9997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/08/2023] [Indexed: 06/19/2023]
Abstract
RNA editing, the endogenous modification of nucleic acids, is known to be altered in genes with important neurological function in schizophrenia (SCZ). However, the global profile and molecular functions of disease-associated RNA editing remain unclear. Here, we analyzed RNA editing in postmortem brains of four SCZ cohorts and uncovered a significant and reproducible trend of hypoediting in patients of European descent. We report a set of SCZ-associated editing sites via WGCNA analysis, shared across cohorts. Using massively parallel reporter assays and bioinformatic analyses, we observed that differential 3' untranslated region (3'UTR) editing sites affecting host gene expression were enriched for mitochondrial processes. Furthermore, we characterized the impact of two recoding sites in the mitofusin 1 (MFN1) gene and showed their functional relevance to mitochondrial fusion and cellular apoptosis. Our study reveals a global reduction of editing in SCZ and a compelling link between editing and mitochondrial function in the disease.
Collapse
Affiliation(s)
- Mudra Choudhury
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Ting Fu
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Kofi Amoah
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Hyun-Ik Jun
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Tracey W. Chan
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Sungwoo Park
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Jae Hoon Bahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Xinshu Xiao
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
79
|
Bell AS, Wagner J, Rosoff DB, Lohoff FW. Proprotein convertase subtilisin/kexin type 9 (PCSK9) in the central nervous system. Neurosci Biobehav Rev 2023; 149:105155. [PMID: 37019248 DOI: 10.1016/j.neubiorev.2023.105155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023]
Abstract
The gene encoding proprotein convertase subtilisin/kexin type 9 (PCSK9) and its protein product have been widely studied for their role in cholesterol and lipid metabolism. PCSK9 increases the rate of metabolic degradation of low-density lipoprotein receptors, preventing the diffusion of low-density lipoprotein (LDL) from plasma into cells and contributes to high lipoprotein-bound cholesterol levels in the plasma. While most research has focused on the regulation and disease relevance of PCSK9 to the cardiovascular system and lipid metabolism, there is a growing body of evidence that PCSK9 plays a crucial role in pathogenic processes in other organ systems, including the central nervous system. PCSK9's impact on the brain is not yet fully understood, though several recent studies have sought to illuminate its impact on various neurodegenerative and psychiatric disorders, as well as its connection with ischemic stroke. Cerebral PCSK9 expression is low but is highly upregulated during disease states. Among others, PCSK9 is known to play a role in neurogenesis, neural cell differentiation, central LDL receptor metabolism, neural cell apoptosis, neuroinflammation, Alzheimer's Disease, Alcohol Use Disorder, and stroke. The PCSK9 gene contains several polymorphisms, including both gain-of-function and loss-of-function mutations which profoundly impact normal PCSK9 signaling and cholesterol metabolism. Gain-of-function mutations lead to persistent hypercholesterolemia and poor health outcomes, while loss-of-function mutations generally lead to hypocholesterolemia and may serve as a protective factor against diseases of the liver, cardiovascular system, and central nervous system. Recent genomic studies have sought to identify the end-organ effects of such mutations and continue to identify evidence of a much broader role for PCSK9 in extrahepatic organ systems. Despite this, there remain large gaps in our understanding of PCSK9, its regulation, and its effects on disease risk outside the liver. This review, which incorporates data from a wide range of scientific disciplines and experimental paradigms, is intended to describe PCSK9's role in the central nervous system as it relates to cerebral disease and neuropsychiatric disorders, and to examine the clinical potential of PCSK9 inhibitors and genetic variation in the PCSK9 gene on disease outcomes, including neurological and neuropsychiatric disease.
Collapse
|
80
|
Zhang W, Zhang M, Xu Z, Yan H, Wang H, Jiang J, Wan J, Tang B, Liu C, Chen C, Meng Q. Human forebrain organoids-based multi-omics analyses reveal PCCB's regulation on GABAergic system contributing to schizophrenia. RESEARCH SQUARE 2023:rs.3.rs-2674668. [PMID: 37034773 PMCID: PMC10081387 DOI: 10.21203/rs.3.rs-2674668/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Identifying genes whose expression is associated with schizophrenia (SCZ) risk by transcriptome-wide association studies (TWAS) facilitates downstream experimental studies. Here, we integrated multiple published datasets of TWAS (including FUSION, PrediXcan, summary-data-based Mendelian randomization (SMR), joint-tissue imputation approach with Mendelian randomization (MR-JTI)), gene coexpression, and differential gene expression analysis to prioritize SCZ candidate genes for functional study. Convergent evidence prioritized Propionyl-CoA Carboxylase Subunit Beta ( PCCB ), a nuclear-encoded mitochondrial gene, as an SCZ risk gene. However, the PCCB ’s contribution to SCZ risk has not been investigated before. Using dual luciferase reporter assay, we identified that SCZ-associated SNP rs35874192, an eQTL SNP for PCCB , showed differential allelic effects on transcriptional activities. PCCB knockdown in human forebrain organoids (hFOs) followed by RNA-seq revealed dysregulation of genes enriched with multiple neuronal functions including gamma-aminobutyric acid (GABA)-ergic synapse, as well as genes dysregulated in postmortem brains of SCZ patients or in cerebral organoids derived from SCZ patients. The metabolomic and mitochondrial function analyses confirmed the deceased GABA levels resulted from reduced tricarboxylic acid cycle in PCCB knockdown hFOs. Multielectrode array recording analysis showed that PCCB knockdown in hFOs resulted into SCZ-related phenotypes including hyper-neuroactivities and decreased synchronization of neural network. In summary, this study utilized hFOs-based multi-omics data and revealed that PCCB downregulation may contribute to SCZ risk through regulating GABAergic system, highlighting the mitochondrial function in SCZ.
Collapse
Affiliation(s)
- Wendiao Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University
| | - Ming Zhang
- School of Life Sciences, Central South University
| | - Zhenhong Xu
- The First Affiliated Hospital of University of South China
| | - Hongye Yan
- The First Affiliated Hospital of University of South China
| | - Huimin Wang
- The First Affiliated Hospital of University of South China
| | - Jiamei Jiang
- The First Affiliated Hospital of University of South China
| | - Juan Wan
- The First Affiliated Hospital of University of South China
| | | | | | | | - Qingtuan Meng
- The First Affiliated Hospital of University of South China
| |
Collapse
|
81
|
Dai R, Chu T, Zhang M, Wang X, Jourdon A, Wu F, Mariani J, Vaccarino FM, Lee D, Fullard JF, Hoffman GE, Roussos P, Wang Y, Wang X, Pinto D, Wang SH, Zhang C, Chen C, Liu C. Evaluating performance and applications of sample-wise cell deconvolution methods on human brain transcriptomic data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532468. [PMID: 36993743 PMCID: PMC10054947 DOI: 10.1101/2023.03.13.532468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Sample-wise deconvolution methods have been developed to estimate cell-type proportions and gene expressions in bulk-tissue samples. However, the performance of these methods and their biological applications has not been evaluated, particularly on human brain transcriptomic data. Here, nine deconvolution methods were evaluated with sample-matched data from bulk-tissue RNAseq, single-cell/nuclei (sc/sn) RNAseq, and immunohistochemistry. A total of 1,130,767 nuclei/cells from 149 adult postmortem brains and 72 organoid samples were used. The results showed the best performance of dtangle for estimating cell proportions and bMIND for estimating sample-wise cell-type gene expression. For eight brain cell types, 25,273 cell-type eQTLs were identified with deconvoluted expressions (decon-eQTLs). The results showed that decon-eQTLs explained more schizophrenia GWAS heritability than bulk-tissue or single-cell eQTLs alone. Differential gene expression associated with multiple phenotypes were also examined using the deconvoluted data. Our findings, which were replicated in bulk-tissue RNAseq and sc/snRNAseq data, provided new insights into the biological applications of deconvoluted data.
Collapse
Affiliation(s)
- Rujia Dai
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Tianyao Chu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Ming Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Xuan Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | | | - Feinan Wu
- Child Study Center, Yale University, New Haven, CT, USA
| | | | - Flora M Vaccarino
- Child Study Center, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Donghoon Lee
- Center for Disease Neurogenomics, Departments of Psychiatry and Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Departments of Psychiatry and Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Departments of Psychiatry and Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Departments of Psychiatry and Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, VA, USA
| | - Xusheng Wang
- Department of Biology, University of North Dakota, Grand Forks, ND, USA
| | - Dalila Pinto
- Department of Psychiatry, Department of Genetics and Genomic Sciences, Mindich Child Health and Development Institute, and Icahn Genomics Institute for Data Science and Genomic Technology, Seaver Autism Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sidney H Wang
- Center for Human Genetics, The Brown foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chunling Zhang
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Chao Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
82
|
Bressan E, Reed X, Bansal V, Hutchins E, Cobb MM, Webb MG, Alsop E, Grenn FP, Illarionova A, Savytska N, Violich I, Broeer S, Fernandes N, Sivakumar R, Beilina A, Billingsley KJ, Berghausen J, Pantazis CB, Pitz V, Patel D, Daida K, Meechoovet B, Reiman R, Courtright-Lim A, Logemann A, Antone J, Barch M, Kitchen R, Li Y, Dalgard CL, The American Genome Center, Rizzu P, Hernandez DG, Hjelm BE, Nalls M, Gibbs JR, Finkbeiner S, Cookson MR, Van Keuren-Jensen K, Craig DW, Singleton AB, Heutink P, Blauwendraat C. The Foundational Data Initiative for Parkinson Disease: Enabling efficient translation from genetic maps to mechanism. CELL GENOMICS 2023; 3:100261. [PMID: 36950378 PMCID: PMC10025424 DOI: 10.1016/j.xgen.2023.100261] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/22/2022] [Accepted: 01/12/2023] [Indexed: 02/08/2023]
Abstract
The Foundational Data Initiative for Parkinson Disease (FOUNDIN-PD) is an international collaboration producing fundamental resources for Parkinson disease (PD). FOUNDIN-PD generated a multi-layered molecular dataset in a cohort of induced pluripotent stem cell (iPSC) lines differentiated to dopaminergic (DA) neurons, a major affected cell type in PD. The lines were derived from the Parkinson's Progression Markers Initiative study, which included participants with PD carrying monogenic PD variants, variants with intermediate effects, and variants identified by genome-wide association studies and unaffected individuals. We generated genetic, epigenetic, regulatory, transcriptomic, and longitudinal cellular imaging data from iPSC-derived DA neurons to understand molecular relationships between disease-associated genetic variation and proximate molecular events. These data reveal that iPSC-derived DA neurons provide a valuable cellular context and foundational atlas for modeling PD genetic risk. We have integrated these data into a FOUNDIN-PD data browser as a resource for understanding the molecular pathogenesis of PD.
Collapse
Affiliation(s)
| | - Xylena Reed
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Vikas Bansal
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Elizabeth Hutchins
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Melanie M. Cobb
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
| | - Michelle G. Webb
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA, USA
| | - Eric Alsop
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Francis P. Grenn
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | - Natalia Savytska
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ivo Violich
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA, USA
| | - Stefanie Broeer
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Noémia Fernandes
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ramiyapriya Sivakumar
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA, USA
| | - Alexandra Beilina
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Kimberley J. Billingsley
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Joos Berghausen
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Caroline B. Pantazis
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Vanessa Pitz
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Dhairya Patel
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Kensuke Daida
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Bessie Meechoovet
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Rebecca Reiman
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Amanda Courtright-Lim
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Amber Logemann
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Jerry Antone
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Mariya Barch
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
| | - Robert Kitchen
- Massachusetts General Hospital, Cardiovascular Research Center, Charlestown, MA, USA
| | - Yan Li
- Protein/Peptide Sequencing Facility, National Institute of Neurological, Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Clifton L. Dalgard
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - The American Genome Center
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA, USA
- Massachusetts General Hospital, Cardiovascular Research Center, Charlestown, MA, USA
- Protein/Peptide Sequencing Facility, National Institute of Neurological, Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Dena G. Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Brooke E. Hjelm
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA, USA
| | - Mike Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
| | - J. Raphael Gibbs
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Mark R. Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | | - David W. Craig
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA, USA
| | - Andrew B. Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
83
|
Hatoum AS, Colbert SM, Johnson EC, Huggett SB, Deak JD, Pathak G, Jennings MV, Paul SE, Karcher NR, Hansen I, Baranger DA, Edwards A, Grotzinger A, Substance Use Disorder Working Group of the Psychiatric Genomics
Consortium, Tucker-Drob EM, Kranzler HR, Davis LK, Sanchez-Roige S, Polimanti R, Gelernter J, Edenberg HJ, Bogdan R, Agrawal A. Multivariate genome-wide association meta-analysis of over 1 million subjects identifies loci underlying multiple substance use disorders. NATURE. MENTAL HEALTH 2023; 1:210-223. [PMID: 37250466 PMCID: PMC10217792 DOI: 10.1038/s44220-023-00034-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/10/2023] [Indexed: 05/31/2023]
Abstract
Genetic liability to substance use disorders can be parsed into loci that confer general or substance-specific addiction risk. We report a multivariate genome-wide association meta-analysis that disaggregates general and substance-specific loci for published summary statistics of problematic alcohol use, problematic tobacco use, cannabis use disorder, and opioid use disorder in a sample of 1,025,550 individuals of European descent and 92,630 individuals of African descent. Nineteen independent SNPs were genome-wide significant (P < 5e-8) for the general addiction risk factor (addiction-rf), which showed high polygenicity. Across ancestries, PDE4B was significant (among other genes), suggesting dopamine regulation as a cross-substance vulnerability. An addiction-rf polygenic risk score was associated with substance use disorders, psychopathologies, somatic conditions, and environments associated with the onset of addictions. Substance-specific loci (9 for alcohol, 32 for tobacco, 5 for cannabis, 1 for opioids) included metabolic and receptor genes. These findings provide insight into genetic risk loci for substance use disorders that could be leveraged as treatment targets.
Collapse
Affiliation(s)
- Alexander S. Hatoum
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Sarah M.C. Colbert
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Emma C. Johnson
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | | | - Joseph D. Deak
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven,
CT, USA
| | - Gita Pathak
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
| | - Mariela V. Jennings
- UC San Diego School of Medicine, Department of Psychiatry,
San Diego, CA, USA
| | - Sarah E. Paul
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - Nicole R. Karcher
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Isabella Hansen
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - David A.A. Baranger
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Alexis Edwards
- Virginia Institute of Psychiatric and Behavioral Genetics,
Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Grotzinger
- University of Colorado-Boulder, Institute for Behavioral
Genetics, Boulder, CO, USA
| | | | - Elliot M. Tucker-Drob
- University of Texas at Austin, Department of Psychology and
Population Research Center, Austin, TX, USA
| | - Henry R. Kranzler
- Center for Studies of Addiction, Department of
Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia,
PA, USA
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, USA
| | - Lea K. Davis
- Department of Medicine, Division of Genetic Medicine,
Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences,
Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt
University Medical Center, Nashville, TN, USA
| | - Sandra Sanchez-Roige
- UC San Diego School of Medicine, Department of Psychiatry,
San Diego, CA, USA
- Department of Medicine, Division of Genetic Medicine,
Vanderbilt University, Nashville, TN, USA
| | - Renato Polimanti
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven,
CT, USA
| | - Joel Gelernter
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- University of Texas at Austin, Department of Psychology and
Population Research Center, Austin, TX, USA
- Department of Genetics, Yale School of Medicine, New
Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New
Haven, CT, USA
| | - Howard J. Edenberg
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana
University School of Medicine, Indianapolis, IN, USA
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - Arpana Agrawal
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| |
Collapse
|
84
|
Johnson EC, Kapoor M, Hatoum AS, Zhou H, Polimanti R, Wendt FR, Walters RK, Lai D, Kember RL, Hartz S, Meyers JL, Peterson RE, Ripke S, Bigdeli TB, Fanous AH, Pato CN, Pato MT, Goate AM, Kranzler HR, O'Donovan MC, Walters JTR, Gelernter J, Edenberg HJ, Agrawal A. Investigation of convergent and divergent genetic influences underlying schizophrenia and alcohol use disorder. Psychol Med 2023; 53:1196-1204. [PMID: 34231451 PMCID: PMC8738774 DOI: 10.1017/s003329172100266x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Alcohol use disorder (AUD) and schizophrenia (SCZ) frequently co-occur, and large-scale genome-wide association studies (GWAS) have identified significant genetic correlations between these disorders. METHODS We used the largest published GWAS for AUD (total cases = 77 822) and SCZ (total cases = 46 827) to identify genetic variants that influence both disorders (with either the same or opposite direction of effect) and those that are disorder specific. RESULTS We identified 55 independent genome-wide significant single nucleotide polymorphisms with the same direction of effect on AUD and SCZ, 8 with robust effects in opposite directions, and 98 with disorder-specific effects. We also found evidence for 12 genes whose pleiotropic associations with AUD and SCZ are consistent with mediation via gene expression in the prefrontal cortex. The genetic covariance between AUD and SCZ was concentrated in genomic regions functional in brain tissues (p = 0.001). CONCLUSIONS Our findings provide further evidence that SCZ shares meaningful genetic overlap with AUD.
Collapse
Affiliation(s)
- Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Manav Kapoor
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander S Hatoum
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hang Zhou
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Renato Polimanti
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Frank R Wendt
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rachel L Kember
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, USA
| | - Sarah Hartz
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jacquelyn L Meyers
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Henri Begleiter Neurodynamics Laboratory, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Roseann E Peterson
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Tim B Bigdeli
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Ayman H Fanous
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Carlos N Pato
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Michele T Pato
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Alison M Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Henry R Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, USA
| | - Michael C O'Donovan
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| | - James T R Walters
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| | - Joel Gelernter
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Howard J Edenberg
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
85
|
Yang G, Mishra M, Perera MA. Multi-Omics Studies in Historically Excluded Populations: The Road to Equity. Clin Pharmacol Ther 2023; 113:541-556. [PMID: 36495075 PMCID: PMC10323857 DOI: 10.1002/cpt.2818] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
Over the past few decades, genomewide association studies (GWASs) have identified the specific genetics variants contributing to many complex diseases by testing millions of genetic variations across the human genome against a variety of phenotypes. However, GWASs are limited in their ability to uncover mechanistic insight given that most significant associations are found in non-coding region of the genome. Furthermore, the lack of diversity in studies has stymied the advance of precision medicine for many historically excluded populations. In this review, we summarize most popular multi-omics approaches (genomics, transcriptomics, proteomics, and metabolomics) related to precision medicine and highlight if diverse populations have been included and how their findings have advance biological understanding of disease and drug response. New methods that incorporate local ancestry have been to improve the power of GWASs for admixed populations (such as African Americans and Latinx). Because most signals from GWAS are in the non-coding region, other machine learning and omics approaches have been developed to identify the potential causative single-nucleotide polymorphisms and genes that explain these phenotypes. These include polygenic risk scores, expression quantitative trait locus mapping, and transcriptome-wide association studies. Analogous protein methods, such as proteins quantitative trait locus mapping, proteome-wide association studies, and metabolomic approaches provide insight into the consequences of genetic variation on protein abundance. Whereas, integrated multi-omics studies have improved our understanding of the mechanisms for genetic association, we still lack the datasets and cohorts for historically excluded populations to provide equity in precision medicine and pharmacogenomics.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mrinal Mishra
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Minoli A. Perera
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
86
|
Arnatkeviciute A, Markello RD, Fulcher BD, Misic B, Fornito A. Toward Best Practices for Imaging Transcriptomics of the Human Brain. Biol Psychiatry 2023; 93:391-404. [PMID: 36725139 DOI: 10.1016/j.biopsych.2022.10.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/03/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Modern brainwide transcriptional atlases provide unprecedented opportunities for investigating the molecular correlates of brain organization, as quantified using noninvasive neuroimaging. However, integrating neuroimaging data with transcriptomic measures is not straightforward, and careful consideration is required to make valid inferences. In this article, we review recent work exploring how various methodological choices affect 3 main phases of imaging transcriptomic analyses, including 1) processing of transcriptional atlas data; 2) relating transcriptional measures to independently derived neuroimaging phenotypes; and 3) evaluating the functional implications of identified associations through gene enrichment analyses. Our aim is to facilitate the development of standardized and reproducible approaches for this rapidly growing field. We identify sources of methodological variability, key choices that can affect findings, and considerations for mitigating false positive and/or spurious results. Finally, we provide an overview of freely available open-source toolboxes implementing current best-practice procedures across all 3 analysis phases.
Collapse
Affiliation(s)
- Aurina Arnatkeviciute
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Melbourne, Victoria, Australia.
| | - Ross D Markello
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Ben D Fulcher
- School of Physics, The University of Sydney, Sydney, New South Wales, Australia
| | - Bratislav Misic
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
87
|
He C, Kalafut NC, Sandoval SO, Risgaard R, Sirois CL, Yang C, Khullar S, Suzuki M, Huang X, Chang Q, Zhao X, Sousa AM, Wang D. BOMA, a machine-learning framework for comparative gene expression analysis across brains and organoids. CELL REPORTS METHODS 2023; 3:100409. [PMID: 36936070 PMCID: PMC10014309 DOI: 10.1016/j.crmeth.2023.100409] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/21/2022] [Accepted: 01/25/2023] [Indexed: 02/17/2023]
Abstract
Our machine-learning framework, brain and organoid manifold alignment (BOMA), first performs a global alignment of developmental gene expression data between brains and organoids. It then applies manifold learning to locally refine the alignment, revealing conserved and specific developmental trajectories across brains and organoids. Using BOMA, we found that human cortical organoids better align with certain brain cortical regions than with other non-cortical regions, implying organoid-preserved developmental gene expression programs specific to brain regions. Additionally, our alignment of non-human primate and human brains reveals highly conserved gene expression around birth. Also, we integrated and analyzed developmental single-cell RNA sequencing (scRNA-seq) data of human brains and organoids, showing conserved and specific cell trajectories and clusters. Further identification of expressed genes of such clusters and enrichment analyses reveal brain- or organoid-specific developmental functions and pathways. Finally, we experimentally validated important specific expressed genes through the use of immunofluorescence. BOMA is open-source available as a web tool for community use.
Collapse
Affiliation(s)
- Chenfeng He
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Noah Cohen Kalafut
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Soraya O. Sandoval
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Risgaard
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Carissa L. Sirois
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Chen Yang
- Department of Mathematics, University of Wisconsin-Madison, Madison, WI, USA
| | - Saniya Khullar
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Marin Suzuki
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Xiang Huang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Departments of Medical Genetics and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Andre M.M. Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Daifeng Wang
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
88
|
Li S, Yan B, Li TKT, Lu J, Gu Y, Tan Y, Gong F, Lam TW, Xie P, Wang Y, Lin G, Luo R. Ultra-low-coverage genome-wide association study-insights into gestational age using 17,844 embryo samples with preimplantation genetic testing. Genome Med 2023; 15:10. [PMID: 36788602 PMCID: PMC9926832 DOI: 10.1186/s13073-023-01158-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Very low-coverage (0.1 to 1×) whole genome sequencing (WGS) has become a promising and affordable approach to discover genomic variants of human populations for genome-wide association study (GWAS). To support genetic screening using preimplantation genetic testing (PGT) in a large population, the sequencing coverage goes below 0.1× to an ultra-low level. However, the feasibility and effectiveness of ultra-low-coverage WGS (ulcWGS) for GWAS remains undetermined. METHODS We built a pipeline to carry out analysis of ulcWGS data for GWAS. To examine its effectiveness, we benchmarked the accuracy of genotype imputation at the combination of different coverages below 0.1× and sample sizes from 2000 to 16,000, using 17,844 embryo PGT samples with approximately 0.04× average coverage and the standard Chinese sample HG005 with known genotypes. We then applied the imputed genotypes of 1744 transferred embryos who have gestational ages and complete follow-up records to GWAS. RESULTS The accuracy of genotype imputation under ultra-low coverage can be improved by increasing the sample size and applying a set of filters. From 1744 born embryos, we identified 11 genomic risk loci associated with gestational ages and 166 genes mapped to these loci according to positional, expression quantitative trait locus, and chromatin interaction strategies. Among these mapped genes, CRHBP, ICAM1, and OXTR were more frequently reported as preterm birth related. By joint analysis of gene expression data from previous studies, we constructed interrelationships of mainly CRHBP, ICAM1, PLAGL1, DNMT1, CNTLN, DKK1, and EGR2 with preterm birth, infant disease, and breast cancer. CONCLUSIONS This study not only demonstrates that ulcWGS could achieve relatively high accuracy of adequate genotype imputation and is capable of GWAS, but also provides insights into the associations between gestational age and genetic variations of the fetal embryos from Chinese population.
Collapse
Affiliation(s)
- Shumin Li
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Bin Yan
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Thomas K. T. Li
- grid.415550.00000 0004 1764 4144Department of Obstetrics & Gynecology, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Jianliang Lu
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Yifan Gu
- grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008 Hunan China ,grid.477823.d0000 0004 1756 593XClinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013 Hunan China
| | - Yueqiu Tan
- grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008 Hunan China ,grid.477823.d0000 0004 1756 593XClinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013 Hunan China
| | - Fei Gong
- grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008 Hunan China ,grid.477823.d0000 0004 1756 593XClinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013 Hunan China
| | - Tak-Wah Lam
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Pingyuan Xie
- Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China. .,National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China.
| | - Yuexuan Wang
- Department of Computer Science, The University of Hong Kong, Hong Kong, China. .,College of Computer Science and Technology, Zhejiang University, Hangzhou, China.
| | - Ge Lin
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008, Hunan, China. .,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013, Hunan, China. .,National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China.
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
89
|
Chen Y, Dai J, Tang L, Mikhailova T, Liang Q, Li M, Zhou J, Kopp RF, Weickert C, Chen C, Liu C. Neuroimmune transcriptome changes in patient brains of psychiatric and neurological disorders. Mol Psychiatry 2023; 28:710-721. [PMID: 36424395 PMCID: PMC9911365 DOI: 10.1038/s41380-022-01854-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 10/07/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2022]
Abstract
Neuroinflammation has been implicated in multiple brain disorders but the extent and the magnitude of change in immune-related genes (IRGs) across distinct brain disorders has not been directly compared. In this study, 1275 IRGs were curated and their expression changes investigated in 2467 postmortem brains of controls and patients with six major brain disorders, including schizophrenia (SCZ), bipolar disorder (BD), autism spectrum disorder (ASD), major depressive disorder (MDD), Alzheimer's disease (AD), and Parkinson's disease (PD). There were 865 IRGs present across all microarray and RNA-seq datasets. More than 60% of the IRGs had significantly altered expression in at least one of the six disorders. The differentially expressed immune-related genes (dIRGs) shared across disorders were mainly related to innate immunity. Moreover, sex, tissue, and putative cell type were systematically evaluated for immune alterations in different neuropsychiatric disorders. Co-expression networks revealed that transcripts of the neuroimmune systems interacted with neuronal-systems, both of which contribute to the pathology of brain disorders. However, only a few genes with expression changes were also identified as containing risk variants in genome-wide association studies. The transcriptome alterations at gene and network levels may clarify the immune-related pathophysiology and help to better define neuropsychiatric and neurological disorders.
Collapse
Affiliation(s)
- Yu Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jiacheng Dai
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
| | - Longfei Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tatiana Mikhailova
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Qiuman Liang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Miao Li
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiaqi Zhou
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Richard F Kopp
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Cynthia Weickert
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
- School of Psychiatry, UNSW, Sydney, NSW, Australia
| | - Chao Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China.
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
90
|
Sathyanarayanan A, Mueller TT, Ali Moni M, Schueler K, Baune BT, Lio P, Mehta D, Baune BT, Dierssen M, Ebert B, Fabbri C, Fusar-Poli P, Gennarelli M, Harmer C, Howes OD, Janzing JGE, Lio P, Maron E, Mehta D, Minelli A, Nonell L, Pisanu C, Potier MC, Rybakowski F, Serretti A, Squassina A, Stacey D, van Westrhenen R, Xicota L. Multi-omics data integration methods and their applications in psychiatric disorders. Eur Neuropsychopharmacol 2023; 69:26-46. [PMID: 36706689 DOI: 10.1016/j.euroneuro.2023.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/22/2022] [Accepted: 01/02/2023] [Indexed: 01/27/2023]
Abstract
To study mental illness and health, in the past researchers have often broken down their complexity into individual subsystems (e.g., genomics, transcriptomics, proteomics, clinical data) and explored the components independently. Technological advancements and decreasing costs of high throughput sequencing has led to an unprecedented increase in data generation. Furthermore, over the years it has become increasingly clear that these subsystems do not act in isolation but instead interact with each other to drive mental illness and health. Consequently, individual subsystems are now analysed jointly to promote a holistic understanding of the underlying biological complexity of health and disease. Complementing the increasing data availability, current research is geared towards developing novel methods that can efficiently combine the information rich multi-omics data to discover biologically meaningful biomarkers for diagnosis, treatment, and prognosis. However, clinical translation of the research is still challenging. In this review, we summarise conventional and state-of-the-art statistical and machine learning approaches for discovery of biomarker, diagnosis, as well as outcome and treatment response prediction through integrating multi-omics and clinical data. In addition, we describe the role of biological model systems and in silico multi-omics model designs in clinical translation of psychiatric research from bench to bedside. Finally, we discuss the current challenges and explore the application of multi-omics integration in future psychiatric research. The review provides a structured overview and latest updates in the field of multi-omics in psychiatry.
Collapse
Affiliation(s)
- Anita Sathyanarayanan
- Queensland University of Technology, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Kelvin Grove, Queensland 4059, Australia
| | - Tamara T Mueller
- Institute for Artificial Intelligence and Informatics in Medicine, TU Munich, 80333 Munich, Germany
| | - Mohammad Ali Moni
- Artificial Intelligence and Digital Health Data Science, School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Katja Schueler
- Clinic for Psychosomatics, Hospital zum Heiligen Geist, Frankfurt am Main, Germany; Frankfurt Psychoanalytic Institute, Frankfurt am Main, Germany
| | - Bernhard T Baune
- Department of Psychiatry and Psychotherapy, University of Münster, Germany; Department of Psychiatry, Melbourne Medical School, University of Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Australia
| | - Pietro Lio
- Department of Computer Science and Technology, University of Cambridge, Cambridge, United Kingdom
| | - Divya Mehta
- Queensland University of Technology, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Kelvin Grove, Queensland 4059, Australia.
| | | | - Bernhard T Baune
- Department of Psychiatry and Psychotherapy, University of Münster, Germany; Department of Psychiatry, Melbourne Medical School, University of Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Australia
| | - Mara Dierssen
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Bjarke Ebert
- Medical Strategy & Communication, H. Lundbeck A/S, Valby, Denmark
| | - Chiara Fabbri
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy; Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Paolo Fusar-Poli
- Early Psychosis: Intervention and Clinical-detection (EPIC) Lab, Department of Psychosis Studies, King's College London, United Kingdom; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia; Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Oliver D Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Psychiatric Imaging, Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | | | - Pietro Lio
- Department of Computer Science and Technology, University of Cambridge, Cambridge, United Kingdom
| | - Eduard Maron
- Department of Psychiatry, University of Tartu, Tartu, Estonia; Centre for Neuropsychopharmacology, Division of Brain Sciences, Imperial College London, London, United Kingdom; Documental Ltd, Tallin, Estonia; West Tallinn Central Hospital, Tallinn, Estonia
| | - Divya Mehta
- Queensland University of Technology, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Kelvin Grove, Queensland 4059, Australia
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, University of Brescia; Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Lara Nonell
- MARGenomics, IMIM (Hospital del Mar Research Institute), Barcelona, Spain
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | | | - Filip Rybakowski
- Department of Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Alessandro Serretti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - David Stacey
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Roos van Westrhenen
- Parnassia Psychiatric Institute, Amsterdam, the Netherlands; Department of Psychiatry and Neuropsychology, Faculty of Health and Sciences, Maastricht University, Maastricht, the Netherlands; Institute of Psychiatry, Psychology & Neuroscience (IoPPN) King's College London, United Kingdom
| | - Laura Xicota
- Paris Brain Institute ICM, Salpetriere Hospital, Paris, France
| |
Collapse
|
91
|
Lafferty MJ, Aygün N, Patel NK, Krupa O, Liang D, Wolter JM, Geschwind DH, de la Torre-Ubieta L, Stein JL. MicroRNA-eQTLs in the developing human neocortex link miR-4707-3p expression to brain size. eLife 2023; 12:e79488. [PMID: 36629315 PMCID: PMC9859047 DOI: 10.7554/elife.79488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 01/10/2023] [Indexed: 01/12/2023] Open
Abstract
Expression quantitative trait loci (eQTL) data have proven important for linking non-coding loci to protein-coding genes. But eQTL studies rarely measure microRNAs (miRNAs), small non-coding RNAs known to play a role in human brain development and neurogenesis. Here, we performed small-RNA sequencing across 212 mid-gestation human neocortical tissue samples, measured 907 expressed miRNAs, discovering 111 of which were novel, and identified 85 local-miRNA-eQTLs. Colocalization of miRNA-eQTLs with GWAS summary statistics yielded one robust colocalization of miR-4707-3p expression with educational attainment and brain size phenotypes, where the miRNA expression increasing allele was associated with decreased brain size. Exogenous expression of miR-4707-3p in primary human neural progenitor cells decreased expression of predicted targets and increased cell proliferation, indicating miR-4707-3p modulates progenitor gene regulation and cell fate decisions. Integrating miRNA-eQTLs with existing GWAS yielded evidence of a miRNA that may influence human brain size and function via modulation of neocortical brain development.
Collapse
Affiliation(s)
- Michael J Lafferty
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Nil Aygün
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Niyanta K Patel
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Oleh Krupa
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Dan Liang
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Justin M Wolter
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Luis de la Torre-Ubieta
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel HillChapel HillUnited States
| |
Collapse
|
92
|
Gedik H, Peterson RE, Riley BP, Vladimirov VI, Bacanu SA. Integrative Post-Genome-Wide Association Study Analyses Relevant to Psychiatric Disorders: Imputing Transcriptome and Proteome Signals. Complex Psychiatry 2023; 9:130-144. [PMID: 37588130 PMCID: PMC10425719 DOI: 10.1159/000530223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 03/09/2023] [Indexed: 08/18/2023] Open
Abstract
Background The genome-wide association study (GWAS) is a common tool to identify genetic variants associated with complex traits, including psychiatric disorders (PDs). However, post-GWAS analyses are needed to extend the statistical inference to biologically relevant entities, e.g., genes, proteins, and pathways. To achieve this goal, researchers developed methods that incorporate biologically relevant intermediate molecular phenotypes, such as gene expression and protein abundance, which are posited to mediate the variant-trait association. Transcriptome-wide association study (TWAS) and proteome-wide association study (PWAS) are commonly used methods to test the association between these molecular mediators and the trait. Summary In this review, we discuss the most recent developments in TWAS and PWAS. These methods integrate existing "omic" information with the GWAS summary statistics for trait(s) of interest. Specifically, they impute transcript/protein data and test the association between imputed gene expression/protein level with phenotype of interest by using (i) GWAS summary statistics and (ii) reference transcriptomic/proteomic/genomic datasets. TWAS and PWAS are suitable as analysis tools for (i) primary association scan and (ii) fine-mapping to identify potentially causal genes for PDs. Key Messages As post-GWAS analyses, TWAS and PWAS have the potential to highlight causal genes for PDs. These prioritized genes could indicate targets for the development of novel drug therapies. For researchers attempting such analyses, we recommend Mendelian randomization tools that use GWAS statistics for both trait and reference datasets, e.g., summary Mendelian randomization (SMR). We base our recommendation on (i) being able to use the same tool for both TWAS and PWAS, (ii) not requiring the pre-computed weights (and thus easier to update for larger reference datasets), and (iii) most larger transcriptome reference datasets are publicly available and easy to transform into a compatible format for SMR analysis.
Collapse
Affiliation(s)
- Huseyin Gedik
- Integrative Life Sciences, Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Roseann E. Peterson
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Brien P. Riley
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Vladimir I. Vladimirov
- Department of Psychiatry, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Silviu-Alin Bacanu
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
93
|
Levchenko A, Gusev F, Rogaev E. The evolutionary origin of psychosis. Front Psychiatry 2023; 14:1115929. [PMID: 36741116 PMCID: PMC9894884 DOI: 10.3389/fpsyt.2023.1115929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
Imagination, the driving force of creativity, and primary psychosis are human-specific, since we do not observe behaviors in other species that would convincingly suggest they possess the same traits. Both these traits have been linked to the function of the prefrontal cortex, which is the most evolutionarily novel region of the human brain. A number of evolutionarily novel genetic and epigenetic changes that determine the human brain-specific structure and function have been discovered in recent years. Among them are genomic loci subjected to increased rates of single nucleotide substitutions in humans, called human accelerated regions. These mostly regulatory regions are involved in brain development and sometimes contain genetic variants that confer a risk for schizophrenia. On the other hand, neuroimaging data suggest that mind wandering and related phenomena (as a proxy of imagination) are in many ways similar to rapid eye movement dreaming, a function also present in non-human species. Furthermore, both functions are similar to psychosis in several ways: for example, the same brain areas are activated both in dreams and visual hallucinations. In the present Perspective we hypothesize that imagination is an evolutionary adaptation of dreaming, while primary psychosis results from deficient control by higher-order brain areas over imagination. In the light of this, human accelerated regions might be one of the key drivers in evolution of human imagination and the pathogenesis of psychotic disorders.
Collapse
Affiliation(s)
- Anastasia Levchenko
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Fedor Gusev
- Center for Genetics and Life Sciences, Department of Genetics, Sirius University of Science and Technology, Sochi, Russia.,Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Evgeny Rogaev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia.,Department of Psychiatry, UMass Chan Medical School, Shrewsbury, MA, United States
| |
Collapse
|
94
|
Ardesch DJ, Libedinsky I, Scholtens LH, Wei Y, van den Heuvel MP. Convergence of brain transcriptomic and neuroimaging patterns in schizophrenia, bipolar disorder, autism spectrum disorder and major depression disorder. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023. [DOI: 10.1016/j.bpsc.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
95
|
Levchenko A, Plotnikova M. Genomic regulatory sequences in the pathogenesis of bipolar disorder. Front Psychiatry 2023; 14:1115924. [PMID: 36824672 PMCID: PMC9941178 DOI: 10.3389/fpsyt.2023.1115924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
The lifetime prevalence of bipolar disorder is estimated to be about 2%. Epigenetics defines regulatory mechanisms that determine relatively stable patterns of gene expression by controlling all key steps, from DNA to messenger RNA to protein. This Mini Review highlights recent discoveries of modified epigenetic control resulting from genetic variants associated with bipolar disorder in genome-wide association studies. The revealed epigenetic abnormalities implicate gene transcription and post-transcriptional regulation. In the light of these discoveries, the Mini Review focuses on the genes PACS1, MCHR1, DCLK3, HAPLN4, LMAN2L, TMEM258, GNL3, LRRC57, CACNA1C, CACNA1D, and NOVA2 and their potential biological role in the pathogenesis of bipolar disorder. Molecular mechanisms under control of these genes do not translate into a unified picture and substantially more research is needed to fill the gaps in knowledge and to solve current limitations in prognosis and treatment of bipolar disorder. In conclusion, the genetic and functional studies confirm the complex nature of bipolar disorder and indicate future research directions to explore possible targeted treatment options, eventually working toward a personalized approach.
Collapse
Affiliation(s)
- Anastasia Levchenko
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Maria Plotnikova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia.,Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
96
|
Wang Y, Chen L. DeepPerVar: a multi-modal deep learning framework for functional interpretation of genetic variants in personal genome. Bioinformatics 2022; 38:5340-5351. [PMID: 36271868 PMCID: PMC9750124 DOI: 10.1093/bioinformatics/btac696] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/04/2022] [Accepted: 10/20/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Understanding the functional consequence of genetic variants, especially the non-coding ones, is important but particularly challenging. Genome-wide association studies (GWAS) or quantitative trait locus analyses may be subject to limited statistical power and linkage disequilibrium, and thus are less optimal to pinpoint the causal variants. Moreover, most existing machine-learning approaches, which exploit the functional annotations to interpret and prioritize putative causal variants, cannot accommodate the heterogeneity of personal genetic variations and traits in a population study, targeting a specific disease. RESULTS By leveraging paired whole-genome sequencing data and epigenetic functional assays in a population study, we propose a multi-modal deep learning framework to predict genome-wide quantitative epigenetic signals by considering both personal genetic variations and traits. The proposed approach can further evaluate the functional consequence of non-coding variants on an individual level by quantifying the allelic difference of predicted epigenetic signals. By applying the approach to the ROSMAP cohort studying Alzheimer's disease (AD), we demonstrate that the proposed approach can accurately predict quantitative genome-wide epigenetic signals and in key genomic regions of AD causal genes, learn canonical motifs reported to regulate gene expression of AD causal genes, improve the partitioning heritability analysis and prioritize putative causal variants in a GWAS risk locus. Finally, we release the proposed deep learning model as a stand-alone Python toolkit and a web server. AVAILABILITY AND IMPLEMENTATION https://github.com/lichen-lab/DeepPerVar. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ye Wang
- Department of Computer Science and Software Engineering, Auburn University, Auburn, AL 36849, USA
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, FL 32603, USA
| |
Collapse
|
97
|
Krohn L, Heilbron K, Blauwendraat C, Reynolds RH, Yu E, Senkevich K, Rudakou U, Estiar MA, Gustavsson EK, Brolin K, Ruskey JA, Freeman K, Asayesh F, Chia R, Arnulf I, Hu MTM, Montplaisir JY, Gagnon JF, Desautels A, Dauvilliers Y, Gigli GL, Valente M, Janes F, Bernardini A, Högl B, Stefani A, Ibrahim A, Šonka K, Kemlink D, Oertel W, Janzen A, Plazzi G, Biscarini F, Antelmi E, Figorilli M, Puligheddu M, Mollenhauer B, Trenkwalder C, Sixel-Döring F, Cochen De Cock V, Monaca CC, Heidbreder A, Ferini-Strambi L, Dijkstra F, Viaene M, Abril B, Boeve BF, Scholz SW, Ryten M, Bandres-Ciga S, Noyce A, Cannon P, Pihlstrøm L, Nalls MA, Singleton AB, Rouleau GA, Postuma RB, Gan-Or Z. Genome-wide association study of REM sleep behavior disorder identifies polygenic risk and brain expression effects. Nat Commun 2022; 13:7496. [PMID: 36470867 PMCID: PMC9722930 DOI: 10.1038/s41467-022-34732-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 11/03/2022] [Indexed: 12/11/2022] Open
Abstract
Rapid-eye movement (REM) sleep behavior disorder (RBD), enactment of dreams during REM sleep, is an early clinical symptom of alpha-synucleinopathies and defines a more severe subtype. The genetic background of RBD and its underlying mechanisms are not well understood. Here, we perform a genome-wide association study of RBD, identifying five RBD risk loci near SNCA, GBA, TMEM175, INPP5F, and SCARB2. Expression analyses highlight SNCA-AS1 and potentially SCARB2 differential expression in different brain regions in RBD, with SNCA-AS1 further supported by colocalization analyses. Polygenic risk score, pathway analysis, and genetic correlations provide further insights into RBD genetics, highlighting RBD as a unique alpha-synucleinopathy subpopulation that will allow future early intervention.
Collapse
Affiliation(s)
- Lynne Krohn
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | | | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Regina H Reynolds
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Konstantin Senkevich
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Uladzislau Rudakou
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Mehrdad A Estiar
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Emil K Gustavsson
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Kajsa Brolin
- Lund University, Translational Neurogenetics Unit, Department of Experimental Medical Science, Lund, Sweden
| | - Jennifer A Ruskey
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Kathryn Freeman
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Farnaz Asayesh
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Ruth Chia
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Arnulf
- Sleep Disorders Unit, Pitié Salpêtrière Hospital, APHP-Sorbonne, Paris Brain Insitute and Sorbonne University, Paris, France
| | - Michele T M Hu
- Oxford Parkinson's Disease Centre (OPDC), University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jacques Y Montplaisir
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
- Department of Psychiatry, Université de Montréal, Montréal, QC, Canada
| | - Jean-François Gagnon
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
- Department of Psychology, Université du Québec à Montréal, Montreal, QC, Canada
| | - Alex Desautels
- Centre d'Études Avancées en Médecine du Sommeil, Hôpital du Sacré-Cœur de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Yves Dauvilliers
- National Reference Center for Narcolepsy, Sleep Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, University of Montpellier, Institute Neuroscience Montpellier Inserm, Montpellier, France
| | - Gian Luigi Gigli
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
| | - Mariarosaria Valente
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Francesco Janes
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
| | - Andrea Bernardini
- Clinical Neurology Unit, Department of Neurosciences, University Hospital of Udine, Udine, Italy
| | - Birgit Högl
- Sleep Disorders Clinic, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Sleep Disorders Clinic, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Abubaker Ibrahim
- Sleep Disorders Clinic, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Karel Šonka
- Department of Neurology and Centre of Clinical Neuroscience, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - David Kemlink
- Department of Neurology and Centre of Clinical Neuroscience, Charles University, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Wolfgang Oertel
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Annette Janzen
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Giuseppe Plazzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio-Emilia, Modena, Italy
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - Francesco Biscarini
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Elena Antelmi
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Michela Figorilli
- Department of Medical Sciences and Public Health, Sleep Disorder Research Center, University of Cagliari, Cagliari, Italy
| | - Monica Puligheddu
- Department of Medical Sciences and Public Health, Sleep Disorder Research Center, University of Cagliari, Cagliari, Italy
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| | - Friederike Sixel-Döring
- Department of Neurology, Philipps-University, Marburg, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
| | - Valérie Cochen De Cock
- Sleep and Neurology Unit, Beau Soleil Clinic, Montpellier, France
- EuroMov Digital Health in Motion, University of Montpellier IMT Mines Ales, Montpellier, France
| | - Christelle Charley Monaca
- University Lille North of France, Department of Clinical Neurophysiology and Sleep Center, CHU Lille, Lille, France
| | - Anna Heidbreder
- Institute of Sleep Medicine and Neuromuscular Disorders, University of Münster, Münster, Germany
| | - Luigi Ferini-Strambi
- Department of Neurological Sciences, Università Vita-Salute San Raffaele, Milan, Italy
| | - Femke Dijkstra
- Laboratory for Sleep Disorders, St. Dimpna Regional Hospital, Geel, Belgium
- Department of Neurology, St. Dimpna Regional Hospital, Geel, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Mineke Viaene
- Laboratory for Sleep Disorders, St. Dimpna Regional Hospital, Geel, Belgium
- Department of Neurology, St. Dimpna Regional Hospital, Geel, Belgium
| | - Beatriz Abril
- Sleep disorder Unit, Carémeau Hospital, University Hospital of Nîmes, Nîmes, France
| | | | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Mina Ryten
- Great Ormond Street Institute of Child Health, Genetics and Genomic Medicine, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Alastair Noyce
- Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
- Department of Clinical and Movement Neurosciences, University College London, Institute of Neurology, London, UK
| | | | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Mike A Nalls
- Data Tecnica International, Glen Echo, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Guy A Rouleau
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Ronald B Postuma
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada.
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
98
|
Liu S, Won H, Clarke D, Matoba N, Khullar S, Mu Y, Wang D, Gerstein M. Illuminating links between cis-regulators and trans-acting variants in the human prefrontal cortex. Genome Med 2022; 14:133. [PMID: 36424644 PMCID: PMC9685876 DOI: 10.1186/s13073-022-01133-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/25/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Neuropsychiatric disorders afflict a large portion of the global population and constitute a significant source of disability worldwide. Although Genome-wide Association Studies (GWAS) have identified many disorder-associated variants, the underlying regulatory mechanisms linking them to disorders remain elusive, especially those involving distant genomic elements. Expression quantitative trait loci (eQTLs) constitute a powerful means of providing this missing link. However, most eQTL studies in human brains have focused exclusively on cis-eQTLs, which link variants to nearby genes (i.e., those within 1 Mb of a variant). A complete understanding of disease etiology requires a clearer understanding of trans-regulatory mechanisms, which, in turn, entails a detailed analysis of the relationships between variants and expression changes in distant genes. METHODS By leveraging large datasets from the PsychENCODE consortium, we conducted a genome-wide survey of trans-eQTLs in the human dorsolateral prefrontal cortex. We also performed colocalization and mediation analyses to identify mediators in trans-regulation and use trans-eQTLs to link GWAS loci to schizophrenia risk genes. RESULTS We identified ~80,000 candidate trans-eQTLs (at FDR<0.25) that influence the expression of ~10K target genes (i.e., "trans-eGenes"). We found that many variants associated with these candidate trans-eQTLs overlap with known cis-eQTLs. Moreover, for >60% of these variants (by colocalization), the cis-eQTL's target gene acts as a mediator for the trans-eQTL SNP's effect on the trans-eGene, highlighting examples of cis-mediation as essential for trans-regulation. Furthermore, many of these colocalized variants fall into a discernable pattern wherein cis-eQTL's target is a transcription factor or RNA-binding protein, which, in turn, targets the gene associated with the candidate trans-eQTL. Finally, we show that trans-regulatory mechanisms provide valuable insights into psychiatric disorders: beyond what had been possible using only cis-eQTLs, we link an additional 23 GWAS loci and 90 risk genes (using colocalization between candidate trans-eQTLs and schizophrenia GWAS loci). CONCLUSIONS We demonstrate that the transcriptional architecture of the human brain is orchestrated by both cis- and trans-regulatory variants and found that trans-eQTLs provide insights into brain-disease biology.
Collapse
Affiliation(s)
- Shuang Liu
- Waisman Center, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Hyejung Won
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Declan Clarke
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Saniya Khullar
- Waisman Center, University of Wisconsin - Madison, Madison, WI, 53705, USA.,Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, 53706, USA
| | - Yudi Mu
- Department of Statistics, University of Wisconsin - Madison, Madison, WI, 53706, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin - Madison, Madison, WI, 53705, USA. .,Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, 53706, USA. .,Department of Computer Sciences, University of Wisconsin - Madison, Madison, WI, 53706, USA.
| | - Mark Gerstein
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA. .,Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, 06520, USA. .,Department of Computer Science, Yale University, New Haven, CT, 06520, USA. .,Department of Statistics and Data Science, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
99
|
Fan P, Kofler J, Ding Y, Marks M, Sweet RA, Wang L. Efficacy difference of antipsychotics in Alzheimer's disease and schizophrenia: explained with network efficiency and pathway analysis methods. Brief Bioinform 2022; 23:bbac394. [PMID: 36151774 PMCID: PMC9677501 DOI: 10.1093/bib/bbac394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/14/2022] Open
Abstract
Approximately 50% of Alzheimer's disease (AD) patients will develop psychotic symptoms and these patients will experience severe rapid cognitive decline compared with those without psychosis (AD-P). Currently, no medication has been approved by the Food and Drug Administration for AD with psychosis (AD+P) specifically, although atypical antipsychotics are widely used in clinical practice. These drugs have demonstrated modest efficacy in managing psychosis in individuals with AD, with an increased frequency of adverse events, including excess mortality. We compared the differences between the genetic variations/genes associated with AD+P and schizophrenia from existing Genome-Wide Association Study and differentially expressed genes (DEGs). We also constructed disease-specific protein-protein interaction networks for AD+P and schizophrenia. Network efficiency was then calculated to characterize the topological structures of these two networks. The efficiency of antipsychotics in these two networks was calculated. A weight adjustment based on binding affinity to drug targets was later applied to refine our results, and 2013 and 2123 genes were identified as related to AD+P and schizophrenia, respectively, with only 115 genes shared. Antipsychotics showed a significantly lower efficiency in the AD+P network than in the schizophrenia network (P < 0.001) indicating that antipsychotics may have less impact in AD+P than in schizophrenia. AD+P may be caused by mechanisms distinct from those in schizophrenia which result in a decreased efficacy of antipsychotics in AD+P. In addition, the network analysis methods provided quantitative explanations of the lower efficacy of antipsychotics in AD+P.
Collapse
Affiliation(s)
- Peihao Fan
- School of Pharmacy, University of Pittsburgh
| | | | - Ying Ding
- Department of Biostatistics at the University of Pittsburgh
| | - Michael Marks
- Center for Neuroscience at the University of Pittsburgh and the Department of Neurobiology
| | - Robert A Sweet
- UPMC Endowed Professor of Psychiatric Neuroscience and Professor of Neurology at the University of Pittsburgh
| | - Lirong Wang
- department of pharmaceutical sciences, school of pharmacy at University of Pittsburgh, USA
| |
Collapse
|
100
|
Real R, Martinez-Carrasco A, Reynolds RH, Lawton MA, Tan MMX, Shoai M, Corvol JC, Ryten M, Bresner C, Hubbard L, Brice A, Lesage S, Faouzi J, Elbaz A, Artaud F, Williams N, Hu MTM, Ben-Shlomo Y, Grosset DG, Hardy J, Morris HR. Association between the LRP1B and APOE loci in the development of Parkinson's disease dementia. Brain 2022; 146:1873-1887. [PMID: 36348503 PMCID: PMC10151192 DOI: 10.1093/brain/awac414] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/04/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022] Open
Abstract
Parkinson's disease is one of the most common age-related neurodegenerative disorders. Although predominantly a motor disorder, cognitive impairment and dementia are important features of Parkinson's disease, particularly in the later stages of the disease. However, the rate of cognitive decline varies among Parkinson's disease patients, and the genetic basis for this heterogeneity is incompletely understood. To explore the genetic factors associated with rate of progression to Parkinson's disease dementia, we performed a genome-wide survival meta-analysis of 3,923 clinically diagnosed Parkinson's disease cases of European ancestry from four longitudinal cohorts. In total, 6.7% of individuals with Parkinson's disease developed dementia during study follow-up, on average 4.4 ± 2.4 years from disease diagnosis. We have identified the APOE ε4 allele as a major risk factor for the conversion to Parkinson's disease dementia [hazards ratio = 2.41 (1.94-3.00), P = 2.32 × 10-15], as well as a new locus within the ApoE and APP receptor LRP1B gene [hazards ratio = 3.23 (2.17-4.81), P = 7.07 × 10-09]. In a candidate gene analysis, GBA variants were also identified to be associated with higher risk of progression to dementia [hazards ratio = 2.02 (1.21-3.32), P = 0.007]. CSF biomarker analysis also implicated the amyloid pathway in Parkinson's disease dementia, with significantly reduced levels of amyloid β42 (P = 0.0012) in Parkinson's disease dementia compared to Parkinson's disease without dementia. These results identify a new candidate gene associated with faster conversion to dementia in Parkinson's disease and suggest that amyloid-targeting therapy may have a role in preventing Parkinson's disease dementia.
Collapse
Affiliation(s)
- Raquel Real
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UCL Movement Disorders Centre, University College London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Alejandro Martinez-Carrasco
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UCL Movement Disorders Centre, University College London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Regina H Reynolds
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Michael A Lawton
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Manuela M X Tan
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | - Maryam Shoai
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Jean-Christophe Corvol
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière - Paris Brain Institute - ICM, INSERM, CNRS, 75013 Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Mina Ryten
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Catherine Bresner
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff CF24 4HQ, UK
| | - Leon Hubbard
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff CF24 4HQ, UK
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière - Paris Brain Institute - ICM, INSERM, CNRS, 75013 Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Suzanne Lesage
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière - Paris Brain Institute - ICM, INSERM, CNRS, 75013 Paris, France
- Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Johann Faouzi
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière - Paris Brain Institute - ICM, INSERM, CNRS, 75013 Paris, France
- Centre Inria de Paris, 75012 Paris, France
| | - Alexis Elbaz
- Centre for Research in Epidemiology and Population Health, INSERM U1018, Team "Exposome, heredity, cancer, and health", 94807 Villejuif, France
| | - Fanny Artaud
- Centre for Research in Epidemiology and Population Health, INSERM U1018, Team "Exposome, heredity, cancer, and health", 94807 Villejuif, France
| | - Nigel Williams
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michele T M Hu
- Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford OX3 9DU, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford OX1 3QU, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK
| | - Donald G Grosset
- School of Neuroscience and Psychology, University of Glasgow, Glasgow G51 4TF, UK
| | - John Hardy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- National Institute for Health Research (NIHR) University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UCL Movement Disorders Centre, University College London, London WC1N 3BG, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|