1
|
Wang R, Su Y, O'Donnell K, Caron J, Meaney M, Meng X, Li Y. Differential interactions between gene expressions and stressors across the lifespan in major depressive disorder. J Affect Disord 2024; 362:688-697. [PMID: 39029669 DOI: 10.1016/j.jad.2024.07.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/05/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Both genetic predispositions and exposures to stressors have collectively contributed to the development of major depressive disorder (MDD). To deep dive into their roles in MDD, our study aimed to examine which susceptible gene expression interacts with various dimensions of stressors in the MDD risk among a large population cohort. METHODS Data analyzed were from a longitudinal community-based cohort from Southwest Montreal, Canada (N = 1083). Latent profile models were used to identify distinct patterns of stressors for the study cohort. A transcriptome-wide association study (TWAS) method was performed to examine the interactive effects of three dimensions of stressors (threat, deprivation, and cumulative lifetime stress) and gene expression on the MDD risk in a total of 48 tissues from GTEx. Additional analyses were also conducted to further explore and specify these associations including colocalization, and fine-mapping analyses, in addition to enrichment analysis investigations based on TWAS. RESULTS We identified 3321 genes linked to MDD at the nominal p-value <0.05 and found that different patterns of stressors can amplify the genetic susceptibility to MDD. We also observed specific genes and pathways that interacted with deprivation and cumulative lifetime stressors, particularly in specific brain tissues including basal ganglia, prefrontal cortex, brain amygdala, brain cerebellum, brain cortex, and the whole blood. Colocalization analysis also identified these genes as having a high probability of sharing MDD causal variants. LIMITATIONS The study cohort was composed exclusively of individuals of Caucasians, which restricts the generalizability of the findings to other ethnic population groups. CONCLUSIONS The findings of the study unveiled significant interactions between potential tissue-specific gene expression × stressors in the MDD risk and shed light on the intricate etiological attributes of gene expression and specific stressors across the lifespan in MDD. These genetic and environmental attributes in MDD corroborate the vulnerability-stress theory and direct future stress research to have a closer examination of genetic predisposition and potential involvements of omics studies to specify the intricate relationships between genes and stressful environments.
Collapse
Affiliation(s)
- Ruiyang Wang
- Department of Financial and Risk Engineering, New York University, NY, NYC, USA; Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, Montreal, QC, Canada
| | - Yingying Su
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Kieran O'Donnell
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, Montreal, QC, Canada; Yale Child Study Center, Department of Obstetrics Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, CT, USA; Child & Brain Development Program, CIFAR, Toronto, ON, Canada
| | - Jean Caron
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, Montreal, QC, Canada
| | - Michael Meaney
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, Montreal, QC, Canada
| | - Xiangfei Meng
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Douglas Research Centre, Montreal, QC, Canada.
| | - Yue Li
- School of Computer Science, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Watanabe H, Kobikov Y, Nosova O, Sarkisyan D, Galatenko V, Carvalho L, Maia GH, Lukoyanov N, Lavrov I, Ossipov MH, Hallberg M, Schouenborg J, Zhang M, Bakalkin G. The Left-Right Side-Specific Neuroendocrine Signaling from Injured Brain: An Organizational Principle. FUNCTION 2024; 5:zqae013. [PMID: 38985004 PMCID: PMC11237900 DOI: 10.1093/function/zqae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 07/11/2024] Open
Abstract
A neurological dogma is that the contralateral effects of brain injury are set through crossed descending neural tracts. We have recently identified a novel topographic neuroendocrine system (T-NES) that operates via a humoral pathway and mediates the left-right side-specific effects of unilateral brain lesions. In rats with completely transected thoracic spinal cords, unilateral injury to the sensorimotor cortex produced contralateral hindlimb flexion, a proxy for neurological deficit. Here, we investigated in acute experiments whether T-NES consists of left and right counterparts and whether they differ in neural and molecular mechanisms. We demonstrated that left- and right-sided hormonal signaling is differentially blocked by the δ-, κ- and µ-opioid antagonists. Left and right neurohormonal signaling differed in targeting the afferent spinal mechanisms. Bilateral deafferentation of the lumbar spinal cord abolished the hormone-mediated effects of the left-brain injury but not the right-sided lesion. The sympathetic nervous system was ruled out as a brain-to-spinal cord-signaling pathway since hindlimb responses were induced in rats with cervical spinal cord transections that were rostral to the preganglionic sympathetic neurons. Analysis of gene-gene co-expression patterns identified the left- and right-side-specific gene co-expression networks that were coordinated via the humoral pathway across the hypothalamus and lumbar spinal cord. The coordination was ipsilateral and disrupted by brain injury. These findings suggest that T-NES is bipartite and that its left and right counterparts contribute to contralateral neurological deficits through distinct neural mechanisms, and may enable ipsilateral regulation of molecular and neural processes across distant neural areas along the neuraxis.
Collapse
Affiliation(s)
- Hiroyuki Watanabe
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, SE-751 24, Sweden
- Department of Molecular Medicine, University of Southern Denmark, Odense, DK-5230, Denmark
| | - Yaromir Kobikov
- Volunteer Associate at Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, SE-751 24, Sweden
| | - Olga Nosova
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, SE-751 24, Sweden
| | - Daniil Sarkisyan
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, SE-751 24, Sweden
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, SE-751 08, Sweden
| | | | - Liliana Carvalho
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Porto 4200-319, Portugal
| | - Gisela H Maia
- Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Porto 4200-072, Portugal
- Medibrain, Vila do Conde 4480-807, Portugal
- Brain Research Institute, Porto 4450-208, Portugal
| | - Nikolay Lukoyanov
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Porto 4200-319, Portugal
- Brain Research Institute, Porto 4450-208, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4200-135, Portugal
| | - Igor Lavrov
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael H Ossipov
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724-5050, USA
| | - Mathias Hallberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, SE-751 24, Sweden
| | - Jens Schouenborg
- Neuronano Research Center, Department of Experimental Medical Science, Lund University, Lund 223 63, Sweden
| | - Mengliang Zhang
- Department of Molecular Medicine, University of Southern Denmark, Odense, DK-5230, Denmark
- Neuronano Research Center, Department of Experimental Medical Science, Lund University, Lund 223 63, Sweden
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, SE-751 24, Sweden
| |
Collapse
|
3
|
Morgunova A, Teixeira M, Flores C. Perspective on adolescent psychiatric illness and emerging role of microRNAs as biomarkers of risk. J Psychiatry Neurosci 2024; 49:E282-E288. [PMID: 39209460 PMCID: PMC11374446 DOI: 10.1503/jpn.240072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Affiliation(s)
- Alice Morgunova
- From the Douglas Mental Health University Institute, Montreal, Que. (Morgunova, Flores); the Department of Psychiatry, McGill University, Montreal, Que. (Morgunova, Flores); the Integrated Program in Neuroscience, McGill University, Montreal, Que. (Teixeira); the Department of Neurology and Neurosurgery, McGill University, Montreal, Que. (Flores); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, Que. (Flores)
| | - Maxime Teixeira
- From the Douglas Mental Health University Institute, Montreal, Que. (Morgunova, Flores); the Department of Psychiatry, McGill University, Montreal, Que. (Morgunova, Flores); the Integrated Program in Neuroscience, McGill University, Montreal, Que. (Teixeira); the Department of Neurology and Neurosurgery, McGill University, Montreal, Que. (Flores); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, Que. (Flores)
| | - Cecilia Flores
- From the Douglas Mental Health University Institute, Montreal, Que. (Morgunova, Flores); the Department of Psychiatry, McGill University, Montreal, Que. (Morgunova, Flores); the Integrated Program in Neuroscience, McGill University, Montreal, Que. (Teixeira); the Department of Neurology and Neurosurgery, McGill University, Montreal, Que. (Flores); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montreal, Que. (Flores)
| |
Collapse
|
4
|
Sun R, Tang MY, Yang D, Zhang YY, Xu YH, Qiao Y, Yu B, Cao SX, Wang H, Huang HQ, Zhang H, Li XM, Lian H. C3aR in the medial prefrontal cortex modulates the susceptibility to LPS-induced depressive-like behaviors through glutamatergic neuronal excitability. Prog Neurobiol 2024; 236:102614. [PMID: 38641040 DOI: 10.1016/j.pneurobio.2024.102614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/18/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Complement activation and prefrontal cortical dysfunction both contribute to the pathogenesis of major depressive disorder (MDD), but their interplay in MDD is unclear. We here studied the role of complement C3a receptor (C3aR) in the medial prefrontal cortex (mPFC) and its influence on depressive-like behaviors induced by systematic lipopolysaccharides (LPS) administration. C3aR knockout (KO) or intra-mPFC C3aR antagonism confers resilience, whereas C3aR expression in mPFC neurons makes KO mice susceptible to LPS-induced depressive-like behaviors. Importantly, the excitation and inhibition of mPFC neurons have opposing effects on depressive-like behaviors, aligning with increased and decreased excitability by C3aR deletion and activation in cortical neurons. In particular, inhibiting mPFC glutamatergic (mPFCGlu) neurons, the main neuronal subpopulation expresses C3aR, induces depressive-like behaviors in saline-treated WT and KO mice, but not in LPS-treated KO mice. Compared to hypoexcitable mPFCGlu neurons in LPS-treated WT mice, C3aR-null mPFCGlu neurons display hyperexcitability upon LPS treatment, and enhanced excitation of mPFCGlu neurons is anti-depressant, suggesting a protective role of C3aR deficiency in these circumstances. In conclusion, C3aR modulates susceptibility to LPS-induced depressive-like behaviors through mPFCGlu neuronal excitability. This study identifies C3aR as a pivotal intersection of complement activation, mPFC dysfunction, and depression and a promising therapeutic target for MDD.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Research Center of System Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China; Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Meng-Yu Tang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Dan Yang
- Clinical Research Center, The second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan-Yi Zhang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yi-Heng Xu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yong Qiao
- Department of Neurology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Research Center of System Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Yu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shu-Xia Cao
- Department of Neurology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wang
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui-Qian Huang
- Clinical Research Center, The second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Zhang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Ming Li
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Hong Lian
- Department of Neurology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Research Center of System Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
5
|
Strom NI, Halvorsen MW, Tian C, Rück C, Kvale G, Hansen B, Bybjerg-Grauholm J, Grove J, Boberg J, Nissen JB, Damm Als T, Werge T, de Schipper E, Fundin B, Hultman C, Höffler KD, Pedersen N, Sandin S, Bulik C, Landén M, Karlsson E, Hagen K, Lindblad-Toh K, Hougaard DM, Meier SM, Hellard SL, Mors O, Børglum AD, Haavik J, Hinds DA, Mataix-Cols D, Crowley JJ, Mattheisen M. Genome-wide association study identifies new loci associated with OCD. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.06.24303776. [PMID: 38496634 PMCID: PMC10942538 DOI: 10.1101/2024.03.06.24303776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
To date, four genome-wide association studies (GWAS) of obsessive-compulsive disorder (OCD) have been published, reporting a high single-nucleotide polymorphism (SNP)-heritability of 28% but finding only one significant SNP. A substantial increase in sample size will likely lead to further identification of SNPs, genes, and biological pathways mediating the susceptibility to OCD. We conducted a GWAS meta-analysis with a 2-3-fold increase in case sample size (OCD cases: N = 37,015, controls: N = 948,616) compared to the last OCD GWAS, including six previously published cohorts (OCGAS, IOCDF-GC, IOCDF-GC-trio, NORDiC-nor, NORDiC-swe, and iPSYCH) and unpublished self-report data from 23andMe Inc. We explored the genetic architecture of OCD by conducting gene-based tests, tissue and celltype enrichment analyses, and estimating heritability and genetic correlations with 74 phenotypes. To examine a potential heterogeneity in our data, we conducted multivariable GWASs with MTAG. We found support for 15 independent genome-wide significant loci (14 new) and 79 protein-coding genes. Tissue enrichment analyses implicate multiple cortical regions, the amygdala, and hypothalamus, while cell type analyses yielded 12 cell types linked to OCD (all neurons). The SNP-based heritability of OCD was estimated to be 0.08. Using MTAG we found evidence for specific genetic underpinnings characteristic of different cohort-ascertainment and identified additional significant SNPs. OCD was genetically correlated with 40 disorders or traits-positively with all psychiatric disorders and negatively with BMI, age at first birth and multiple autoimmune diseases. The GWAS meta-analysis identified several biologically informative genes as important contributors to the aetiology of OCD. Overall, we have begun laying the groundwork through which the biology of OCD will be understood and described.
Collapse
Affiliation(s)
- Nora I Strom
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital of Munich, Munich, Germany
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Matthew W Halvorsen
- Department of Genetics, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
| | | | - Christian Rück
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Gerd Kvale
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Psychology, University of Bergen, Bergen, Norway
| | - Bjarne Hansen
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Psychology, University of Bergen, Bergen, Norway
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Jakob Grove
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Julia Boberg
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Judith Becker Nissen
- Departments of Child and Adolescent Psychiatry, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
- Institute of Clinical Medicine, Health, Aarhus University, Health, Aarhus University, Aarhus, Danmark
| | - Thomas Damm Als
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Institute of Biological Psychiatry, Mental Health Services Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
- GLOBE Institute, Center for GeoGenetics, University of Copenhagen, Copenhagen, Denmark
| | - Elles de Schipper
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Fundin
- Department of Medical Epidemiology and Biostatistics, Center for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden
| | - Christina Hultman
- Department of Medical Epidemiology and Biostatistics, Center for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden
| | - Kira D. Höffler
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Nancy Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Cynthia Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, NC, USA
| | - Mikael Landén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Elinor Karlsson
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kristen Hagen
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Psychiatry, Møre og Romsdal Hospital Trust, Molde, Møre og Romsdal, Norway
- Department of Mental Health, Norwegian University for Science and Technology, Trondheim, Sweden
| | - Kerstin Lindblad-Toh
- Department of Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | - David M. Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Sandra M. Meier
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Stéphanie Le Hellard
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole Mors
- Psychosis Research Unit, Aarhus University Hospital - Psychiatry, Aarhus Denmark
| | - Anders D. Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Jan Haavik
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - David Mataix-Cols
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - James J Crowley
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Department of Genetics, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina At Chapel Hill, Chapel Hill, NC, USA
| | - Manuel Mattheisen
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital of Munich, Munich, Germany
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Community Health and Epidemiology and Faculty of Computer Science, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
6
|
Zhang P, Zhang W, Sun W, Xu J, Hu H, Wang L, Wong L. Identification of gene biomarkers for brain diseases via multi-network topological semantics extraction and graph convolutional network. BMC Genomics 2024; 25:175. [PMID: 38350848 PMCID: PMC10865627 DOI: 10.1186/s12864-024-09967-9] [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: 09/06/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Brain diseases pose a significant threat to human health, and various network-based methods have been proposed for identifying gene biomarkers associated with these diseases. However, the brain is a complex system, and extracting topological semantics from different brain networks is necessary yet challenging to identify pathogenic genes for brain diseases. RESULTS In this study, we present a multi-network representation learning framework called M-GBBD for the identification of gene biomarker in brain diseases. Specifically, we collected multi-omics data to construct eleven networks from different perspectives. M-GBBD extracts the spatial distributions of features from these networks and iteratively optimizes them using Kullback-Leibler divergence to fuse the networks into a common semantic space that represents the gene network for the brain. Subsequently, a graph consisting of both gene and large-scale disease proximity networks learns representations through graph convolution techniques and predicts whether a gene is associated which brain diseases while providing associated scores. Experimental results demonstrate that M-GBBD outperforms several baseline methods. Furthermore, our analysis supported by bioinformatics revealed CAMP as a significantly associated gene with Alzheimer's disease identified by M-GBBD. CONCLUSION Collectively, M-GBBD provides valuable insights into identifying gene biomarkers for brain diseases and serves as a promising framework for brain networks representation learning.
Collapse
Affiliation(s)
- Ping Zhang
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, 277100, Shandong, China
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Weihan Zhang
- CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430074, China
| | - Weicheng Sun
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jinsheng Xu
- College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hua Hu
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, 277100, Shandong, China.
| | - Lei Wang
- College of Information Science and Engineering, Zaozhuang University, Zaozhuang, 277100, Shandong, China.
- Guangxi Key Lab of Human-Machine Interaction and Intelligent Decision, Guangxi Academy of Sciences, Nanning, 530007, China.
| | - Leon Wong
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, 518118, China.
| |
Collapse
|
7
|
Meng X, Navoly G, Giannakopoulou O, Levey DF, Koller D, Pathak GA, Koen N, Lin K, Adams MJ, Rentería ME, Feng Y, Gaziano JM, Stein DJ, Zar HJ, Campbell ML, van Heel DA, Trivedi B, Finer S, McQuillin A, Bass N, Chundru VK, Martin HC, Huang QQ, Valkovskaya M, Chu CY, Kanjira S, Kuo PH, Chen HC, Tsai SJ, Liu YL, Kendler KS, Peterson RE, Cai N, Fang Y, Sen S, Scott LJ, Burmeister M, Loos RJF, Preuss MH, Actkins KV, Davis LK, Uddin M, Wani AH, Wildman DE, Aiello AE, Ursano RJ, Kessler RC, Kanai M, Okada Y, Sakaue S, Rabinowitz JA, Maher BS, Uhl G, Eaton W, Cruz-Fuentes CS, Martinez-Levy GA, Campos AI, Millwood IY, Chen Z, Li L, Wassertheil-Smoller S, Jiang Y, Tian C, Martin NG, Mitchell BL, Byrne EM, Awasthi S, Coleman JRI, Ripke S, Sofer T, Walters RG, McIntosh AM, Polimanti R, Dunn EC, Stein MB, Gelernter J, Lewis CM, Kuchenbaecker K. Multi-ancestry genome-wide association study of major depression aids locus discovery, fine mapping, gene prioritization and causal inference. Nat Genet 2024; 56:222-233. [PMID: 38177345 PMCID: PMC10864182 DOI: 10.1038/s41588-023-01596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/26/2023] [Indexed: 01/06/2024]
Abstract
Most genome-wide association studies (GWAS) of major depression (MD) have been conducted in samples of European ancestry. Here we report a multi-ancestry GWAS of MD, adding data from 21 cohorts with 88,316 MD cases and 902,757 controls to previously reported data. This analysis used a range of measures to define MD and included samples of African (36% of effective sample size), East Asian (26%) and South Asian (6%) ancestry and Hispanic/Latin American participants (32%). The multi-ancestry GWAS identified 53 significantly associated novel loci. For loci from GWAS in European ancestry samples, fewer than expected were transferable to other ancestry groups. Fine mapping benefited from additional sample diversity. A transcriptome-wide association study identified 205 significantly associated novel genes. These findings suggest that, for MD, increasing ancestral and global diversity in genetic studies may be particularly important to ensure discovery of core genes and inform about transferability of findings.
Collapse
Affiliation(s)
| | | | | | - Daniel F Levey
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Dora Koller
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Gita A Pathak
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Nastassja Koen
- SAMRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Miguel E Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - J Michael Gaziano
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Dan J Stein
- SAMRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Heather J Zar
- SAMRC Unit on Child and Adolescent Health, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Megan L Campbell
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | | | - Bhavi Trivedi
- Blizard Institute, Queen Mary University of London, London, UK
| | - Sarah Finer
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Nick Bass
- Division of Psychiatry, UCL, London, UK
| | | | | | | | | | | | - Susan Kanjira
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Po-Hsiu Kuo
- Department of Public Health and Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsi-Chung Chen
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
- Center of Sleep Disorders, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Jen Tsai
- Institute of Brain Science and Division of Psychiatry, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | | | - Roseann E Peterson
- Department of Psychiatry, VCU, Richmond, VA, USA
- Department of Psychiatry, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, Neuherberg, Germany
- Computational Health Centre, Helmholtz Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Yu Fang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Srijan Sen
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Laura J Scott
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Margit Burmeister
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael H Preuss
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ky'Era V Actkins
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lea K Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Monica Uddin
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Agaz H Wani
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Derek E Wildman
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Allison E Aiello
- Robert N. Butler Columbia Aging Center, Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Robert J Ursano
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | - Masahiro Kanai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Saori Sakaue
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jill A Rabinowitz
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - George Uhl
- Neurology and Pharmacology, University of Maryland, Maryland VA Healthcare System, Baltimore, MD, USA
| | - William Eaton
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Carlos S Cruz-Fuentes
- Departamento de Genética, Instituto Nacional de Psiquiatría 'Ramón de la Fuente Muñíz', Mexico City, Mexico
| | - Gabriela A Martinez-Levy
- Departamento de Genética, Instituto Nacional de Psiquiatría 'Ramón de la Fuente Muñíz', Mexico City, Mexico
| | - Adrian I Campos
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Iona Y Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | | | - Yunxuan Jiang
- Department of Biostatistics, Emory University, Atlanta, GA, USA
- 23andMe, Inc., Mountain View, CA, USA
| | - Chao Tian
- 23andMe, Inc., Mountain View, CA, USA
| | - Nicholas G Martin
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Brittany L Mitchell
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Enda M Byrne
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Swapnil Awasthi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Jonathan R I Coleman
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Stephan Ripke
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Cambridge, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
- Institute for Genomics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Renato Polimanti
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Erin C Dunn
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Murray B Stein
- Department of Psychiatry, UC San Diego School of Medicine, La Jolla, CA, USA
- Herbert Wertheim School of Public Health and Human Longevity, University of California San Diego, La Jolla, CA, USA
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Joel Gelernter
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Cathryn M Lewis
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | | |
Collapse
|
8
|
Liang J, LaFleur B, Hussainy S, Perry G. Gene Co-Expression Analysis of Multiple Brain Tissues Reveals Correlation of FAM222A Expression with Multiple Alzheimer's Disease-Related Genes. J Alzheimers Dis 2024; 99:S249-S263. [PMID: 37092222 PMCID: PMC11091573 DOI: 10.3233/jad-221241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/25/2023]
Abstract
Background Alzheimer's disease (AD) is the most common form of dementia in the elderly marked by central nervous system (CNS) neuronal loss and amyloid plaques. FAM222A, encoding an amyloid plaque core protein, is an AD brain atrophy susceptibility gene that mediates amyloid-β aggregation. However, the expression interplay between FAM222A and other AD-related pathway genes is unclear. Objective Our goal was to study FAM222A's whole-genome co-expression profile in multiple tissues and investigate its interplay with other AD-related genes. Methods We analyzed gene expression correlations in Genotype-Tissue Expression (GTEx) tissues to identify FAM222A co-expressed genes and performed functional enrichment analysis on identified genes in CNS system. Results Genome-wide gene expression profiling identified 673 genes significantly correlated with FAM222A (p < 2.5×10-6) in 48 human tissues, including 298 from 13 CNS tissues. Functional enrichment analysis revealed that FAM222A co-expressed CNS genes were enriched in multiple AD-related pathways. Gene co-expression network analysis for identified genes in each brain region predicted other disease associated genes with similar biological function. Furthermore, co-expression of 25 out of 31 AD-related pathways genes with FAM222A was replicated in brain samples from 107 aged subjects from the Aging, Dementia and TBI Study. Conclusion This gene co-expression study identified multiple AD-related genes that are associated with FAM222A, indicating that FAM222A and AD-associated genes can be active simultaneously in similar biological processes, providing evidence that supports the association of FAM222A with AD.
Collapse
Affiliation(s)
- Jingjing Liang
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Bonnie LaFleur
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Sadiya Hussainy
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
9
|
Zang JCS, Hohoff C, Van Assche E, Lange P, Kraft M, Sandmann S, Varghese J, Jörgens S, Knight MJ, Baune BT. Immune gene co-expression signatures implicated in occurence and persistence of cognitive dysfunction in depression. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110826. [PMID: 37451594 DOI: 10.1016/j.pnpbp.2023.110826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/29/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
Cognitive dysfunction contributes significantly to the burden caused by Major Depressive Disorder (MDD). Yet, while compelling evidence suggests that different biological processes play a part in both MDD aetiology and the development of cognitive decline more generally, we only begin to understand the molecular underpinnings of depression-related cognitive impairment. Developments in psychometric assessments, molecular high-throughput methods and systems biology derived analysis strategies advance this endeavour. Here, we aim to identify gene expression signatures associated with cognitive dysfunction and cognitive improvement following therapy using RNA sequencing to analyze the whole blood-derived transcriptome of altogether 101 MDD patients who enrolled in the CERT-D study. The mRNA(Nova)Seq based transcriptome was analyzed from whole blood taken at baseline assessment, and patients' cognitive performance was measured twice at baseline and following eight weeks of therapy by means of the THINC integrated tool. Thirty-six patients showed comparatively low cognitive performance at baseline assessment, and 32 patients showed comparatively strong cognitive improvement following therapy. Differential gene expression analysis was performed using limma to a significance threshold of 0.05 and a logFC cutoff of |1.2|. Although we observed some indications for expression differences related to low cognitive performance and cognitive therapy response, signals did not withstand adjustment for multiple testing. Applying WGCNA, we retrieved altogether 25 modules of co-expressed genes and we used a combination of correlational and linear analyses to identify modules related to baseline cognitive performance and cognitive improvement following therapy. Three immune modules reflected distinct but interrelated immune processes (the yellow module: neutrophil-mediated immunity, the darkorange module: interferon signaling, the tan module: platelet activation), and higher expression of the yellow (r = -0.21, p < .05), the dark orange (r = 0.2, p < .05), and the tan (r = -0.23, p < .05) module correlated significantly negatively with patients' cognitive baseline performance. Patients' cognitive baseline performance was a significant predictor of the darkorange module (b = -0.039, p < .05) and the tan module's expression (b = 0.02, p < .05) and was close to becoming a significant predictor of the yellow module's expression (b = -0.02, p = .05). Furthermore, patients characterized by comparatively low cognitive performance at baseline showed significantly higher expression of the tan module when compared to all other patients F(1,97) = 4.32, p < .05, η= 0.04. Following eight weeks of treatment, we observed altogether significant improvement in patients' cognitive performance (b = 0.30, p < .001), and patients with comparatively high cognitive gain showed noticeably lower, but not significantly lower F(1,98) = 3.76, p = .058, expression of a dark turquoise module, which reflects complement and B-cell-associated immune processes. Noteworthy, the relation between cognitive performance and module expression remained observable after controlling for symptom severity and BMI, which partly accounted for variance in module expression. As such, our findings provide further evidence for the involvement of immune processes in MDD related cognitive dysfunction and they suggest that different immune processes contribute to the development and long-term persistence of cognitive dysfunction in the context of depression.
Collapse
Affiliation(s)
- Johannes C S Zang
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
| | - Christa Hohoff
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
| | - Evelien Van Assche
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
| | - Pia Lange
- Institute of Medical Informatics, University of Münster, Münster, Germany.
| | - Manuel Kraft
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, Münster, Germany.
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany.
| | - Silke Jörgens
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
| | - Matthew J Knight
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, 48149 Münster, Germany; Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville, VIC 3010, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
10
|
Russell M, Aqi A, Saitou M, Gokcumen O, Masuda N. Gene communities in co-expression networks across different tissues. ARXIV 2023:arXiv:2305.12963v2. [PMID: 37292479 PMCID: PMC10246089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
With the recent availability of tissue-specific gene expression data, e.g., provided by the GTEx Consortium, there is interest in comparing gene co-expression patterns across tissues. One promising approach to this problem is to use a multilayer network analysis framework and perform multilayer community detection. Communities in gene co-expression networks reveal groups of genes similarly expressed across individuals, potentially involved in related biological processes responding to specific environmental stimuli or sharing common regulatory variations. We construct a multilayer network in which each of the four layers is an exocrine gland tissue-specific gene co-expression network. We develop methods for multilayer community detection with correlation matrix input and an appropriate null model. Our correlation matrix input method identifies five groups of genes that are similarly co-expressed in multiple tissues (a community that spans multiple layers, which we call a generalist community) and two groups of genes that are co-expressed in just one tissue (a community that lies primarily within just one layer, which we call a specialist community). We further found gene co-expression communities where the genes physically cluster across the genome significantly more than expected by chance (on chromosomes 1 and 11). This clustering hints at underlying regulatory elements determining similar expression patterns across individuals and cell types. We suggest that KRTAP3-1, KRTAP3-3, and KRTAP3-5 share regulatory elements in skin and pancreas. Furthermore, we find that CELA3A and CELA3B share associated expression quantitative trait loci in the pancreas. The results indicate that our multilayer community detection method for correlation matrix input extracts biologically interesting communities of genes.
Collapse
Affiliation(s)
| | - Alber Aqi
- Department of Biological Sciences, University at Buffalo
| | - Marie Saitou
- Faculty of Biosciences, Norwegian University of Life Sciences
| | - Omer Gokcumen
- Department of Biological Sciences, University at Buffalo
| | - Naoki Masuda
- Department of Mathematics, University at Buffalo
- Institute for Artificial Intelligence and Data Science, University at Buffalo
| |
Collapse
|
11
|
Brasher MS, Mize TJ, Thomas AL, Hoeffer CA, Ehringer MA, Evans LM. Testing associations between human anxiety and genes previously implicated by mouse anxiety models. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12851. [PMID: 37259642 PMCID: PMC10733569 DOI: 10.1111/gbb.12851] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 05/02/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
Anxiety disorders are common and can be debilitating, with effective treatments remaining hampered by an incomplete understanding of the underlying genetic etiology. Improvements have been made in understanding the genetic influences on mouse behavioral models of anxiety, yet it is unclear the extent to which genes identified in these experimental systems contribute to genetic variation in human anxiety phenotypes. Leveraging new and existing large-scale human genome-wide association studies, we tested whether sets of genes previously identified in mouse anxiety-like behavior studies contribute to a range of human anxiety disorders. When tested as individual genes, 13 mouse-identified genes were associated with human anxiety phenotypes, suggesting an overlap of individual genes contributing to both mouse models of anxiety-like behaviors and human anxiety traits. When genes were tested as sets, we did identify 14 significant associations between mouse gene sets and human anxiety, but the majority of gene sets showed no significant association with human anxiety phenotypes. These few significant associations indicate a need to identify and develop more translatable mouse models by identifying sets of genes that "match" between model systems and specific human phenotypes of interest. We suggest that continuing to develop improved behavioral paradigms and finer-scale experimental data, for instance from individual neuronal subtypes or cell-type-specific expression data, is likely to improve our understanding of the genetic etiology and underlying functional changes in anxiety disorders.
Collapse
Affiliation(s)
- Maizy S. Brasher
- Department of Ecology and Evolutionary BiologyUniversity of Colorado BoulderBoulderColoradoUSA
- Institute for Behavioral GeneticsBoulderColoradoUSA
| | - Travis J. Mize
- Department of Ecology and Evolutionary BiologyUniversity of Colorado BoulderBoulderColoradoUSA
- Institute for Behavioral GeneticsBoulderColoradoUSA
| | | | - Charles A. Hoeffer
- Institute for Behavioral GeneticsBoulderColoradoUSA
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Marissa A. Ehringer
- Institute for Behavioral GeneticsBoulderColoradoUSA
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Luke M. Evans
- Department of Ecology and Evolutionary BiologyUniversity of Colorado BoulderBoulderColoradoUSA
- Institute for Behavioral GeneticsBoulderColoradoUSA
| |
Collapse
|
12
|
Moore A, Marks JA, Quach BC, Guo Y, Bierut LJ, Gaddis NC, Hancock DB, Page GP, Johnson EO. Evaluating 17 methods incorporating biological function with GWAS summary statistics to accelerate discovery demonstrates a tradeoff between high sensitivity and high positive predictive value. Commun Biol 2023; 6:1199. [PMID: 38001305 PMCID: PMC10673847 DOI: 10.1038/s42003-023-05413-w] [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: 04/05/2022] [Accepted: 10/03/2023] [Indexed: 11/26/2023] Open
Abstract
Where sufficiently large genome-wide association study (GWAS) samples are not currently available or feasible, methods that leverage increasing knowledge of the biological function of variants may illuminate discoveries without increasing sample size. We comprehensively evaluated 17 functional weighting methods for identifying novel associations. We assessed the performance of these methods using published results from multiple GWAS waves across each of five complex traits. Although no method achieved both high sensitivity and positive predictive value (PPV) for any trait, a subset of methods utilizing pleiotropy and expression quantitative trait loci nominated variants with high PPV (>75%) for multiple traits. Application of functionally weighting methods to enhance GWAS power for locus discovery is unlikely to circumvent the need for larger sample sizes in truly underpowered GWAS, but these results suggest that applying functional weighting to GWAS can accurately nominate additional novel loci from available samples for follow-up studies.
Collapse
Affiliation(s)
- Amy Moore
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC, 27709, USA.
| | - Jesse A Marks
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC, 27709, USA
| | - Bryan C Quach
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC, 27709, USA
| | - Yuelong Guo
- GeneCentric Therapeutics, Inc., Cary, NC, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Nathan C Gaddis
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC, 27709, USA
| | - Dana B Hancock
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC, 27709, USA
| | - Grier P Page
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC, 27709, USA
- Fellow Program, RTI International, Research Triangle Park, NC, 27709, USA
| | - Eric O Johnson
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC, 27709, USA.
- Fellow Program, RTI International, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
13
|
Russell M, Aqil A, Saitou M, Gokcumen O, Masuda N. Gene communities in co-expression networks across different tissues. PLoS Comput Biol 2023; 19:e1011616. [PMID: 37976327 PMCID: PMC10691702 DOI: 10.1371/journal.pcbi.1011616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 12/01/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023] Open
Abstract
With the recent availability of tissue-specific gene expression data, e.g., provided by the GTEx Consortium, there is interest in comparing gene co-expression patterns across tissues. One promising approach to this problem is to use a multilayer network analysis framework and perform multilayer community detection. Communities in gene co-expression networks reveal groups of genes similarly expressed across individuals, potentially involved in related biological processes responding to specific environmental stimuli or sharing common regulatory variations. We construct a multilayer network in which each of the four layers is an exocrine gland tissue-specific gene co-expression network. We develop methods for multilayer community detection with correlation matrix input and an appropriate null model. Our correlation matrix input method identifies five groups of genes that are similarly co-expressed in multiple tissues (a community that spans multiple layers, which we call a generalist community) and two groups of genes that are co-expressed in just one tissue (a community that lies primarily within just one layer, which we call a specialist community). We further found gene co-expression communities where the genes physically cluster across the genome significantly more than expected by chance (on chromosomes 1 and 11). This clustering hints at underlying regulatory elements determining similar expression patterns across individuals and cell types. We suggest that KRTAP3-1, KRTAP3-3, and KRTAP3-5 share regulatory elements in skin and pancreas. Furthermore, we find that CELA3A and CELA3B share associated expression quantitative trait loci in the pancreas. The results indicate that our multilayer community detection method for correlation matrix input extracts biologically interesting communities of genes.
Collapse
Affiliation(s)
- Madison Russell
- Department of Mathematics, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Alber Aqil
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Marie Saitou
- Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Omer Gokcumen
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Naoki Masuda
- Department of Mathematics, State University of New York at Buffalo, Buffalo, New York, United States of America
- Institute for Artificial Intelligence and Data Science, State University of New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
14
|
Chatzinakos C, Pernia CD, Morrison FG, Iatrou A, McCullough KM, Schuler H, Snijders C, Bajaj T, DiPietro CP, Soliva Estruch M, Gassen NC, Anastasopoulos C, Bharadwaj RA, Bowlby BC, Hartmann J, Maihofer AX, Nievergelt CM, Ressler NM, Wolf EJ, Carlezon WA, Krystal JH, Kleinman JE, Girgenti MJ, Huber BR, Kellis M, Logue MW, Miller MW, Ressler KJ, Daskalakis NP. Single-Nucleus Transcriptome Profiling of Dorsolateral Prefrontal Cortex: Mechanistic Roles for Neuronal Gene Expression, Including the 17q21.31 Locus, in PTSD Stress Response. Am J Psychiatry 2023; 180:739-754. [PMID: 37491937 PMCID: PMC11406458 DOI: 10.1176/appi.ajp.20220478] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
OBJECTIVE Multidisciplinary studies of posttraumatic stress disorder (PTSD) and major depressive disorder (MDD) implicate the dorsolateral prefrontal cortex (DLPFC) in disease risk and pathophysiology. Postmortem brain studies have relied on bulk-tissue RNA sequencing (RNA-seq), but single-cell RNA-seq is needed to dissect cell-type-specific mechanisms. The authors conducted the first single-nucleus RNA-seq postmortem brain study in PTSD to elucidate disease transcriptomic pathology with cell-type-specific resolution. METHOD Profiling of 32 DLPFC samples from 11 individuals with PTSD, 10 with MDD, and 11 control subjects was conducted (∼415K nuclei; >13K cells per sample). A replication sample included 15 DLPFC samples (∼160K nuclei; >11K cells per sample). RESULTS Differential gene expression analyses identified significant single-nucleus RNA-seq differentially expressed genes (snDEGs) in excitatory (EX) and inhibitory (IN) neurons and astrocytes, but not in other cell types or bulk tissue. MDD samples had more false discovery rate-corrected significant snDEGs, and PTSD samples had a greater replication rate. In EX and IN neurons, biological pathways that were differentially enriched in PTSD compared with MDD included glucocorticoid signaling. Furthermore, glucocorticoid signaling in induced pluripotent stem cell (iPSC)-derived cortical neurons demonstrated greater relevance in PTSD and opposite direction of regulation compared with MDD, especially in EX neurons. Many snDEGs were from the 17q21.31 locus and are particularly interesting given causal roles in disease pathogenesis and DLPFC-based neuroimaging (PTSD: ARL17B, LINC02210-CRHR1, and LRRC37A2; MDD: LRRC37A and LRP4), while others were regulated by glucocorticoids in iPSC-derived neurons (PTSD: SLC16A6, TAF1C; MDD: CDH3). CONCLUSIONS The study findings point to cell-type-specific mechanisms of brain stress response in PTSD and MDD, highlighting the importance of examining cell-type-specific gene expression and indicating promising novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Chris Chatzinakos
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Cameron D Pernia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Filomene G Morrison
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Artemis Iatrou
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Kenneth M McCullough
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Heike Schuler
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Clara Snijders
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Thomas Bajaj
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Christopher P DiPietro
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Marina Soliva Estruch
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Nils C Gassen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Constantin Anastasopoulos
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Rahul A Bharadwaj
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Benjamin C Bowlby
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Jakob Hartmann
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Adam X Maihofer
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Caroline M Nievergelt
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Nicholas M Ressler
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Erika J Wolf
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - William A Carlezon
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - John H Krystal
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Joel E Kleinman
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Matthew J Girgenti
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Bertrand R Huber
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Manolis Kellis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Mark W Logue
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Mark W Miller
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Kerry J Ressler
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Mass. (Chatzinakos, Pernia, Iatrou, McCullough, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Hartmann, N.M. Ressler, Carlezon, K.J. Ressler, Daskalakis); Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Mass. (Chatzinakos, Pernia, Iatrou, Schuler, Snijders, DiPietro, Soliva Estruch, Anastasopoulos, Bowlby, Daskalakis); National Center for PTSD, VA Boston Healthcare System, Boston (Morrison, Wolf, Logue, Miller); Department of Psychiatry (Morrison, Wolf, Logue, Miller), Department of Neurology (Huber), and Department of Biomedical Genetics (Logue), Boston University School of Medicine, Boston; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands (Soliva Estruch, Snijders); RG Neurohomeostasis, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Bonn, Bonn, Germany (Bajaj, Gassen); Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland (Anastasopoulos); Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore (Bharadwaj, Kleinman); Department of Psychiatry, University of California San Diego, La Jolla (Maihofer, Nievergelt); Center for Excellence in Stress and Mental Health (Maihofer, Nievergelt) and Research Service (Maihofer, Nievergelt), Veterans Affairs San Diego Healthcare System, San Diego; Department of Psychiatry, Yale University School of Medicine, New Haven, Conn. (Krystal, Girgenti); Psychiatry Service, VA Connecticut Healthcare System, West Haven (Krystal, Girgenti); National Center for PTSD, Clinical Neurosciences Division, U.S. Department of Veterans Affairs, West Haven, Conn. (Krystal, Girgenti); Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore (Kleinman); Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston (Huber); Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, and Broad Institute of MIT and Harvard, Cambridge, Mass. (Kellis); Department of Biostatistics, Boston University School of Public Health, Boston (Logue)
| |
Collapse
|
15
|
Islam MK, Islam MR, Rahman MH, Islam MZ, Hasan MM, Mamun MMI, Moni MA. Integrated bioinformatics and statistical approach to identify the common molecular mechanisms of obesity that are linked to the development of two psychiatric disorders: Schizophrenia and major depressive disorder. PLoS One 2023; 18:e0276820. [PMID: 37494308 PMCID: PMC10370737 DOI: 10.1371/journal.pone.0276820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 10/13/2022] [Indexed: 07/28/2023] Open
Abstract
Obesity is a chronic multifactorial disease characterized by the accumulation of body fat and serves as a gateway to a number of metabolic-related diseases. Epidemiologic data indicate that Obesity is acting as a risk factor for neuro-psychiatric disorders such as schizophrenia, major depression disorder and vice versa. However, how obesity may biologically interact with neurodevelopmental or neurological psychiatric conditions influenced by hereditary, environmental, and other factors is entirely unknown. To address this issue, we have developed a pipeline that integrates bioinformatics and statistical approaches such as transcriptomic analysis to identify differentially expressed genes (DEGs) and molecular mechanisms in patients with psychiatric disorders that are also common in obese patients. Biomarker genes expressed in schizophrenia, major depression, and obesity have been used to demonstrate such relationships depending on the previous research studies. The highly expressed genes identify commonly altered signalling pathways, gene ontology pathways, and gene-disease associations across disorders. The proposed method identified 163 significant genes and 134 significant pathways shared between obesity and schizophrenia. Similarly, there are 247 significant genes and 65 significant pathways that are shared by obesity and major depressive disorder. These genes and pathways increase the likelihood that psychiatric disorders and obesity are pathogenic. Thus, this study may help in the development of a restorative approach that will ameliorate the bidirectional relation between obesity and psychiatric disorder. Finally, we also validated our findings using genome-wide association study (GWAS) and whole-genome sequence (WGS) data from SCZ, MDD, and OBE. We confirmed the likely involvement of four significant genes both in transcriptomic and GWAS/WGS data. Moreover, we have performed co-expression cluster analysis of the transcriptomic data and compared it with the results of transcriptomic differential expression analysis and GWAS/WGS.
Collapse
Affiliation(s)
- Md Khairul Islam
- Dept. of Information Communication Technology, Islamic University, Kushtia, Bangladesh
| | - Md Rakibul Islam
- Dept. of Information Communication Technology, Islamic University, Kushtia, Bangladesh
| | - Md Habibur Rahman
- Dept. of Computer Science Engineering, Islamic University, Kushtia, Bangladesh
| | - Md Zahidul Islam
- Dept. of Information Communication Technology, Islamic University, Kushtia, Bangladesh
| | - Md Mehedi Hasan
- Department of Statistics, University of Rajshahi, Rajshahi, Bangladesh
| | - Md Mainul Islam Mamun
- Department of Applied Physics and Electronic Engineering, University of Rajshahi, Rajshahi, Bangladesh
| | - Mohammad Ali Moni
- Dept. of Computer Science and Engineering, Pabna University of Science and Technology, Pabna, Bangladesh
| |
Collapse
|
16
|
Hicks EM, Seah C, Cote A, Marchese S, Brennand KJ, Nestler EJ, Girgenti MJ, Huckins LM. Integrating genetics and transcriptomics to study major depressive disorder: a conceptual framework, bioinformatic approaches, and recent findings. Transl Psychiatry 2023; 13:129. [PMID: 37076454 PMCID: PMC10115809 DOI: 10.1038/s41398-023-02412-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 04/21/2023] Open
Abstract
Major depressive disorder (MDD) is a complex and heterogeneous psychiatric syndrome with genetic and environmental influences. In addition to neuroanatomical and circuit-level disturbances, dysregulation of the brain transcriptome is a key phenotypic signature of MDD. Postmortem brain gene expression data are uniquely valuable resources for identifying this signature and key genomic drivers in human depression; however, the scarcity of brain tissue limits our capacity to observe the dynamic transcriptional landscape of MDD. It is therefore crucial to explore and integrate depression and stress transcriptomic data from numerous, complementary perspectives to construct a richer understanding of the pathophysiology of depression. In this review, we discuss multiple approaches for exploring the brain transcriptome reflecting dynamic stages of MDD: predisposition, onset, and illness. We next highlight bioinformatic approaches for hypothesis-free, genome-wide analyses of genomic and transcriptomic data and their integration. Last, we summarize the findings of recent genetic and transcriptomic studies within this conceptual framework.
Collapse
Affiliation(s)
- Emily M Hicks
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Carina Seah
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Alanna Cote
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Shelby Marchese
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Kristen J Brennand
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Matthew J Girgenti
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
17
|
Mize TJ, Funkhouser SA, Buck JM, Stitzel JA, Ehringer MA, Evans LM. Testing Association of Previously Implicated Gene Sets and Gene-Networks in Nicotine Exposed Mouse Models with Human Smoking Phenotypes. Nicotine Tob Res 2023; 25:1030-1038. [PMID: 36444815 PMCID: PMC10077928 DOI: 10.1093/ntr/ntac269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 08/15/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Smoking behaviors are partly heritable, yet the genetic and environmental mechanisms underlying smoking phenotypes are not fully understood. Developmental nicotine exposure (DNE) is a significant risk factor for smoking and leads to gene expression changes in mouse models; however, it is unknown whether the same genes whose expression is impacted by DNE are also those underlying smoking genetic liability. We examined whether genes whose expression in D1-type striatal medium spiny neurons due to DNE in the mouse are also associated with human smoking behaviors. METHODS Specifically, we assessed whether human orthologs of mouse-identified genes, either individually or as a set, were genetically associated with five human smoking traits using MAGMA and S-LDSC while implementing a novel expression-based gene-SNP annotation methodology. RESULTS We found no strong evidence that these genes sets were more strongly associated with smoking behaviors than the rest of the genome, but ten of these individual genes were significantly associated with three of the five human smoking traits examined (p < 2.5e-6). Three of these genes have not been reported previously and were discovered only when implementing the expression-based annotation. CONCLUSIONS These results suggest the genes whose expression is impacted by DNE in mice are largely distinct from those contributing to smoking genetic liability in humans. However, examining a single mouse neuronal cell type may be too fine a resolution for comparison, suggesting that experimental manipulation of nicotine consumption, reward, or withdrawal in mice may better capture genes related to the complex genetics of human tobacco use. IMPLICATIONS Genes whose expression is impacted by DNE in mouse D1-type striatal medium spiny neurons were not found to be, as a whole, more strongly associated with human smoking behaviors than the rest of the genome, though ten individual mouse-identified genes were associated with human smoking traits. This suggests little overlap between the genetic mechanisms impacted by DNE and those influencing heritable liability to smoking phenotypes in humans. Further research is warranted to characterize how developmental nicotine exposure paradigms in mice can be translated to understand nicotine use in humans and their heritable effects on smoking.
Collapse
Affiliation(s)
- Travis J Mize
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA
| | - Scott A Funkhouser
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, USA
| | - Marissa A Ehringer
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, USA
| | - Luke M Evans
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA
| |
Collapse
|
18
|
Ye X, Shi T, Cui Y, Sakurai T. Interactive gene identification for cancer subtyping based on multi-omics clustering. Methods 2023; 211:61-67. [PMID: 36804215 DOI: 10.1016/j.ymeth.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/06/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Recent advances in multi-omics databases offer the opportunity to explore complex systems of cancers across hierarchical biological levels. Some methods have been proposed to identify the genes that play a vital role in disease development by integrating multi-omics. However, the existing methods identify the related genes separately, neglecting the gene interactions that are related to the multigenic disease. In this study, we develop a learning framework to identify the interactive genes based on multi-omics data including gene expression. Firstly, we integrate different omics based on their similarities and apply spectral clustering for cancer subtype identification. Then, a gene co-expression network is construct for each cancer subtype. Finally, we detect the interactive genes in the co-expression network by learning the dense subgraphs based on the L1 prosperities of eigenvectors in the modularity matrix. We apply the proposed learning framework on a multi-omics cancer dataset to identify the interactive genes for each cancer subtype. The detected genes are examined by DAVID and KEGG tools for systematic gene ontology enrichment analysis. The analysis results show that the detected genes have relationships to cancer development and the genes in different cancer subtypes are related to different biological processes and pathways, which are expected to yield important references for understanding tumor heterogeneity and improving patient survival.
Collapse
Affiliation(s)
- Xiucai Ye
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Tianyi Shi
- Tsukuba Life Science Innovation Program, University of Tsukuba, Tsukuba 3058577, Japan
| | - Yaxuan Cui
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| | - Tetsuya Sakurai
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan; Tsukuba Life Science Innovation Program, University of Tsukuba, Tsukuba 3058577, Japan
| |
Collapse
|
19
|
Li A, Liu S, Bakshi A, Jiang L, Chen W, Zheng Z, Sullivan PF, Visscher PM, Wray NR, Yang J, Zeng J. mBAT-combo: A more powerful test to detect gene-trait associations from GWAS data. Am J Hum Genet 2023; 110:30-43. [PMID: 36608683 PMCID: PMC9892780 DOI: 10.1016/j.ajhg.2022.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Gene-based association tests aggregate multiple SNP-trait associations into sets defined by gene boundaries and are widely used in post-GWAS analysis. A common approach for gene-based tests is to combine SNPs associations by computing the sum of χ2 statistics. However, this strategy ignores the directions of SNP effects, which could result in a loss of power for SNPs with masking effects, e.g., when the product of two SNP effects and the linkage disequilibrium (LD) correlation is negative. Here, we introduce "mBAT-combo," a set-based test that is better powered than other methods to detect multi-SNP associations in the context of masking effects. We validate the method through simulations and applications to real data. We find that of 35 blood and urine biomarker traits in the UK Biobank, 34 traits show evidence for masking effects in a total of 4,273 gene-trait pairs, indicating that masking effects is common in complex traits. We further validate the improved power of our method in height, body mass index, and schizophrenia with different GWAS sample sizes and show that on average 95.7% of the genes detected only by mBAT-combo with smaller sample sizes can be identified by the single-SNP approach with a 1.7-fold increase in sample sizes. Eleven genes significant only in mBAT-combo for schizophrenia are confirmed by functionally informed fine-mapping or Mendelian randomization integrating gene expression data. The framework of mBAT-combo can be applied to any set of SNPs to refine trait-association signals hidden in genomic regions with complex LD structures.
Collapse
Affiliation(s)
- Ang Li
- Institute for Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Shouye Liu
- Institute for Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Andrew Bakshi
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | | | - Wenhan Chen
- Epigenetics Research Laboratory, Genomics and Epigenetics Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Zhili Zheng
- Institute for Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden; Departments of Genetics and Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Peter M Visscher
- Institute for Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Naomi R Wray
- Institute for Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia; Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Jian Yang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Jian Zeng
- Institute for Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
20
|
Dovrolis N, Filidou E, Tarapatzi G, Kokkotis G, Spathakis M, Kandilogiannakis L, Drygiannakis I, Valatas V, Arvanitidis K, Karakasiliotis I, Vradelis S, Manolopoulos VG, Paspaliaris V, Bamias G, Kolios G. Co-expression of fibrotic genes in inflammatory bowel disease; A localized event? Front Immunol 2022; 13:1058237. [PMID: 36632136 PMCID: PMC9826764 DOI: 10.3389/fimmu.2022.1058237] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/08/2022] [Indexed: 12/27/2022] Open
Abstract
Introduction Extracellular matrix turnover, a ubiquitous dynamic biological process, can be diverted to fibrosis. The latter can affect the intestine as a serious complication of Inflammatory Bowel Diseases (IBD) and is resistant to current pharmacological interventions. It embosses the need for out-of-the-box approaches to identify and target molecular mechanisms of fibrosis. Methods and results In this study, a novel mRNA sequencing dataset of 22 pairs of intestinal biopsies from the terminal ileum (TI) and the sigmoid of 7 patients with Crohn's disease, 6 with ulcerative colitis and 9 control individuals (CI) served as a validation cohort of a core fibrotic transcriptomic signature (FIBSig), This signature, which was identified in publicly available data (839 samples from patients and healthy individuals) of 5 fibrotic disorders affecting different organs (GI tract, lung, skin, liver, kidney), encompasses 241 genes and the functional pathways which derive from their interactome. These genes were used in further bioinformatics co-expression analyses to elucidate the site-specific molecular background of intestinal fibrosis highlighting their involvement, particularly in the terminal ileum. We also confirmed different transcriptomic profiles of the sigmoid and terminal ileum in our validation cohort. Combining the results of these analyses we highlight 21 core hub genes within a larger single co-expression module, highly enriched in the terminal ileum of CD patients. Further pathway analysis revealed known and novel inflammation-regulated, fibrogenic pathways operating in the TI, such as IL-13 signaling and pyroptosis, respectively. Discussion These findings provide a rationale for the increased incidence of fibrosis at the terminal ileum of CD patients and highlight operating pathways in intestinal fibrosis for future evaluation with mechanistic and translational studies.
Collapse
Affiliation(s)
- Nikolas Dovrolis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Laboratory of Biology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece,*Correspondence: George Kolios, ; Nikolas Dovrolis,
| | - Eirini Filidou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Gesthimani Tarapatzi
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Georgios Kokkotis
- Gastrointestinal (GI) Unit, 3 Department of Internal Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Michail Spathakis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Leonidas Kandilogiannakis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Ioannis Drygiannakis
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, Heraklion, Greece
| | - Vassilis Valatas
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, Heraklion, Greece
| | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | - Ioannis Karakasiliotis
- Laboratory of Biology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Stergios Vradelis
- Second Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece
| | | | - Giorgos Bamias
- Gastrointestinal (GI) Unit, 3 Department of Internal Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - George Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), Alexandroupolis, Greece,*Correspondence: George Kolios, ; Nikolas Dovrolis,
| |
Collapse
|
21
|
Roy B, Ochi S, Dwivedi Y. M6A RNA Methylation-Based Epitranscriptomic Modifications in Plasticity-Related Genes via miR-124-C/EBPα-FTO-Transcriptional Axis in the Hippocampus of Learned Helplessness Rats. Int J Neuropsychopharmacol 2022; 25:1037-1049. [PMID: 36161325 PMCID: PMC9743968 DOI: 10.1093/ijnp/pyac068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Impaired synaptic plasticity has been linked to dynamic gene regulatory network changes. Recently, gene regulation has been introduced with the emerging concept of unique N6-methyladenosine (m6A)-based reversible transcript methylation. In this study, we tested whether m6A RNA methylation may potentially serve as a link between the stressful insults and altered expression of plasticity-related genes. METHODS Expression of plasticity genes Nr3c1, Creb1, Ntrk2; m6A-modifying enzymes Fto, methyltransferase like (Mettl)-3 and 14; DNA methylation enzymes Dnmt1, Dnmt3a; transcription factor C/ebp-α; and miRNA-124-3p were determined by quantitative polymerase chain reaction (qPCR) in the hippocampus of rats that showed susceptibility to develop stress-induced depression (learned helplessness). M6A methylation of plasticity-related genes was determined following m6A mRNA immunoprecipitation. Chromatin immunoprecipitation was used to examine the endogenous binding of C/EBP-α to the Fto promoter. MiR-124-mediated post-transcriptional inhibition of Fto via C/EBPα was determined using an in vitro model. RESULTS Hippocampus of learned helplessness rats showed downregulation of Nr3c1, Creb1, and Ntrk2 along with enrichment in their m6A methylation. A downregulation in demethylating enzyme Fto and upregulation in methylating enzyme Mettl3 were also noted. The Fto promoter was hypomethylated due to the lower expression of Dnmt1 and Dnmt3a. At the same time, there was a lower occupancy of transcription factor C/EBPα on the Fto promoter. Conversely, C/ebp-α transcript was downregulated via induced miR-124-3p expression. CONCLUSIONS Our study mechanistically linked defective C/EBP-α-FTO-axis, epigenetically influenced by induced expression of miR-124-3p, in modifying m6A enrichment in plasticity-related genes. This could potentially be linked with abnormal neuronal plasticity in depression.
Collapse
Affiliation(s)
- Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama atBirmingham, Birmingham, Alabama, USA
| | - Shinichiro Ochi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama atBirmingham, Birmingham, Alabama, USA,Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, Japan
| | - Yogesh Dwivedi
- Correspondence: Yogesh Dwivedi, PhD, Elesabeth Ridgely Shook Professor, Director of Translational Research, UAB Mood Disorder Program, Codirector, Depression and Suicide Center, Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, SC711 Sparks Center, 1720 2nd Avenue South, Birmingham, AL, USA ()
| |
Collapse
|
22
|
Cabana-Domínguez J, Soler Artigas M, Arribas L, Alemany S, Vilar-Ribó L, Llonga N, Fadeuilhe C, Corrales M, Richarte V, Ramos-Quiroga JA, Ribasés M. Comprehensive analysis of omics data identifies relevant gene networks for Attention-Deficit/Hyperactivity Disorder (ADHD). Transl Psychiatry 2022; 12:409. [PMID: 36153331 PMCID: PMC9509350 DOI: 10.1038/s41398-022-02182-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly prevalent neurodevelopmental disorder that results from the interaction of both genetic and environmental risk factors. Genome-wide association studies have started to identify multiple genetic risk loci associated with ADHD, however, the exact causal genes and biological mechanisms remain largely unknown. We performed a multi-step analysis to identify and characterize modules of co-expressed genes associated with ADHD using data from peripheral blood mononuclear cells of 270 ADHD cases and 279 controls. We identified seven ADHD-associated modules of co-expressed genes, some of them enriched in both genetic and epigenetic signatures for ADHD and in biological pathways relevant for psychiatric disorders, such as the regulation of gene expression, epigenetics and immune system. In addition, for some of the modules, we found evidence of potential regulatory mechanisms, including microRNAs and common genetic variants. In conclusion, our results point to promising genes and pathways for ADHD, supporting the use of peripheral blood to assess gene expression signatures in psychiatric disorders. Furthermore, they highlight that the combination of multi-omics signals provides deeper and broader insights into the biological mechanisms underlying ADHD.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain.
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
| | - María Soler Artigas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Lorena Arribas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Silvia Alemany
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Laura Vilar-Ribó
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Natalia Llonga
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Christian Fadeuilhe
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Montse Corrales
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vanesa Richarte
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Josep Antoni Ramos-Quiroga
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Ribasés
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain.
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
23
|
Bankier S, Michoel T. eQTLs as causal instruments for the reconstruction of hormone linked gene networks. Front Endocrinol (Lausanne) 2022; 13:949061. [PMID: 36060942 PMCID: PMC9428692 DOI: 10.3389/fendo.2022.949061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Hormones act within in highly dynamic systems and much of the phenotypic response to variation in hormone levels is mediated by changes in gene expression. The increase in the number and power of large genetic association studies has led to the identification of hormone linked genetic variants. However, the biological mechanisms underpinning the majority of these loci are poorly understood. The advent of affordable, high throughput next generation sequencing and readily available transcriptomic databases has shown that many of these genetic variants also associate with variation in gene expression levels as expression Quantitative Trait Loci (eQTLs). In addition to further dissecting complex genetic variation, eQTLs have been applied as tools for causal inference. Many hormone networks are driven by transcription factors, and many of these genes can be linked to eQTLs. In this mini-review, we demonstrate how causal inference and gene networks can be used to describe the impact of hormone linked genetic variation upon the transcriptome within an endocrinology context.
Collapse
Affiliation(s)
- Sean Bankier
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | | |
Collapse
|
24
|
Novel functional genomics approaches bridging neuroscience and psychiatry. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022. [PMID: 37519472 PMCID: PMC10382709 DOI: 10.1016/j.bpsgos.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The possibility of establishing a metric of individual genetic risk for a particular disease or trait has sparked the interest of the clinical and research communities, with many groups developing and validating genomic profiling methodologies for their potential application in clinical care. Current approaches for calculating genetic risk to specific psychiatric conditions consist of aggregating genome-wide association studies-derived estimates into polygenic risk scores, which broadly represent the number of inherited risk alleles for an individual. While the traditional approach for polygenic risk score calculation aggregates estimates of gene-disease associations, novel alternative approaches have started to consider functional molecular phenotypes that are closer to genetic variation and are less penalized by the multiple testing required in genome-wide association studies. Moving the focus from genotype-disease to genotype-gene regulation frameworks, these novel approaches incorporate prior knowledge regarding biological processes involved in disease and aggregate estimates for the association of genotypes and phenotypes using multi-omics data modalities. In this review, we discuss and list different functional genomics tools that can be used and integrated to inform researchers and clinicians for a better understanding and diagnosis of psychopathology. We suggest that these novel approaches can help generate biologically driven hypotheses for polygenic signals that can ultimately serve the clinical community as potential biomarkers of psychiatric disease susceptibility.
Collapse
|
25
|
Gerring ZF, Thorp JG, Gamazon ER, Derks EM. An analysis of genetically regulated gene expression and the role of co-expression networks across 16 psychiatric and substance use phenotypes. Eur J Hum Genet 2022; 30:560-566. [PMID: 35217801 PMCID: PMC9090912 DOI: 10.1038/s41431-022-01037-6] [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: 04/30/2021] [Revised: 12/15/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified thousands of risk loci for psychiatric and substance use phenotypes, however the biological consequences of these loci remain largely unknown. We performed a transcriptome-wide association study of 10 psychiatric disorders and 6 substance use phenotypes (GWAS sample size range, N = 9725-807,553) using expression quantitative trait loci data from 532 prefrontal cortex samples. We estimated the correlation of genetically regulated expression between phenotype pairs, and compared the results with the genetic correlations. We identified 393 genes with at least one significant phenotype association, comprising 458 significant associations across 16 phenotypes. Overall, the transcriptomic correlations for phenotype pairs were significantly higher than the respective genetic correlations. For example, attention deficit hyperactivity disorder and autism spectrum disorder, both childhood developmental disorders, had significantly higher transcriptomic correlation (r = 0.84) than genetic correlation (r = 0.35). Finally, we tested the enrichment of phenotype-associated genes in gene co-expression networks built from human prefrontal cortex samples. Phenotype-associated genes were enriched in multiple gene co-expression modules and the implicated modules contained genes involved in mRNA splicing and glutamatergic receptors, among others. Together, our results highlight the utility of gene expression data in the understanding of functional gene mechanisms underlying psychiatric disorders and substance use phenotypes.
Collapse
Affiliation(s)
- Zachary F Gerring
- Translational Neurogenomics Laboratory; QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| | - Jackson G Thorp
- Translational Neurogenomics Laboratory; QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Clare Hall & MRC Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Eske M Derks
- Translational Neurogenomics Laboratory; QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Li X, Jiang L, Xue C, Li MJ, Li M. A conditional gene-based association framework integrating isoform-level eQTL data reveals new susceptibility genes for schizophrenia. eLife 2022; 11:e70779. [PMID: 35412455 PMCID: PMC9005191 DOI: 10.7554/elife.70779] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Linkage disequilibrium and disease-associated variants in the non-coding regions make it difficult to distinguish the truly associated genes from the redundantly associated genes for complex diseases. In this study, we proposed a new conditional gene-based framework called eDESE that leveraged an improved effective chi-squared statistic to control the type I error rates and remove the redundant associations. eDESE initially performed the association analysis by mapping variants to genes according to their physical distance. We further demonstrated that the isoform-level eQTLs could be more powerful than the gene-level eQTLs in the association analysis using a simulation study. Then the eQTL-guided strategies, that is, mapping variants to genes according to their gene/isoform-level variant-gene cis-eQTLs associations, were also integrated with eDESE. We then applied eDESE to predict the potential susceptibility genes of schizophrenia and found that the potential susceptibility genes were enriched with many neuronal or synaptic signaling-related terms in the Gene Ontology knowledgebase and antipsychotics-gene interaction terms in the drug-gene interaction database (DGIdb). More importantly, seven potential susceptibility genes identified by eDESE were the target genes of multiple antipsychotics in DrugBank. Comparing the potential susceptibility genes identified by eDESE and other benchmark approaches (i.e., MAGMA and S-PrediXcan) implied that strategy based on the isoform-level eQTLs could be an important supplement for the other two strategies (physical distance and gene-level eQTLs). We have implemented eDESE in our integrative platform KGGSEE (http://pmglab.top/kggsee/#/) and hope that eDESE can facilitate the prediction of candidate susceptibility genes and isoforms for complex diseases in a multi-tissue context.
Collapse
Affiliation(s)
- Xiangyi Li
- Program in Bioinformatics, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of EducationGuangzhouChina
- Center for Precision Medicine, Sun Yat-sen UniversityGuangzhouChina
| | - Lin Jiang
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouChina
| | - Chao Xue
- Program in Bioinformatics, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of EducationGuangzhouChina
- Center for Precision Medicine, Sun Yat-sen UniversityGuangzhouChina
| | - Mulin Jun Li
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical UniversityTianjinChina
| | - Miaoxin Li
- Program in Bioinformatics, Zhongshan School of Medicine and The Fifth Affiliated Hospital, Sun Yat-sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of EducationGuangzhouChina
- Center for Precision Medicine, Sun Yat-sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen UniversityZhuhaiChina
| |
Collapse
|
27
|
Identification of Key Modules and Genes Associated with Major Depressive Disorder in Adolescents. Genes (Basel) 2022; 13:genes13030464. [PMID: 35328018 PMCID: PMC8949287 DOI: 10.3390/genes13030464] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 12/25/2022] Open
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide. Adolescence is a crucial period for the occurrence and development of depression. There are essential distinctions between adolescent and adult depression patients, and the etiology of depressive disorder is unclear. The interactions of multiple genes in a co-expression network are likely to be involved in the physiopathology of MDD. In the present study, RNA-Seq data of mRNA were acquired from the peripheral blood of MDD in adolescents and healthy control (HC) subjects. Co-expression modules were constructed via weighted gene co-expression network analysis (WGCNA) to investigate the relationships between the underlying modules and MDD in adolescents. In the combined MDD and HC groups, the dynamic tree cutting method was utilized to assign genes to modules through hierarchical clustering. Moreover, functional enrichment analysis was conducted on those co-expression genes from interested modules. The results showed that eight modules were constructed by WGCNA. The blue module was significantly associated with MDD after multiple comparison adjustment. Several Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways associated with stress and inflammation were identified in this module, including histone methylation, apoptosis, NF-kappa β signaling pathway, and TNF signaling pathway. Five genes related to inflammation, immunity, and the nervous system were identified as hub genes: CNTNAP3, IL1RAP, MEGF9, UBE2W, and UBE2D1. All of these findings supported that MDD was associated with stress, inflammation, and immune responses, helping us to obtain a better understanding of the internal molecular mechanism and to explore biomarkers for the diagnosis or treatment of depression in adolescents.
Collapse
|
28
|
Xiang J, Zhang J, Zhao Y, Wu FX, Li M. Biomedical data, computational methods and tools for evaluating disease-disease associations. Brief Bioinform 2022; 23:6522999. [PMID: 35136949 DOI: 10.1093/bib/bbac006] [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: 10/19/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, exploring potential relationships between diseases has been an active research field. With the rapid accumulation of disease-related biomedical data, a lot of computational methods and tools/platforms have been developed to reveal intrinsic relationship between diseases, which can provide useful insights to the study of complex diseases, e.g. understanding molecular mechanisms of diseases and discovering new treatment of diseases. Human complex diseases involve both external phenotypic abnormalities and complex internal molecular mechanisms in organisms. Computational methods with different types of biomedical data from phenotype to genotype can evaluate disease-disease associations at different levels, providing a comprehensive perspective for understanding diseases. In this review, available biomedical data and databases for evaluating disease-disease associations are first summarized. Then, existing computational methods for disease-disease associations are reviewed and classified into five groups in terms of the usages of biomedical data, including disease semantic-based, phenotype-based, function-based, representation learning-based and text mining-based methods. Further, we summarize software tools/platforms for computation and analysis of disease-disease associations. Finally, we give a discussion and summary on the research of disease-disease associations. This review provides a systematic overview for current disease association research, which could promote the development and applications of computational methods and tools/platforms for disease-disease associations.
Collapse
Affiliation(s)
- Ju Xiang
- School of Computer Science and Engineering, Central South University, China
| | - Jiashuai Zhang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Yichao Zhao
- School of Computer Science and Engineering, Central South University, China
| | - Fang-Xiang Wu
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Min Li
- Division of Biomedical Engineering and Department of Mechanical Engineering at University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
29
|
Singla RK, Joon S, Shen L, Shen B. Translational Informatics for Natural Products as Antidepressant Agents. Front Cell Dev Biol 2022; 9:738838. [PMID: 35127696 PMCID: PMC8811306 DOI: 10.3389/fcell.2021.738838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/13/2021] [Indexed: 12/18/2022] Open
Abstract
Depression, a neurological disorder, is a universally common and debilitating illness where social and economic issues could also become one of its etiologic factors. From a global perspective, it is the fourth leading cause of long-term disability in human beings. For centuries, natural products have proven their true potential to combat various diseases and disorders, including depression and its associated ailments. Translational informatics applies informatics models at molecular, imaging, individual, and population levels to promote the translation of basic research to clinical applications. The present review summarizes natural-antidepressant-based translational informatics studies and addresses challenges and opportunities for future research in the field.
Collapse
Affiliation(s)
- Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Shikha Joon
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Li Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Zhang S, Yang X, Si S, Zhang J. The neurobiological basis of divergent thinking: Insight from gene co-expression network-based analysis. Neuroimage 2021; 245:118762. [PMID: 34838948 DOI: 10.1016/j.neuroimage.2021.118762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/25/2021] [Accepted: 11/23/2021] [Indexed: 11/30/2022] Open
Abstract
Although many efforts have been made to explore the genetic basis of divergent thinking (DT), there is still a gap in the understanding of how these findings relate to the neurobiology of DT. In a combined sample of 1,682 Chinese participants, by integrating GWAS with previously identified brain-specific gene co-expression network modules, this study explored for the first time the functional brain-specific gene co-expression networks underlying DT. The results showed that gene co-expression network modules in anterior cingulate cortex, caudate, amygdala and substantia nigra were enriched with DT association signals. Further functional enrichment analysis showed that these DT-related gene co-expression network modules were enriched for key biological process and cellular component related to myelination, suggesting that cortical and sub-cortical grey matter myelination may serve as important neurobiological basis of DT. Although the underlying mechanisms need to be further refined, this exploratory study may provide new insight into the neurobiology of DT.
Collapse
Affiliation(s)
- Shun Zhang
- Department of Psychology, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, China
| | - Xiaolei Yang
- College of Life Science, Qilu Normal University, Jinan, China
| | - Si Si
- Department of Psychology, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, China
| | - Jinghuan Zhang
- Department of Psychology, Shandong Normal University, No. 88 East Wenhua Road, Jinan 250014, China.
| |
Collapse
|
31
|
Remes O, Mendes JF, Templeton P. Biological, Psychological, and Social Determinants of Depression: A Review of Recent Literature. Brain Sci 2021; 11:1633. [PMID: 34942936 PMCID: PMC8699555 DOI: 10.3390/brainsci11121633] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022] Open
Abstract
Depression is one of the leading causes of disability, and, if left unmanaged, it can increase the risk for suicide. The evidence base on the determinants of depression is fragmented, which makes the interpretation of the results across studies difficult. The objective of this study is to conduct a thorough synthesis of the literature assessing the biological, psychological, and social determinants of depression in order to piece together the puzzle of the key factors that are related to this condition. Titles and abstracts published between 2017 and 2020 were identified in PubMed, as well as Medline, Scopus, and PsycInfo. Key words relating to biological, social, and psychological determinants as well as depression were applied to the databases, and the screening and data charting of the documents took place. We included 470 documents in this literature review. The findings showed that there are a plethora of risk and protective factors (relating to biological, psychological, and social determinants) that are related to depression; these determinants are interlinked and influence depression outcomes through a web of causation. In this paper, we describe and present the vast, fragmented, and complex literature related to this topic. This review may be used to guide practice, public health efforts, policy, and research related to mental health and, specifically, depression.
Collapse
Affiliation(s)
- Olivia Remes
- Institute for Manufacturing, University of Cambridge, Cambridge CB3 0FS, UK
| | | | - Peter Templeton
- IfM Engage Limited, Institute for Manufacturing, University of Cambridge, Cambridge CB3 0FS, UK;
- The William Templeton Foundation for Young People’s Mental Health (YPMH), Cambridge CB2 0AH, UK
| |
Collapse
|
32
|
Polikowsky HG, Shaw DM, Petty LE, Chen HH, Pruett DG, Linklater JP, Viljoen KZ, Beilby JM, Highland HM, Levitt B, Avery CL, Mullan Harris K, Jones RM, Below JE, Kraft SJ. Population-based genetic effects for developmental stuttering. HGG ADVANCES 2021; 3:100073. [PMID: 35047858 PMCID: PMC8756529 DOI: 10.1016/j.xhgg.2021.100073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Despite a lifetime prevalence of at least 5%, developmental stuttering, characterized by prolongations, blocks, and repetitions of speech sounds, remains a largely idiopathic speech disorder. Family, twin, and segregation studies overwhelmingly support a strong genetic influence on stuttering risk; however, its complex mode of inheritance combined with thus-far underpowered genetic studies contribute to the challenge of identifying and reproducing genes implicated in developmental stuttering susceptibility. We conducted a trans-ancestry genome-wide association study (GWAS) and meta-analysis of developmental stuttering in two primary datasets: The International Stuttering Project comprising 1,345 clinically ascertained cases from multiple global sites and 6,759 matched population controls from the biobank at Vanderbilt University Medical Center (VUMC), and 785 self-reported stuttering cases and 7,572 controls ascertained from The National Longitudinal Study of Adolescent to Adult Health (Add Health). Meta-analysis of these genome-wide association studies identified a genome-wide significant (GWS) signal for clinically reported developmental stuttering in the general population: a protective variant in the intronic or genic upstream region of SSUH2 (rs113284510, protective allele frequency = 7.49%, Z = -5.576, p = 2.46 × 10-8) that acts as an expression quantitative trait locus (eQTL) in esophagus-muscularis tissue by reducing its gene expression. In addition, we identified 15 loci reaching suggestive significance (p < 5 × 10-6). This foundational population-based genetic study of a common speech disorder reports the findings of a clinically ascertained study of developmental stuttering and highlights the need for further research.
Collapse
Affiliation(s)
- Hannah G. Polikowsky
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas M. Shaw
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lauren E. Petty
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dillon G. Pruett
- Hearing and Speech Sciences, Vanderbilt University, Nashville, TN, USA
| | | | | | - Janet M. Beilby
- Curtin School of Allied Health, Curtin University, Perth, WA, Australia
| | - Heather M. Highland
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brandt Levitt
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christy L. Avery
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kathleen Mullan Harris
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robin M. Jones
- Hearing and Speech Sciences, Vanderbilt University, Nashville, TN, USA
| | - Jennifer E. Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA,Corresponding author
| | - Shelly Jo Kraft
- Communication Sciences and Disorders, Wayne State University, Detroit, MI, USA,Corresponding author
| |
Collapse
|
33
|
Yurko R, Roeder K, Devlin B, G'Sell M. An approach to gene-based testing accounting for dependence of tests among nearby genes. Brief Bioinform 2021; 22:6359004. [PMID: 34459489 DOI: 10.1093/bib/bbab329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/20/2021] [Accepted: 07/29/2021] [Indexed: 11/14/2022] Open
Abstract
In genome-wide association studies (GWAS), it has become commonplace to test millions of single-nucleotide polymorphisms (SNPs) for phenotypic association. Gene-based testing can improve power to detect weak signal by reducing multiple testing and pooling signal strength. While such tests account for linkage disequilibrium (LD) structure of SNP alleles within each gene, current approaches do not capture LD of SNPs falling in different nearby genes, which can induce correlation of gene-based test statistics. We introduce an algorithm to account for this correlation. When a gene's test statistic is independent of others, it is assessed separately; when test statistics for nearby genes are strongly correlated, their SNPs are agglomerated and tested as a locus. To provide insight into SNPs and genes driving association within loci, we develop an interactive visualization tool to explore localized signal. We demonstrate our approach in the context of weakly powered GWAS for autism spectrum disorder, which is contrasted to more highly powered GWAS for schizophrenia and educational attainment. To increase power for these analyses, especially those for autism, we use adaptive $P$-value thresholding, guided by high-dimensional metadata modeled with gradient boosted trees, highlighting when and how it can be most useful. Notably our workflow is based on summary statistics.
Collapse
Affiliation(s)
- Ronald Yurko
- Department of Statistics & Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Kathryn Roeder
- Department of Computational Biology, Carnegie Mellon University, USA
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, USA
| | - Max G'Sell
- Department of Statistics & Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
34
|
Integration of functional genomics data to uncover cell type-specific pathways affected in Parkinson's disease. Biochem Soc Trans 2021; 49:2091-2100. [PMID: 34581766 PMCID: PMC8589426 DOI: 10.1042/bst20210128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent late-onset neurodegenerative disorder worldwide after Alzheimer's disease for which available drugs only deliver temporary symptomatic relief. Loss of dopaminergic neurons (DaNs) in the substantia nigra and intracellular alpha-synuclein inclusions are the main hallmarks of the disease but the events that cause this degeneration remain uncertain. Despite cell types other than DaNs such as astrocytes, microglia and oligodendrocytes have been recently associated with the pathogenesis of PD, we still lack an in-depth characterisation of PD-affected brain regions at cell-type resolution that could help our understanding of the disease mechanisms. Nevertheless, publicly available large-scale brain-specific genomic, transcriptomic and epigenomic datasets can be further exploited to extract different layers of cell type-specific biological information for the reconstruction of cell type-specific transcriptional regulatory networks. By intersecting disease risk variants within the networks, it may be possible to study the functional role of these risk variants and their combined effects at cell type- and pathway levels, that, in turn, can facilitate the identification of key regulators involved in disease progression, which are often potential therapeutic targets.
Collapse
|
35
|
Wang W, Han R, Zhang M, Wang Y, Wang T, Wang Y, Shang X, Peng J. A network-based method for brain disease gene prediction by integrating brain connectome and molecular network. Brief Bioinform 2021; 23:6415315. [PMID: 34727570 DOI: 10.1093/bib/bbab459] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/18/2021] [Accepted: 10/07/2021] [Indexed: 12/27/2022] Open
Abstract
Brain disease gene identification is critical for revealing the biological mechanism and developing drugs for brain diseases. To enhance the identification of brain disease genes, similarity-based computational methods, especially network-based methods, have been adopted for narrowing down the searching space. However, these network-based methods only use molecular networks, ignoring brain connectome data, which have been widely used in many brain-related studies. In our study, we propose a novel framework, named brainMI, for integrating brain connectome data and molecular-based gene association networks to predict brain disease genes. For the consistent representation of molecular-based network data and brain connectome data, brainMI first constructs a novel gene network, called brain functional connectivity (BFC)-based gene network, based on resting-state functional magnetic resonance imaging data and brain region-specific gene expression data. Then, a multiple network integration method is proposed to learn low-dimensional features of genes by integrating the BFC-based gene network and existing protein-protein interaction networks. Finally, these features are utilized to predict brain disease genes based on a support vector machine-based model. We evaluate brainMI on four brain diseases, including Alzheimer's disease, Parkinson's disease, major depressive disorder and autism. brainMI achieves of 0.761, 0.729, 0.728 and 0.744 using the BFC-based gene network alone and enhances the molecular network-based performance by 6.3% on average. In addition, the results show that brainMI achieves higher performance in predicting brain disease genes compared to the existing three state-of-the-art methods.
Collapse
Affiliation(s)
- Wei Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| | - Ruijiang Han
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| | - Menghan Zhang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| | - Yuxian Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| | - Tao Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| | - Yongtian Wang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| | - Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China.,Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, 710072, China
| |
Collapse
|
36
|
Dalvie S, Chatzinakos C, Al Zoubi O, Georgiadis F, Lancashire L, Daskalakis NP. From genetics to systems biology of stress-related mental disorders. Neurobiol Stress 2021; 15:100393. [PMID: 34584908 PMCID: PMC8456113 DOI: 10.1016/j.ynstr.2021.100393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/22/2021] [Accepted: 09/08/2021] [Indexed: 01/20/2023] Open
Abstract
Many individuals will be exposed to some form of traumatic stress in their lifetime which, in turn, increases the likelihood of developing stress-related disorders such as post-traumatic stress disorder (PTSD), major depressive disorder (MDD) and anxiety disorders (ANX). The development of these disorders is also influenced by genetics and have heritability estimates ranging between ∼30 and 70%. In this review, we provide an overview of the findings of genome-wide association studies for PTSD, depression and ANX, and we observe a clear genetic overlap between these three diagnostic categories. We go on to highlight the results from transcriptomic and epigenomic studies, and, given the multifactorial nature of stress-related disorders, we provide an overview of the gene-environment studies that have been conducted to date. Finally, we discuss systems biology approaches that are now seeing wider utility in determining a more holistic view of these complex disorders.
Collapse
Affiliation(s)
- Shareefa Dalvie
- South African Medical Research Council (SAMRC), Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council (SAMRC), Unit on Child & Adolescent Health, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Chris Chatzinakos
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Obada Al Zoubi
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Foivos Georgiadis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, USA
| | | | - Lee Lancashire
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Data Science, Cohen Veterans Bioscience, New York, USA
| | - Nikolaos P. Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, USA
| |
Collapse
|
37
|
Lin YS, Wang CC, Chen CY. GWAS Meta-Analysis Reveals Shared Genes and Biological Pathways between Major Depressive Disorder and Insomnia. Genes (Basel) 2021; 12:1506. [PMID: 34680902 PMCID: PMC8536096 DOI: 10.3390/genes12101506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 11/27/2022] Open
Abstract
Major depressive disorder (MDD) is one of the most prevalent and disabling mental disorders worldwide. Among the symptoms of MDD, sleep disturbance such as insomnia is prominent, and the first reason patients may seek professional help. However, the underlying pathophysiology of this comorbidity is still elusive. Recently, genome-wide association studies (GWAS) have begun to unveil the genetic background of several psychiatric disorders, including MDD and insomnia. Identifying the shared genomic risk loci between comorbid psychiatric disorders could be a valuable strategy to understanding their comorbidity. This study seeks to identify the shared genes and biological pathways between MDD and insomnia based on their shared genetic variants. First, we performed a meta-analysis based on the GWAS summary statistics of MDD and insomnia obtained from Psychiatric Genomics Consortium and UK Biobank, respectively. Next, we associated shared genetic variants to genes using two gene mapping strategies: (a) positional mapping based on genomic proximity and (b) expression quantitative trait loci (eQTL) mapping based on gene expression linkage across multiple tissues. As a result, a total of 719 shared genes were identified. Over half (51%) of them are protein-coding genes. Functional enrichment analysis shows that the most enriched biological pathways are related to epigenetic modification, sensory perception, and immunologic signatures. We also identified druggable targets using a network approach. Together, these results may provide insights into understanding the genetic predisposition and underlying biological pathways of comorbid MDD and insomnia symptoms.
Collapse
Affiliation(s)
- Yi-Sian Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.L.); (C.-C.W.)
| | - Chia-Chun Wang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.L.); (C.-C.W.)
| | - Cho-Yi Chen
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.L.); (C.-C.W.)
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
38
|
Gerring ZF, Gamazon ER, White A, Derks EM. Integrative Network-Based Analysis Reveals Gene Networks and Novel Drug Repositioning Candidates for Alzheimer Disease. NEUROLOGY-GENETICS 2021; 7:e622. [PMID: 34532569 PMCID: PMC8441674 DOI: 10.1212/nxg.0000000000000622] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 07/13/2021] [Indexed: 12/30/2022]
Abstract
Background and Objectives To integrate genome-wide association study data with tissue-specific gene expression information to identify coexpression networks, biological pathways, and drug repositioning candidates for Alzheimer disease. Methods We integrated genome-wide association summary statistics for Alzheimer disease with tissue-specific gene coexpression networks from brain tissue samples in the Genotype-Tissue Expression study. We identified gene coexpression networks enriched with genetic signals for Alzheimer disease and characterized the associated networks using biological pathway analysis. The disease-implicated modules were subsequently used as a molecular substrate for a computational drug repositioning analysis, in which we (1) imputed genetically regulated gene expression within Alzheimer disease implicated modules; (2) integrated the imputed gene expression levels with drug-gene signatures from the connectivity map to identify compounds that normalize dysregulated gene expression underlying Alzheimer disease; and (3) prioritized drug compounds and mechanisms of action based on the extent to which they normalize dysregulated expression signatures. Results Genetic factors for Alzheimer disease are enriched in brain gene coexpression networks involved in the immune response. Computational drug repositioning analyses of expression changes within the disease-associated networks retrieved known Alzheimer disease drugs (e.g., memantine) as well as biologically meaningful drug categories (e.g., glutamate receptor antagonists). Discussion Our results improve the biological interpretation of genetic data for Alzheimer disease and provide a list of potential antidementia drug repositioning candidates for which the efficacy should be investigated in functional validation studies.
Collapse
Affiliation(s)
- Zachary F Gerring
- Translational Neurogenomics Laboratory (Z.F.G., E.M.D.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Division of Genetic Medicine (E.R.G.), Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN; and Cellular and Molecular Neurodegeneration (A.W.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Eric R Gamazon
- Translational Neurogenomics Laboratory (Z.F.G., E.M.D.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Division of Genetic Medicine (E.R.G.), Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN; and Cellular and Molecular Neurodegeneration (A.W.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anthony White
- Translational Neurogenomics Laboratory (Z.F.G., E.M.D.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Division of Genetic Medicine (E.R.G.), Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN; and Cellular and Molecular Neurodegeneration (A.W.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Eske M Derks
- Translational Neurogenomics Laboratory (Z.F.G., E.M.D.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; Division of Genetic Medicine (E.R.G.), Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN; and Cellular and Molecular Neurodegeneration (A.W.), QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
Lukoyanov N, Watanabe H, Carvalho LS, Kononenko O, Sarkisyan D, Zhang M, Andersen MS, Lukoyanova EA, Galatenko V, Tonevitsky A, Bazov I, Iakovleva T, Schouenborg J, Bakalkin G. Left-right side-specific endocrine signaling complements neural pathways to mediate acute asymmetric effects of brain injury. eLife 2021; 10:e65247. [PMID: 34372969 PMCID: PMC8354641 DOI: 10.7554/elife.65247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Brain injuries can interrupt descending neural pathways that convey motor commands from the cortex to spinal motoneurons. Here, we demonstrate that a unilateral injury of the hindlimb sensorimotor cortex of rats with completely transected thoracic spinal cord produces hindlimb postural asymmetry with contralateral flexion and asymmetric hindlimb withdrawal reflexes within 3 hr, as well as asymmetry in gene expression patterns in the lumbar spinal cord. The injury-induced postural effects were abolished by hypophysectomy and were mimicked by transfusion of serum from animals with brain injury. Administration of the pituitary neurohormones β-endorphin or Arg-vasopressin-induced side-specific hindlimb responses in naive animals, while antagonists of the opioid and vasopressin receptors blocked hindlimb postural asymmetry in rats with brain injury. Thus, in addition to the well-established involvement of motor pathways descending from the brain to spinal circuits, the side-specific humoral signaling may also add to postural and reflex asymmetries seen after brain injury.
Collapse
Affiliation(s)
- Nikolay Lukoyanov
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Instituto de Investigação e Inovação em Saúde, Instituto de Biologia Molecular e CelularPortoPortugal
| | - Hiroyuki Watanabe
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Liliana S Carvalho
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Instituto de Investigação e Inovação em Saúde, Instituto de Biologia Molecular e CelularPortoPortugal
| | - Olga Kononenko
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Daniil Sarkisyan
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Mengliang Zhang
- Neuronano Research Center, Department of Experimental Medical Science, Lund UniversityLundSweden
- Department of Molecular Medicine, University of Southern DenmarkOdenseDenmark
| | | | - Elena A Lukoyanova
- Departamento de Biomedicina da Faculdade de Medicina da Universidade do Porto, Instituto de Investigação e Inovação em Saúde, Instituto de Biologia Molecular e CelularPortoPortugal
| | - Vladimir Galatenko
- Faculty of Mechanics and Mathematics, Lomonosov Moscow State UniversityMoscowRussian Federation
| | - Alex Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of EconomicsMoscowRussian Federation
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry RASMoscowRussian Federation
| | - Igor Bazov
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Tatiana Iakovleva
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| | - Jens Schouenborg
- Neuronano Research Center, Department of Experimental Medical Science, Lund UniversityLundSweden
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, Uppsala UniversityUppsalaSweden
| |
Collapse
|
40
|
Pisanu C, Congiu D, Severino G, Ardau R, Chillotti C, Del Zompo M, Baune BT, Squassina A. Investigation of genetic loci shared between bipolar disorder and risk-taking propensity: potential implications for pharmacological interventions. Neuropsychopharmacology 2021; 46:1680-1692. [PMID: 34035470 PMCID: PMC8280111 DOI: 10.1038/s41386-021-01045-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 11/09/2022]
Abstract
Patients with bipolar disorder (BD) often show increased risk-taking propensity, which may contribute to poor clinical outcome. While these two phenotypes are genetically correlated, there is scarce knowledge on the shared genetic determinants. Using GWAS datasets on BD (41,917 BD cases and 371,549 controls) and risk-taking (n = 466,571), we dissected shared genetic determinants using conjunctional false discovery rate (conjFDR) and local genetic covariance analysis. We investigated specificity of identified targets using GWAS datasets on schizophrenia (SCZ) and attention-deficit hyperactivity disorder (ADHD). The putative functional role of identified targets was evaluated using different tools and GTEx v. 8. Target druggability was evaluated using DGIdb and enrichment for drug targets with genome for REPositioning drugs (GREP). Among 102 loci shared between BD and risk-taking, 87% showed the same direction of effect. Sixty-two were specifically shared between risk-taking propensity and BD, while the others were also shared between risk-taking propensity and either SCZ or ADHD. By leveraging pleiotropic enrichment, we reported 15 novel and specific loci associated with BD and 22 with risk-taking. Among cross-disorder genes, CACNA1C (a known target of calcium channel blockers) was significantly associated with risk-taking propensity and both BD and SCZ using conjFDR (p = 0.001 for both) as well as local genetic covariance analysis, and predicted to be differentially expressed in the cerebellar hemisphere in an eQTL-informed gene-based analysis (BD, Z = 7.48, p = 3.8E-14; risk-taking: Z = 4.66, p = 1.6E-06). We reported for the first time shared genetic determinants between BD and risk-taking propensity. Further investigation into calcium channel blockers or development of innovative ligands of calcium channels might form the basis for innovative pharmacotherapy in patients with BD with increased risk-taking propensity.
Collapse
Affiliation(s)
- Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Donatella Congiu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Giovanni Severino
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Raffaella Ardau
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Caterina Chillotti
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Maria Del Zompo
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
- Unit of Clinical Pharmacology of the University Hospital of Cagliari, Cagliari, Italy
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
41
|
Mulvey B, Dougherty JD. Transcriptional-regulatory convergence across functional MDD risk variants identified by massively parallel reporter assays. Transl Psychiatry 2021; 11:403. [PMID: 34294677 PMCID: PMC8298436 DOI: 10.1038/s41398-021-01493-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Family and population studies indicate clear heritability of major depressive disorder (MDD), though its underlying biology remains unclear. The majority of single-nucleotide polymorphism (SNP) linkage blocks associated with MDD by genome-wide association studies (GWASes) are believed to alter transcriptional regulators (e.g., enhancers, promoters) based on enrichment of marks correlated with these functions. A key to understanding MDD pathophysiology will be elucidation of which SNPs are functional and how such functional variants biologically converge to elicit the disease. Furthermore, retinoids can elicit MDD in patients and promote depressive-like behaviors in rodent models, acting via a regulatory system of retinoid receptor transcription factors (TFs). We therefore sought to simultaneously identify functional genetic variants and assess retinoid pathway regulation of MDD risk loci. Using Massively Parallel Reporter Assays (MPRAs), we functionally screened over 1000 SNPs prioritized from 39 neuropsychiatric trait/disease GWAS loci, selecting SNPs based on overlap with predicted regulatory features-including expression quantitative trait loci (eQTL) and histone marks-from human brains and cell cultures. We identified >100 SNPs with allelic effects on expression in a retinoid-responsive model system. Functional SNPs were enriched for binding sequences of retinoic acid-receptive transcription factors (TFs), with additional allelic differences unmasked by treatment with all-trans retinoic acid (ATRA). Finally, motifs overrepresented across functional SNPs corresponded to TFs highly specific to serotonergic neurons, suggesting an in vivo site of action. Our application of MPRAs to screen MDD-associated SNPs suggests a shared transcriptional-regulatory program across loci, a component of which is unmasked by retinoids.
Collapse
Affiliation(s)
- Bernard Mulvey
- Departments of Genetics and Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Joseph D Dougherty
- Departments of Genetics and Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
42
|
Chen J, Dong G, Song L, Zhao X, Cao J, Luo X, Feng J, Zhao XM. Integration of Multimodal Data for Deciphering Brain Disorders. Annu Rev Biomed Data Sci 2021; 4:43-56. [PMID: 34465176 DOI: 10.1146/annurev-biodatasci-092820-020354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The accumulation of vast amounts of multimodal data for the human brain, in both normal and disease conditions, has provided unprecedented opportunities for understanding why and how brain disorders arise. Compared with traditional analyses of single datasets, the integration of multimodal datasets covering different types of data (i.e., genomics, transcriptomics, imaging, etc.) has shed light on the mechanisms underlying brain disorders in greater detail across both the microscopic and macroscopic levels. In this review, we first briefly introduce the popular large datasets for the brain. Then, we discuss in detail how integration of multimodal human brain datasets can reveal the genetic predispositions and the abnormal molecular pathways of brain disorders. Finally, we present an outlook on how future data integration efforts may advance the diagnosis and treatment of brain disorders.
Collapse
Affiliation(s)
- Jingqi Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; , .,MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, Ministry of Education, Shanghai 200433, China.,Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200433, China
| | - Guiying Dong
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; ,
| | - Liting Song
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; ,
| | - Xingzhong Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; ,
| | - Jixin Cao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; ,
| | - Xiaohui Luo
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; ,
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; , .,MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, Ministry of Education, Shanghai 200433, China.,Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200433, China.,Department of Computer Science, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Xing-Ming Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; , .,MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, Ministry of Education, Shanghai 200433, China.,Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200433, China
| |
Collapse
|
43
|
Multilayer modelling of the human transcriptome and biological mechanisms of complex diseases and traits. NPJ Syst Biol Appl 2021; 7:24. [PMID: 34045472 PMCID: PMC8160250 DOI: 10.1038/s41540-021-00186-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/28/2021] [Indexed: 01/03/2023] Open
Abstract
Here, we performed a comprehensive intra-tissue and inter-tissue multilayer network analysis of the human transcriptome. We generated an atlas of communities in gene co-expression networks in 49 tissues (GTEx v8), evaluated their tissue specificity, and investigated their methodological implications. UMAP embeddings of gene expression from the communities (representing nearly 18% of all genes) robustly identified biologically-meaningful clusters. Notably, new gene expression data can be embedded into our algorithmically derived models to accelerate discoveries in high-dimensional molecular datasets and downstream diagnostic or prognostic applications. We demonstrate the generalisability of our approach through systematic testing in external genomic and transcriptomic datasets. Methodologically, prioritisation of the communities in a transcriptome-wide association study of the biomarker C-reactive protein (CRP) in 361,194 individuals in the UK Biobank identified genetically-determined expression changes associated with CRP and led to considerably improved performance. Furthermore, a deep learning framework applied to the communities in nearly 11,000 tumors profiled by The Cancer Genome Atlas across 33 different cancer types learned biologically-meaningful latent spaces, representing metastasis (p < 2.2 × 10−16) and stemness (p < 2.2 × 10−16). Our study provides a rich genomic resource to catalyse research into inter-tissue regulatory mechanisms, and their downstream consequences on human disease.
Collapse
|
44
|
Gerring ZF, Vargas AM, Gamazon ER, Derks EM. An integrative systems-based analysis of substance use: eQTL-informed gene-based tests, gene networks, and biological mechanisms. Am J Med Genet B Neuropsychiatr Genet 2021; 186:162-172. [PMID: 33369091 PMCID: PMC8137546 DOI: 10.1002/ajmg.b.32829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/17/2020] [Accepted: 11/27/2020] [Indexed: 01/03/2023]
Abstract
Genome-wide association studies have identified multiple genetic risk factors underlying susceptibility to substance use, however, the functional genes and biological mechanisms remain poorly understood. The discovery and characterization of risk genes can be facilitated by the integration of genome-wide association data and gene expression data across biologically relevant tissues and/or cell types to identify genes whose expression is altered by DNA sequence variation (expression quantitative trait loci; eQTLs). The integration of gene expression data can be extended to the study of genetic co-expression, under the biologically valid assumption that genes form co-expression networks to influence the manifestation of a disease or trait. Here, we integrate genome-wide association data with gene expression data from 13 brain tissues to identify candidate risk genes for 8 substance use phenotypes. We then test for the enrichment of candidate risk genes within tissue-specific gene co-expression networks to identify modules (or groups) of functionally related genes whose dysregulation is associated with variation in substance use. We identified eight gene modules in brain that were enriched with gene-based association signals for substance use phenotypes. For example, a single module of 40 co-expressed genes was enriched with gene-based associations for drinks per week and biological pathways involved in GABA synthesis, release, reuptake and degradation. Our study demonstrates the utility of eQTL and gene co-expression analysis to uncover novel biological mechanisms for substance use traits.
Collapse
Affiliation(s)
- Zachary F Gerring
- Translational Neurogenomics Laboratory; QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Angela Mina Vargas
- Translational Neurogenomics Laboratory; QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA,Clare Hall, University of Cambridge, Cambridge, United Kingdom
| | - Eske M Derks
- Translational Neurogenomics Laboratory; QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
45
|
Silberstein M, Nesbit N, Cai J, Lee PH. Pathway analysis for genome-wide genetic variation data: Analytic principles, latest developments, and new opportunities. J Genet Genomics 2021; 48:173-183. [PMID: 33896739 PMCID: PMC8286309 DOI: 10.1016/j.jgg.2021.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/23/2022]
Abstract
Pathway analysis, also known as gene-set enrichment analysis, is a multilocus analytic strategy that integrates a priori, biological knowledge into the statistical analysis of high-throughput genetics data. Originally developed for the studies of gene expression data, it has become a powerful analytic procedure for in-depth mining of genome-wide genetic variation data. Astonishing discoveries were made in the past years, uncovering genes and biological mechanisms underlying common and complex disorders. However, as massive amounts of diverse functional genomics data accrue, there is a pressing need for newer generations of pathway analysis methods that can utilize multiple layers of high-throughput genomics data. In this review, we provide an intellectual foundation of this powerful analytic strategy, as well as an update of the state-of-the-art in recent method developments. The goal of this review is threefold: (1) introduce the motivation and basic steps of pathway analysis for genome-wide genetic variation data; (2) review the merits and the shortcomings of classic and newly emerging integrative pathway analysis tools; and (3) discuss remaining challenges and future directions for further method developments.
Collapse
Affiliation(s)
- Micah Silberstein
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nicholas Nesbit
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jacquelyn Cai
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Phil H Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
46
|
Dall’Aglio L, Lewis CM, Pain O. Delineating the Genetic Component of Gene Expression in Major Depression. Biol Psychiatry 2021; 89:627-636. [PMID: 33279206 PMCID: PMC7886308 DOI: 10.1016/j.biopsych.2020.09.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/17/2020] [Accepted: 09/08/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Major depression (MD) is determined by a multitude of factors including genetic risk variants that regulate gene expression. We examined the genetic component of gene expression in MD by performing a transcriptome-wide association study (TWAS), inferring gene expression-trait relationships from genetic, transcriptomic, and phenotypic information. METHODS Genes differentially expressed in depression were identified with the TWAS FUSION method, based on summary statistics from the largest genome-wide association analysis of MD (n = 135,458 cases, n = 344,901 controls) and gene expression levels from 21 tissue datasets (brain; blood; thyroid, adrenal, and pituitary glands). Follow-up analyses were performed to extensively characterize the identified associations: colocalization, conditional, and fine-mapping analyses together with TWAS-based pathway investigations. RESULTS Transcriptome-wide significant differences between cases and controls were found at 94 genes, approximately half of which were novel. Of the 94 significant genes, 6 represented strong, colocalized, and potentially causal associations with depression. Such high-confidence associations include NEGR1, CTC-467M3.3, TMEM106B, LRFN5, ESR2, and PROX2. Lastly, TWAS-based enrichment analysis highlighted dysregulation of gene sets for, among others, neuronal and synaptic processes. CONCLUSIONS This study sheds further light on the genetic component of gene expression in depression by characterizing the identified associations, unraveling novel risk genes, and determining which associations are congruent with a causal model. These findings can be used as a resource for prioritizing and designing subsequent functional studies of MD.
Collapse
Affiliation(s)
- Lorenza Dall’Aglio
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom,Department of Child and Adolescent Psychiatry, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands,Generation R Study Group, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Cathryn M. Lewis
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom,Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Oliver Pain
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
47
|
Gerring ZF. Dissecting Genetically Regulated Gene Expression in Major Depression. Biol Psychiatry 2021; 89:e31-e33. [PMID: 33594984 DOI: 10.1016/j.biopsych.2020.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Zachary F Gerring
- Translational Neurogenomics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
48
|
Gerring ZF, Mina-Vargas A, Gamazon ER, Derks EM. E-MAGMA: an eQTL-informed method to identify risk genes using genome-wide association study summary statistics. Bioinformatics 2021; 37:2245-2249. [PMID: 33624746 PMCID: PMC8388029 DOI: 10.1093/bioinformatics/btab115] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/02/2021] [Accepted: 02/18/2021] [Indexed: 01/29/2023] Open
Abstract
MOTIVATION Genome-wide association studies have successfully identified multiple independent genetic loci that harbour variants associated with human traits and diseases, but the exact causal genes are largely unknown. Common genetic risk variants are enriched in non-protein-coding regions of the genome and often affect gene expression (expression quantitative trait loci, eQTL) in a tissue-specific manner. To address this challenge, we developed a methodological framework, E-MAGMA, which converts genome-wide association summary statistics into gene-level statistics by assigning risk variants to their putative genes based on tissue-specific eQTL information. RESULTS We compared E-MAGMA to three eQTL informed gene-based approaches using simulated phenotype data. Phenotypes were simulated based on eQTL reference data using GCTA for all genes with at least one eQTL at chromosome 1. We performed 10 simulations per gene. The eQTL-h2 (i.e., the proportion of variation explained by the eQTLs) was set at 1%, 2%, and 5%. We found E-MAGMA outperforms other gene-based approaches across a range of simulated parameters (e.g. the number of identified causal genes). When applied to genome-wide association summary statistics for five neuropsychiatric disorders, E-MAGMA identified more putative candidate causal genes compared to other eQTL-based approaches. By integrating tissue-specific eQTL information, these results show E-MAGMA will help to identify novel candidate causal genes from genome-wide association summary statistics and thereby improve the understanding of the biological basis of complex disorders. AVAILABILITY A tutorial and input files are made available in a github repository: https://github.com/eskederks/eMAGMA-tutorial. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zachary F Gerring
- Translational Neurogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Angela Mina-Vargas
- Translational Neurogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.,Clare Hall, University of Cambridge, Cambridge, United Kingdom
| | - Eske M Derks
- Translational Neurogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
49
|
Li HJ, Su X, Zhang LW, Zhang CY, Wang L, Li WQ, Yang YF, Lv LX, Li M, Xiao X. Transcriptomic analyses of humans and mice provide insights into depression. Zool Res 2021; 41:632-643. [PMID: 32987454 PMCID: PMC7671914 DOI: 10.24272/j.issn.2095-8137.2020.174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Accumulating studies have been conducted to identify risk genes and relevant biological mechanisms underlying major depressive disorder (MDD). In particular, transcriptomic analyses in brain regions engaged in cognitive and emotional processes, e.g., the dorsolateral prefrontal cortex (DLPFC), have provided essential insights. Based on three independent DLPFC RNA-seq datasets of 79 MDD patients and 75 healthy controls, we performed differential expression analyses using two alternative approaches for cross-validation. We also conducted transcriptomic analyses in mice undergoing chronic variable stress (CVS) and chronic social defeat stress (CSDS). We identified 12 differentially expressed genes (DEGs) through both analytical methods in MDD patients, the majority of which were also dysregulated in stressed mice. Notably, the mRNA level of the immediate early gene FOS ( Fos proto-oncogene) was significantly decreased in both MDD patients and CVS-exposed mice, and CSDS-susceptible mice exhibited a greater reduction in Fos expression compared to resilient mice. These findings suggest the potential key roles of this gene in the pathogenesis of MDD related to stress exposure. Altered transcriptomes in the DLPFC of MDD patients might be, at least partially, the result of stress exposure, supporting that stress is a primary risk factor for MDD.
Collapse
Affiliation(s)
- Hui-Juan 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, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Xi Su
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Lu-Wen Zhang
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Chu-Yi Zhang
- 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, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Lu Wang
- 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, Yunnan 650223, China
| | - Wen-Qiang Li
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Yong-Feng Yang
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Lu-Xian Lv
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Province People's Hospital, Zhengzhou, Henan 450003, 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, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| | - 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, Yunnan 650223, China. E-mail:
| |
Collapse
|
50
|
Yang A, Chen J, Zhao XM. nMAGMA: a network-enhanced method for inferring risk genes from GWAS summary statistics and its application to schizophrenia. Brief Bioinform 2020; 22:5998843. [PMID: 33230537 DOI: 10.1093/bib/bbaa298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/21/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022] Open
Abstract
MOTIVATION Annotating genetic variants from summary statistics of genome-wide association studies (GWAS) is crucial for predicting risk genes of various disorders. The multimarker analysis of genomic annotation (MAGMA) is one of the most popular tools for this purpose, where MAGMA aggregates signals of single nucleotide polymorphisms (SNPs) to their nearby genes. In biology, SNPs may also affect genes that are far away in the genome, thus missed by MAGMA. Although different upgrades of MAGMA have been proposed to extend gene-wise variant annotations with more information (e.g. Hi-C or eQTL), the regulatory relationships among genes and the tissue specificity of signals have not been taken into account. RESULTS We propose a new approach, namely network-enhanced MAGMA (nMAGMA), for gene-wise annotation of variants from GWAS summary statistics. Compared with MAGMA and H-MAGMA, nMAGMA significantly extends the lists of genes that can be annotated to SNPs by integrating local signals, long-range regulation signals (i.e. interactions between distal DNA elements), and tissue-specific gene networks. When applied to schizophrenia (SCZ), nMAGMA is able to detect more risk genes (217% more than MAGMA and 57% more than H-MAGMA) that are involved in SCZ compared with MAGMA and H-MAGMA, and more of nMAGMA results can be validated with known SCZ risk genes. Some disease-related functions (e.g. the ATPase pathway in Cortex) are also uncovered in nMAGMA but not in MAGMA or H-MAGMA. Moreover, nMAGMA provides tissue-specific risk signals, which are useful for understanding disorders with multitissue origins.
Collapse
Affiliation(s)
- Anyi Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China
| | - Jingqi Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China
| | | |
Collapse
|