1
|
Laffranchi M, Paraboschi EM, Bianchetto-Aguilera F, Tamassia N, Gasperini S, Gardiman E, Piserà A, Del Prete A, Invernizzi P, Gismondi A, Mantovani A, Cassatella MA, Asselta R, Sozzani S. Neutrophils restricted contribution of CCRL2 genetic variants to COVID-19 severity. Heliyon 2025; 11:e41267. [PMID: 39811276 PMCID: PMC11731188 DOI: 10.1016/j.heliyon.2024.e41267] [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: 07/23/2024] [Revised: 11/12/2024] [Accepted: 12/14/2024] [Indexed: 01/02/2025] Open
Abstract
The 3p21.31 locus is the most robust genomic region associated with COVID-19 severity. This locus contains a main chemokine receptor (CKR) cluster. We tested expression quantitative trait loci (eQTL) targeting the 3p21.31 CKR cluster linked to COVID-19 hospitalization in Europeans from the COVID-19 HGI meta-analysis. Among these, CCRL2, a key regulator of neutrophil trafficking, was targeted by neutrophil-restricted eQTLs. We confirmed these eQTLs in an Italian COVID-19 cohort. Haplotype analysis revealed a link between an increased CCRL2 expression and COVID-19 severity and hospitalization. By the exposure of neutrophils to a TLR8 ligand, reflecting a viral infection, we revealed specific chromatin domains within the 3p21.31 locus exclusive to neutrophils. In addition, the identified variants mapped within these regions altered the binding motif of neutrophils-expressed transcription factors. These results support that CCRL2 eQTL variants contribute to the risk of severe COVID-19 by selectively affecting neutrophil functions.
Collapse
Affiliation(s)
- Mattia Laffranchi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Elvezia Maria Paraboschi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Arianna Piserà
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo Dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Marco A. Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, 37134, Verona, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
2
|
Yao S, Harder A, Darki F, Chang YW, Li A, Nikouei K, Volpe G, Lundström JN, Zeng J, Wray NR, Lu Y, Sullivan PF, Hjerling-Leffler J. Connecting genomic results for psychiatric disorders to human brain cell types and regions reveals convergence with functional connectivity. Nat Commun 2025; 16:395. [PMID: 39755698 DOI: 10.1038/s41467-024-55611-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/17/2024] [Indexed: 01/06/2025] Open
Abstract
Identifying cell types and brain regions critical for psychiatric disorders and brain traits is essential for targeted neurobiological research. By integrating genomic insights from genome-wide association studies with a comprehensive single-cell transcriptomic atlas of the adult human brain, we prioritized specific neuronal clusters significantly enriched for the SNP-heritabilities for schizophrenia, bipolar disorder, and major depressive disorder along with intelligence, education, and neuroticism. Extrapolation of cell-type results to brain regions reveals the whole-brain impact of schizophrenia genetic risk, with subregions in the hippocampus and amygdala exhibiting the most significant enrichment of SNP-heritability. Using functional MRI connectivity, we further confirmed the significance of the central and lateral amygdala, hippocampal body, and prefrontal cortex in distinguishing schizophrenia cases from controls. Our findings underscore the value of single-cell transcriptomics in understanding the polygenicity of psychiatric disorders and suggest a promising alignment of genomic, transcriptomic, and brain imaging modalities for identifying common biological targets.
Collapse
Affiliation(s)
- Shuyang Yao
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Arvid Harder
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fahimeh Darki
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yu-Wei Chang
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Ang Li
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Kasra Nikouei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Giovanni Volpe
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Johan N Lundström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Jian Zeng
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Kim S, Ko H, Myung W, Yoon J, Kim K, Jung SH, Shim I, Cha S, Kim B, Kang JM, Park WY, Natarajan P, Do R, Won HH. Association between genetically predicted leisure and social activities and cardiovascular disease and other health outcomes. NATURE CARDIOVASCULAR RESEARCH 2025:10.1038/s44161-024-00581-2. [PMID: 39747453 DOI: 10.1038/s44161-024-00581-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 11/01/2024] [Indexed: 01/04/2025]
Abstract
Participation in leisure and social activities (LSA) is associated with better health outcomes and lower mortality1-3. Previous observational studies demonstrated a relationship between engagement in LSA and both mental and physical health4,5. Although several studies6 examined the association between LSA and health outcomes, including cardiovascular disease, their possible causal relationship has not been studied. In this study, we investigated the causal relationship between LSA and various health outcomes, including cardiovascular disease, using data from genome-wide association study summary statistics (sample size = 63,926-1,557,411) and a Mendelian randomization approach. Genetically predicted LSA were associated with a reduced risk of several health outcomes, including coronary artery disease and coronary atherosclerosis. Mediation analysis indicated that these effects were partly mediated by modifiable risk factors, such as body mass index, smoking and lipid levels. These findings highlight the importance of LSA in disease prevention and health promotion.
Collapse
Affiliation(s)
- Soyeon Kim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hyunwoong Ko
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea.
| | - Joohyun Yoon
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kiwon Kim
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Sang-Hyuk Jung
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Injeong Shim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Soojin Cha
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Beomsu Kim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Jae Myeong Kang
- Department of Psychiatry, Gil Medical Center, Gachon University, Incheon, South Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Pradeep Natarajan
- Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ron Do
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hong-Hee Won
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea.
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
4
|
Zhang YP, Wang S, Yao TX, Zou XL, Li S, Chen L, Zhang XB, Chen JY, Zeng Y, Zhang L. IL6 and WNK2 polymorphisms and prognosis in patients with intracerebral hemorrhage receiving edaravone. J Stroke Cerebrovasc Dis 2025; 34:108095. [PMID: 39433142 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/03/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE Edaravone is utilized in intra-cerebral hemorrhage (ICH) patients for years, while personalized variances were observed in clinic. To explore the precision medicine strategy for Edaravone, this study investigated the association between genetic polymorphisms and Edaravone efficacy in ICH patients. METHODS We genotyped 7 SNPs in 4 potential genes, including COL4A2, TNF, WNK2 and IL6, from the peripheral blood of 217 ICH patients with or without Edaravone utilizations. PLINK and SPSS were utilized for association tests, Student's t tests, Mann-Whitney U tests and Chi-square tests. RESULTS Rs1800796 (C>G) in IL6 (OR: 0.41, 95 % CI: 0.18-0.94, p-value = 0.03) and rs16936752 (G>T) in WNK2 (OR: 0.28, 95 % CI: 0.09-0.88, p-value = 0.02) were found to be significantly associated with Edaravone efficacy. The association of rs1800796 and rs16936752 were found to be different in patients with or without smoking habit, alcohol drinking habit, hypertension history and hyperlipemia history were found to be different. Both of these two SNPs were found to be associated with the concentration of high-density lipoprotein cholesterol (HDL) in ICH patients. CONCLUSIONS The IL6 rs1800796 and the WNK2 rs16936752 could be useful biomarkers for prognosis prediction during Edaravone treatment. These SNPs should be considered in the personalized medicine strategy for Edaravone in the future.
Collapse
Affiliation(s)
- Yu-Peng Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Sai Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Tian-Xing Yao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xue-Lun Zou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Si Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lei Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiang-Bin Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jun-You Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yi Zeng
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Le Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
5
|
Jasper EA, Hellwege JN, Greene CA, Edwards TL, Velez Edwards DR. Genomic insights into gestational weight gain uncover tissue-specific mechanisms and pathways. NPJ WOMEN'S HEALTH 2024; 2:42. [PMID: 39651376 PMCID: PMC11624131 DOI: 10.1038/s44294-024-00035-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/18/2024] [Indexed: 12/11/2024]
Abstract
Gestational weight gain (GWG) is linked to adverse outcomes in pregnant persons and offspring. The Early Growth Genetics Consortium previously identified genetic variants contributing to GWG from fetal and maternal genomes. However, their biologic mechanisms and tissue-specificity are unknown. We evaluated the association between genetically predicted gene expression in relevant maternal (subcutaneous and visceral adipose, breast, uterus, and whole blood) tissues from GTEx (v7) and fetal (placenta) tissue and early, late, and total GWG using S-PrediXcan. We tested for pathway enrichment using the GENE2FUNC module from Functional Mapping and Annotation of Genome-Wide Association Studies. After Bonferroni correction, we found no associations between maternal or fetal gene expression and GWG. Among nominally significant (P < 0.05) maternal genes, there was enrichment of several biological pathways, including metabolic processes, secretion, and intracellular transport, that varied across pregnancy. These results indicate the likely influence of diverse pathways, varying by tissue and weeks of gestation, on GWG.
Collapse
Affiliation(s)
- Elizabeth A. Jasper
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN USA
- Center for Precision Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN USA
| | - Jacklyn N. Hellwege
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Catherine A. Greene
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN USA
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN USA
| | - Todd L. Edwards
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN USA
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Digna R. Velez Edwards
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN USA
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN USA
- Vanderbilt Epidemiology Center, Vanderbilt University, Nashville, TN USA
| |
Collapse
|
6
|
Townsend HA, Rosenberger KJ, Vanderlinden LA, Inamo J, Zhang F. Evaluating methods for integrating single-cell data and genetics to understand inflammatory disease complexity. Front Immunol 2024; 15:1454263. [PMID: 39703500 PMCID: PMC11655331 DOI: 10.3389/fimmu.2024.1454263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024] Open
Abstract
Background Understanding genetic underpinnings of immune-mediated inflammatory diseases is crucial to improve treatments. Single-cell RNA sequencing (scRNA-seq) identifies cell states expanded in disease, but often overlooks genetic causality due to cost and small genotyping cohorts. Conversely, large genome-wide association studies (GWAS) are commonly accessible. Methods We present a 3-step robust benchmarking analysis of integrating GWAS and scRNA-seq to identify genetically relevant cell states and genes in inflammatory diseases. First, we applied and compared the results of three recent algorithms, based on pathways (scGWAS), single-cell disease scores (scDRS), or both (scPagwas), according to accuracy/sensitivity and interpretability. While previous studies focused on coarse cell types, we used disease-specific, fine-grained single-cell atlases (183,742 and 228,211 cells) and GWAS data (Ns of 97,173 and 45,975) for rheumatoid arthritis (RA) and ulcerative colitis (UC). Second, given the lack of scRNA-seq for many diseases with GWAS, we further tested the tools' resolution limits by differentiating between similar diseases with only one fine-grained scRNA-seq atlas. Lastly, we provide a novel evaluation of noncoding SNP incorporation methods by testing which enabled the highest sensitivity/accuracy of known cell-state calls. Results We first found that single-cell based tools scDRS and scPagwas called superior numbers of supported cell states that were overlooked by scGWAS. While scGWAS and scPagwas were advantageous for gene exploration, scDRS effectively accounted for batch effect and captured cellular heterogeneity of disease-relevance without single-cell genotyping. For noncoding SNP integration, we found a key trade-off between statistical power and confidence with positional (e.g. MAGMA) and non-positional approaches (e.g. chromatin-interaction, eQTL). Even when directly incorporating noncoding SNPs through 5' scRNA-seq measures of regulatory elements, non disease-specific atlases gave misleading results by not containing disease-tissue specific transcriptomic patterns. Despite this criticality of tissue-specific scRNA-seq, we showed that scDRS enabled deconvolution of two similar diseases with a single fine-grained scRNA-seq atlas and separate GWAS. Indeed, we identified supported and novel genetic-phenotype linkages separating RA and ankylosing spondylitis, and UC and crohn's disease. Overall, while noting evolving single-cell technologies, our study provides key findings for integrating expanding fine-grained scRNA-seq, GWAS, and noncoding SNP resources to unravel the complexities of inflammatory diseases.
Collapse
Affiliation(s)
- Hope A. Townsend
- Biofrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
- Department of Molecular, Cellular, Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
| | - Kaylee J. Rosenberger
- Biofrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, CO, United States
| | - Lauren A. Vanderlinden
- Department of Medicine, Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, United States
- Department of Biomedical Informatics, Center for Health AI, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Jun Inamo
- Department of Medicine, Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, United States
- Department of Biomedical Informatics, Center for Health AI, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Fan Zhang
- Biofrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
- Department of Medicine, Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, United States
- Department of Biomedical Informatics, Center for Health AI, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| |
Collapse
|
7
|
Loughnan R, Ahern J, Boyle M, Jernigan TL, Hagler DJ, Iversen JR, Frei O, Smith DM, Andreassen O, Zaitlen N, Sugrue L, Thompson WK, Dale A, Schork AJ, Fan CC. Hemochromatosis neural archetype reveals iron disruption in motor circuits. SCIENCE ADVANCES 2024; 10:eadp4431. [PMID: 39576859 PMCID: PMC11584016 DOI: 10.1126/sciadv.adp4431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Our understanding of brain iron regulation and its disruption in disease is limited. Excess iron affects motor circuitry, contributing to Parkinson's disease (PD) risk. The molecular mechanisms regulating central iron levels, beyond a few well-known genes controlling peripheral iron, remain unclear. We generated scores based on the archetypal brain iron accumulation observed in magnetic resonance imaging scans of individuals with excessive dietary iron absorption and hemochromatosis risk. Genome-wide analysis revealed that this score is highly heritable, identifying loci associated with iron homeostasis, and driven by peripheral iron levels. Our score predicted gait abnormalities and showed a U-shaped relationship with PD risk, identifying individuals with threefold increased risk. These results establish a hormetic relationship between brain iron and PD risk, where central iron levels are strongly determined by genetics via peripheral iron. This framework combining forward and reverse genetics is a powerful study design to understand genomic drivers underlying high dimensional phenotypes.
Collapse
Affiliation(s)
- Robert Loughnan
- Department of Cognitive Science, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Center for Human Development, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92161, USA
- Center for Multimodal Imaging and Genetics, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, OK 74103, USA
| | - Jonathan Ahern
- Department of Cognitive Science, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Center for Human Development, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92161, USA
| | - Mary Boyle
- Department of Cognitive Science, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Terry L Jernigan
- Department of Cognitive Science, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Center for Human Development, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92161, USA
- Department of Psychiatry, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
- Department of Radiology, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - Donald J Hagler
- Center for Multimodal Imaging and Genetics, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
- Department of Radiology, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - John R Iversen
- Center for Human Development, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92161, USA
- Swartz Center for Computational Neuroscience, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92161, USA
| | - Oleksandr Frei
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Diana M Smith
- Center for Human Development, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92161, USA
- Center for Multimodal Imaging and Genetics, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
- Medical Scientist Training Program, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Ole Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Noah Zaitlen
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Leo Sugrue
- Department of Radiology and Biomedical Imaging and Department of Psychiatry, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Wesley K Thompson
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, OK 74103, USA
| | - Anders Dale
- Department of Cognitive Science, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Center for Multimodal Imaging and Genetics, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
- Department of Radiology, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
- Department of Neuroscience, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - Andrew J Schork
- Institute of Biological Psychiatry, Mental Health Center-Sct Hans, Copenhagen University Hospital, Copenhagen, Denmark
- Section for Geogenetics, GLOBE Institute, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Chun Chieh Fan
- Center for Multimodal Imaging and Genetics, University of California, San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA 92037, USA
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, OK 74103, USA
| |
Collapse
|
8
|
Webber CJ, van de Spek SJF, Cruz AL, Puri S, Zhang C, Aw JTM, Papadimitriou GZ, Roberts R, Jiang K, Tran TN, Zhang L, Taylor A, Wang Z, Porter J, Sotiropoulos I, Emili A, Silva J, Li H, Wolozin B. TIA1 Mediates Divergent Inflammatory Responses to Tauopathy in Microglia and Macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622325. [PMID: 39574689 PMCID: PMC11580906 DOI: 10.1101/2024.11.06.622325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
The RNA binding protein TIA1 is known to regulate stress responses. Here we show that TIA1 plays a much broader role in inflammatory cells, being required for the microglial sensome. We crossed TIA1 cKO mice (using a CX3CR1 driven cre element) to PS19 MAPT P301S tauopathy mice. The peripheral macrophages of TIA1 cKO mice exhibited a hyper-inflammatory phenotype with increased cytokine signaling, as expected. Surprisingly, the brains of these mice showed striking reductions in inflammation, including decreases in microglial inflammatory cytokines (TNFα and IL-1β) and sensome markers (CLEC7A, TREM2, ITGAX); these reductions were accompanied by corresponding decreases in tau pathology. Analysis of the brain TIA1 protein interactome identified brain selective TIA1 protein mediated pathways, including strong interactions with the microglial protein C1q, which directs pruning of dystrophic neurons. These results uncover a previously unknown regulatory role for TIA1 in microglial activation in the context of neurodegenerative disease and highlights the divergent regulation of two mononuclear phagocytic lineages: microglia and macrophages.
Collapse
Affiliation(s)
- Chelsea J. Webber
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
- Dept. of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Sophie J. F. van de Spek
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Anna Lourdes Cruz
- Dept. of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
- Dept. of Neuroscience, Johns Hopkins Medical Center, Baltimore, MD
| | - Sambhavi Puri
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Cheng Zhang
- Dept. of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Jacqueline T. M. Aw
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Georgia-Zeta Papadimitriou
- Laboratory of Brain Exosomes & Pathology, Institute of Biosciences & Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Rebecca Roberts
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Kiki Jiang
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Thuc Nhan Tran
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Lushuang Zhang
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Alexandria Taylor
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Zihan Wang
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
| | - Jacob Porter
- Department of Chemical Physiology & Biochemistry, Oregon Health Sciences University, Portland, OR
| | - Ionnis Sotiropoulos
- Laboratory of Brain Exosomes & Pathology, Institute of Biosciences & Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andrew Emili
- Department of Chemical Physiology & Biochemistry, Oregon Health Sciences University, Portland, OR
| | - Joana Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Hu Li
- Dept. of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Benjamin Wolozin
- Dept. of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
- Dept. of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA
- Department of Chemical Physiology & Biochemistry, Oregon Health Sciences University, Portland, OR
| |
Collapse
|
9
|
Park S, Kim S, Kim B, Kim DS, Kim J, Ahn Y, Kim H, Song M, Shim I, Jung SH, Cho C, Lim S, Hong S, Jo H, Fahed AC, Natarajan P, Ellinor PT, Torkamani A, Park WY, Yu TY, Myung W, Won HH. Multivariate genomic analysis of 5 million people elucidates the genetic architecture of shared components of the metabolic syndrome. Nat Genet 2024; 56:2380-2391. [PMID: 39349817 PMCID: PMC11549047 DOI: 10.1038/s41588-024-01933-1] [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: 10/19/2023] [Accepted: 08/29/2024] [Indexed: 11/10/2024]
Abstract
Metabolic syndrome (MetS) is a complex hereditary condition comprising various metabolic traits as risk factors. Although the genetics of individual MetS components have been investigated actively through large-scale genome-wide association studies, the conjoint genetic architecture has not been fully elucidated. Here, we performed the largest multivariate genome-wide association study of MetS in Europe (nobserved = 4,947,860) by leveraging genetic correlation between MetS components. We identified 1,307 genetic loci associated with MetS that were enriched primarily in brain tissues. Using transcriptomic data, we identified 11 genes associated strongly with MetS. Our phenome-wide association and Mendelian randomization analyses highlighted associations of MetS with diverse diseases beyond cardiometabolic diseases. Polygenic risk score analysis demonstrated better discrimination of MetS and predictive power in European and East Asian populations. Altogether, our findings will guide future studies aimed at elucidating the genetic architecture of MetS.
Collapse
Affiliation(s)
- Sanghyeon Park
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soyeon Kim
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beomsu Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dan Say Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Jaeyoung Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Yeeun Ahn
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hyejin Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Minku Song
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Injeong Shim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chamlee Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soohyun Lim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Sanghoon Hong
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Hyeonbin Jo
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Akl C Fahed
- Department of Medicine, Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Pradeep Natarajan
- Department of Medicine, Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Department of Medicine, Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Ali Torkamani
- Scripps Research Translational Institute, Scripps Research, La Jolla, CA, USA
| | - Woong-Yang Park
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Tae Yang Yu
- Department of Medicine, Division of Endocrinology and Metabolism, Wonkwang Medical Center, Wonkwang University School of Medicine, Iksan, South Korea
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.
- Department of Neuropsychiatry, College of Medicine, Seoul National University, Seoul, South Korea.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea.
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
10
|
Zhang H, Zheng R, Yu B, Yu Y, Luo X, Yin S, Zheng Y, Shi J, Ai S. Dissecting shared genetic architecture between depression and body mass index. BMC Med 2024; 22:455. [PMID: 39394142 PMCID: PMC11481102 DOI: 10.1186/s12916-024-03681-9] [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: 07/16/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND A growing body of evidence supports the comorbidity between depression (DEP) and obesity, yet the genetic mechanisms underlying this association remain unclear. Our study explored the shared genetic architecture and causal associations of DEP with BMI. METHODS We investigated the multigene overlap and genetic correlation between DEP (N > 1.3 million) and BMI (N = 806,834) based on genome-wide association studies (GWAS) and using the bivariate causal mixture model and linkage disequilibrium score regression (LDSC). The causal association was explored by bi-directional Mendelian randomization (MR). Common risk loci were identified through cross-trait meta-analyses. Stratified LDSC and multi-marker gene annotation analyses were applied to investigate single-nucleotide polymorphisms enrichment across tissue types, cell types, and functional categories. Finally, we explored shared functional genes by Summary Data-Based Mendelian Randomization (SMR) and further detected differential expression genes (DEG) in brain tissues of individuals with depression and obesity. RESULTS We found a positive genetic correlation between DEP and BMI (rg = 0.19, P = 4.07 × 10-26), which was more evident in local genomic regions. Cross-trait meta-analyses identified 16 shared genetic loci, 5 of which were newly identified, and they had influence on both diseases in the same direction. MR analysis showed a bidirectional causal association between DEP and BMI, with comparable effect sizes estimated in both directions. Combined with gene expression information, we found that genetic correlations between DEP and BMI were enriched in 6 brain regions, predominantly in the nucleus accumbens and anterior cingulate cortex. Moreover, 6 specific cell types and 23 functional genes were found to have an impact on both DEP and BMI across the brain regions. Of which, NEGR1 was identified as the most significant functional gene and associated with DEP and BMI at the genome-wide significance level (P < 5 × 10-8). Compared with healthy controls, the expression levels of NEGR1 gene were significant lower in brain tissues of individuals with depression and obesity. CONCLUSIONS Our study reveals shared genetic basis underpinnings between DEP and BMI, including genetic correlations and common genes. These insights offer novel opportunities and avenues for future research into their comorbidities.
Collapse
Affiliation(s)
- Hengyu Zhang
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Guangzhou, Guangdong, China
| | - Rui Zheng
- Center for Sleep and Circadian Medicine, The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Guangzhou, Guangdong, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 511436, China
| | - Binhe Yu
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center, Weihui, 453100, Henan, China
| | - Yuefeng Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China
| | - Xiaomin Luo
- Center for Sleep and Circadian Medicine, The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Guangzhou, Guangdong, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shujuan Yin
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Guangzhou, Guangdong, China
| | - Yingjun Zheng
- Department of Psychiatry, The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Guangzhou, Guangdong, China.
| | - Jie Shi
- National Institute On Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Haidian District, 38 Xueyuan Road, Beijing, 100191, China.
- The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
- The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing, 100191, China.
| | - Sizhi Ai
- Center for Sleep and Circadian Medicine, The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Guangzhou, Guangdong, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 511436, China.
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center, Weihui, 453100, Henan, China.
| |
Collapse
|
11
|
Davis CN, Khan Y, Toikumo S, Jinwala Z, Boomsma DI, Levey DF, Gelernter J, Kember RL, Kranzler HR. Genetic Insights into Externalizing and Internalizing Traits through Integration of the Research Domain Criteria and Hierarchical Taxonomy of Psychopathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.06.24305166. [PMID: 38645045 PMCID: PMC11030494 DOI: 10.1101/2024.04.06.24305166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background There is considerable comorbidity between externalizing (EXT) and internalizing (INT) psychopathology. Understanding the shared genetic underpinnings of these spectra is crucial for advancing knowledge of their biological bases and potential health impacts, and for informing empirical models like the Research Domain Criteria (RDoC) and Hierarchical Taxonomy of Psychopathology (HiTOP). Methods We conducted a multivariate genome-wide association study (GWAS) of EXT and INT psychopathology by applying genomic structural equation modeling to summary statistics from 16 EXT and INT traits in European-ancestry individuals (n = 16,400 to 1,074,629). Downstream analyses explored associations across RDoC units of analysis (i.e., genes, molecules, cells, circuits, physiology, and behaviors). Results The GWAS identified 409 lead single nucleotide polymorphisms (SNPs) for EXT, 85 for INT, and 256 for EXT+INT (i.e., shared) traits. Bivariate causal mixture models estimated that nearly all EXT and INT causal variants overlapped, despite a genetic correlation of 0.37 (SE = 0.02). Drug repurposing analyses identified potential therapeutic targets, including perturbagens affecting dopamine and serotonin pathways. EXT genes had enriched expression in GABAergic, cortical, and hippocampal neurons, while INT genes were more narrowly linked to GABAergic neurons. EXT+INT liability was associated with reduced grey matter volumes in the amygdala and subcallosal cortex. Conclusions These findings reveal both genetic overlap and distinct molecular and neurobiological pathways underlying EXT and INT psychopathology. By integrating genomic insights with the RDoC and HiTOP frameworks, this study advances our understanding of the mechanisms driving these dimensions of psychopathology.
Collapse
Affiliation(s)
- Christal N. Davis
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Yousef Khan
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Zeal Jinwala
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Dorret I. Boomsma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands and Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Daniel F. Levey
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Joel Gelernter
- VA Connecticut Healthcare Center, West Haven, CT, USA
- Departments of Psychiatry, Genetics, and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Rachel L. Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Henry R. Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
12
|
Ahmad S, Imtiaz MA, Mishra A, Wang R, Herrera-Rivero M, Bis JC, Fornage M, Roshchupkin G, Hofer E, Logue M, Longstreth WT, Xia R, Bouteloup V, Mosley T, Launer LJ, Khalil M, Kuhle J, Rissman RA, Chene G, Dufouil C, Djoussé L, Lyons MJ, Mukamal KJ, Kremen WS, Franz CE, Schmidt R, Debette S, Breteler MMB, Berger K, Yang Q, Seshadri S, Aziz NA, Ghanbari M, Ikram MA. Genome-wide association study meta-analysis of neurofilament light (NfL) levels in blood reveals novel loci related to neurodegeneration. Commun Biol 2024; 7:1103. [PMID: 39251807 PMCID: PMC11385583 DOI: 10.1038/s42003-024-06804-3] [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: 11/29/2023] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Neurofilament light chain (NfL) levels in circulation have been established as a sensitive biomarker of neuro-axonal damage across a range of neurodegenerative disorders. Elucidation of the genetic architecture of blood NfL levels could provide new insights into molecular mechanisms underlying neurodegenerative disorders. In this meta-analysis of genome-wide association studies (GWAS) of blood NfL levels from eleven cohorts of European ancestry, we identify two genome-wide significant loci at 16p12 (UMOD) and 17q24 (SLC39A11). We observe association of three loci at 1q43 (FMN2), 12q14, and 12q21 with blood NfL levels in the meta-analysis of African-American ancestry. In the trans-ethnic meta-analysis, we identify three additional genome-wide significant loci at 1p32 (FGGY), 6q14 (TBX18), and 4q21. In the post-GWAS analyses, we observe the association of higher NfL polygenic risk score with increased plasma levels of total-tau, Aβ-40, Aβ-42, and higher incidence of Alzheimer's disease in the Rotterdam Study. Furthermore, Mendelian randomization analysis results suggest that a lower kidney function could cause higher blood NfL levels. This study uncovers multiple genetic loci of blood NfL levels, highlighting the genes related to molecular mechanism of neurodegeneration.
Collapse
Affiliation(s)
- Shahzad Ahmad
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, 3000, CA, Rotterdam, the Netherlands
- Oxford-GSK Institute of Computational and Molecular Medicine (IMCM), Centre for Human Genetics, Nuffield Department of Medicine (NDM), University of Oxford, Oxford, OX3 7BN, UK
| | - Mohammad Aslam Imtiaz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - Ruiqi Wang
- Boston University, Boston, MA, 02215, USA
| | - Marisol Herrera-Rivero
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, 1730 Minor Ave #1360, Seattle, WA, 98101, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, 1825 Pressler Street Houston, Houston, 77030, TX, USA
| | - Gennady Roshchupkin
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, 3000, CA, Rotterdam, the Netherlands
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Auenbruggerplatz 22, 8036, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Auenbruggerplatz 2, Fifth Floor, Graz, 8036, Austria
| | - Mark Logue
- National Center for PTSD, Behavioral Sciences Division at VA Boston Healthcare System, Boston, 150 South Huntington Avenue, Boston, MA, 02130, USA
- Department of Psychiatry and Biomedical Genetics, Boston University School of Medicine, Boston, 72 East Concord Street E200, Boston, MA, 02118, USA
| | - W T Longstreth
- Departments of Neurology and Epidemiology, University of Washington, Seattle, 3980 15th Ave NE Seattle, Seattle, WA, 98195, USA
| | - Rui Xia
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, 1825 Pressler Street Houston, Houston, 77030, TX, USA
| | - Vincent Bouteloup
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - Thomas Mosley
- MIND Center, University of Mississippi Medical Center, Jackson, 2500 North State Street, Jackson, MS, 39216, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Science, NIA Intramural Research Program, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Michael Khalil
- Department of Neurology, Medical University of Graz, Auenbruggerplatz 22, 8036, Graz, Austria
| | - Jens Kuhle
- Research Center for Clinical Neuroimmunology and Neuroscience University Hospital, Spitalstrasse 2, CH-4031, Basel, Switzerland
| | - Robert A Rissman
- Department of Physiology and Neuroscience, Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, California, USA
| | - Genevieve Chene
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - Carole Dufouil
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - Luc Djoussé
- Brigham and Women's Hospital, Harvard Medical School, Boston, 75 FRANCIS STREET, BOSTON MA 02115, MA, Boston, USA
| | - Michael J Lyons
- Department of Psychological & Brain Sciences, Boston University, Boston, 64 Cummington Mall # 149, Boston, MA, 02215, USA
| | - Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 330 Brookline Avenue Boston, MA, 02215, USA
| | - William S Kremen
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Carol E Franz
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Auenbruggerplatz 22, 8036, Graz, Austria
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
- CHU de Bordeaux, Department of Neurology, Institute for Neurodegenerative Diseases, F-33000, Bordeaux, France
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Institut für Epidemiologie und Sozialmedizin Albert-Schweitzer-Campus 1, Gebäude D3 48149, Münster, Germany
| | - Qiong Yang
- Boston University, Boston, MA, 02215, USA
| | - Sudha Seshadri
- Boston University, Boston, MA, 02215, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, 53127, Bonn, Germany
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, 3000, CA, Rotterdam, the Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, PO Box 2040, 3000, CA, Rotterdam, the Netherlands.
| |
Collapse
|
13
|
Li M, Dang X, Chen Y, Chen Z, Xu X, Zhao Z, Wu D. Cognitive processing speed and accuracy are intrinsically different in genetic architecture and brain phenotypes. Nat Commun 2024; 15:7786. [PMID: 39242605 PMCID: PMC11379965 DOI: 10.1038/s41467-024-52222-8] [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: 03/10/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
Since the birth of cognitive science, researchers have used reaction time and accuracy to measure cognitive ability. Although recognition of these two measures is often based on empirical observations, the underlying consensus is that most cognitive behaviors may be along two fundamental dimensions: cognitive processing speed (CPS) and cognitive processing accuracy (CPA). In this study, we used genomic-wide association studies (GWAS) data from 14 cognitive traits to show the presence of those two factors and revealed the specific neurobiological basis underlying them. We identified that CPS and CPA had distinct brain phenotypes (e.g. white matter microstructure), neurobiological bases (e.g. postsynaptic membrane), and developmental periods (i.e. late infancy). Moreover, those two factors showed differential associations with other health-related traits such as screen exposure and sleep status, and a significant causal relationship with psychiatric disorders such as major depressive disorder and schizophrenia. Utilizing an independent cohort from the Adolescent Brain Cognitive Development (ABCD) study, we also uncovered the distinct contributions of those two factors on the cognitive development of young adolescents. These findings reveal two fundamental factors underlying various cognitive abilities, elucidate the distinct brain structural fingerprint and genetic architecture of CPS and CPA, and hint at the complex interrelationship between cognitive ability, lifestyle, and mental health.
Collapse
Affiliation(s)
- Mingyang Li
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Xixi Dang
- Department of Psychology, Hangzhou Normal University, Hangzhou, China
| | - Yiwei Chen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Zhifan Chen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Xinyi Xu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Zhiyong Zhao
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Yuquan Campus, Hangzhou, 310027, China.
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
- Binjiang Institute, Zhejiang University, Hangzhou, China.
| |
Collapse
|
14
|
Sullivan PF, Yao S, Hjerling-Leffler J. Schizophrenia genomics: genetic complexity and functional insights. Nat Rev Neurosci 2024; 25:611-624. [PMID: 39030273 DOI: 10.1038/s41583-024-00837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/21/2024]
Abstract
Determining the causes of schizophrenia has been a notoriously intractable problem, resistant to a multitude of investigative approaches over centuries. In recent decades, genomic studies have delivered hundreds of robust findings that implicate nearly 300 common genetic variants (via genome-wide association studies) and more than 20 rare variants (via whole-exome sequencing and copy number variant studies) as risk factors for schizophrenia. In parallel, functional genomic and neurobiological studies have provided exceptionally detailed information about the cellular composition of the brain and its interconnections in neurotypical individuals and, increasingly, in those with schizophrenia. Taken together, these results suggest unexpected complexity in the mechanisms that drive schizophrenia, pointing to the involvement of ensembles of genes (polygenicity) rather than single-gene causation. In this Review, we describe what we now know about the genetics of schizophrenia and consider the neurobiological implications of this information.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Shuyang Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
15
|
Weller AE, Ferraro TN, Doyle GA, Reiner BC, Berrettini WH, Crist RC. Analysis of single-cell transcriptome data from a mouse model implicates protein synthesis dysfunction in schizophrenia. Genes Genomics 2024; 46:1071-1084. [PMID: 39083157 DOI: 10.1007/s13258-024-01545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/08/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Schizophrenia is a mental disorder that causes considerable morbidity, whose risk largely results from genetic factors. Setd1a is a gene implicated in schizophrenia. OBJECTIVE To study the gene expression changes found in heterozygous Setd1a± knockout mice in order to gain useful insight into schizophrenia pathogenesis. METHODS We mined a single-cell RNA sequencing (scRNAseq) dataset from the prefrontal cortex (PFC) and striatum of Setd1a± mice and identified cell type-specific differentially expressed genes (DEGs) and differential transcript usage (DTU). DEGs and genes containing DTU found in each cell type were used to identify affected biological pathways using Ingenuity Pathway Analysis (IPA). RESULTS We identified 273 unique DEGs across all cell types in PFC and 675 unique gene peaks containing DTU. In striatum, we identified 327 unique DEGs across all cell types and 8 unique gene peaks containing DTU. Key IPA findings from the analysis of DEGs found in PFC and striatum implicate processes involved in protein synthesis, mitochondrial function, cell metabolism, and inflammation. IPA analysis of genes containing DTU in PFC points to protein synthesis, as well as cellular activities involving intracellular signaling and neurotransmission. One canonical pathway, 'EIF2 Signaling', which is involved in the regulation of protein synthesis, was detected in PFC DEGs, striatum DEGs, and PFC genes containing DTU, drawing attention to its importance in schizophrenia pathophysiology. CONCLUSION Processes involving protein synthesis in general and the 'EIF2 Signaling' pathway in particular could be targets for the development of new research strategies and biomarkers in schizophrenia.
Collapse
Affiliation(s)
- Andrew E Weller
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US.
| | - Thomas N Ferraro
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, US
| | - Glenn A Doyle
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, US
| | - Benjamin C Reiner
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
| | - Wade H Berrettini
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
| | - Richard C Crist
- Molecular and Neural Basis of Psychiatric Disease Section, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 S. 31 St., Suite 2200, Philadelphia, PA, 19104, US
| |
Collapse
|
16
|
Cañadas-Garre M, Maqueda JJ, Baños-Jaime B, Hill C, Skelly R, Cappa R, Brennan E, Doyle R, Godson C, Maxwell AP, McKnight AJ. Mitochondrial related variants associated with cardiovascular traits. Front Physiol 2024; 15:1395371. [PMID: 39258111 PMCID: PMC11385366 DOI: 10.3389/fphys.2024.1395371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Cardiovascular disease (CVD) is responsible for over 30% of mortality worldwide. CVD arises from the complex influence of molecular, clinical, social, and environmental factors. Despite the growing number of autosomal genetic variants contributing to CVD, the cause of most CVDs is still unclear. Mitochondria are crucial in the pathophysiology, development and progression of CVDs; the impact of mitochondrial DNA (mtDNA) variants and mitochondrial haplogroups in the context of CVD has recently been highlighted. Aims We investigated the role of genetic variants in both mtDNA and nuclear-encoded mitochondrial genes (NEMG) in CVD, including coronary artery disease (CAD), hypertension, and serum lipids in the UK Biobank, with sub-group analysis for diabetes. Methods We investigated 371,542 variants in 2,527 NEMG, along with 192 variants in 32 mitochondrial genes in 381,994 participants of the UK Biobank, stratifying by presence of diabetes. Results Mitochondrial variants showed associations with CVD, hypertension, and serum lipids. Mitochondrial haplogroup J was associated with CAD and serum lipids, whereas mitochondrial haplogroups T and U were associated with CVD. Among NEMG, variants within Nitric Oxide Synthase 3 (NOS3) showed associations with CVD, CAD, hypertension, as well as diastolic and systolic blood pressure. We also identified Translocase Of Outer Mitochondrial Membrane 40 (TOMM40) variants associated with CAD; Solute carrier family 22 member 2 (SLC22A2) variants associated with CAD and CVD; and HLA-DQA1 variants associated with hypertension. Variants within these three genes were also associated with serum lipids. Conclusion Our study demonstrates the relevance of mitochondrial related variants in the context of CVD. We have linked mitochondrial haplogroup U to CVD, confirmed association of mitochondrial haplogroups J and T with CVD and proposed new markers of hypertension and serum lipids in the context of diabetes. We have also evidenced connections between the etiological pathways underlying CVDs, blood pressure and serum lipids, placing NOS3, SLC22A2, TOMM40 and HLA-DQA1 genes as common nexuses.
Collapse
Affiliation(s)
- Marisa Cañadas-Garre
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School (Population Health Sciences), University of Bristol Oakfield House, Belfast, United Kingdom
| | - Joaquín J Maqueda
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Blanca Baños-Jaime
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Sevilla, Spain
| | - Claire Hill
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Ryan Skelly
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Ruaidhri Cappa
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| | - Eoin Brennan
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Ross Doyle
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- Mater Misericordiae University Hospital, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Alexander P Maxwell
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
- Regional Nephrology Unit, Belfast City Hospital Belfast, Belfast, United Kingdom
| | - Amy Jayne McKnight
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, United Kingdom
| |
Collapse
|
17
|
Gezsi A, Van der Auwera S, Mäkinen H, Eszlari N, Hullam G, Nagy T, Bonk S, González-Colom R, Gonda X, Garvert L, Paajanen T, Gal Z, Kirchner K, Millinghoffer A, Schmidt CO, Bolgar B, Roca J, Cano I, Kuokkanen M, Antal P, Juhasz G. Unique genetic and risk-factor profiles in clusters of major depressive disorder-related multimorbidity trajectories. Nat Commun 2024; 15:7190. [PMID: 39168988 PMCID: PMC11339304 DOI: 10.1038/s41467-024-51467-7] [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: 08/07/2023] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
The heterogeneity and complexity of symptom presentation, comorbidities and genetic factors pose challenges to the identification of biological mechanisms underlying complex diseases. Current approaches used to identify biological subtypes of major depressive disorder (MDD) mainly focus on clinical characteristics that cannot be linked to specific biological models. Here, we examined multimorbidities to identify MDD subtypes with distinct genetic and non-genetic factors. We leveraged dynamic Bayesian network approaches to determine a minimal set of multimorbidities relevant to MDD and identified seven clusters of disease-burden trajectories throughout the lifespan among 1.2 million participants from cohorts in the UK, Finland, and Spain. The clusters had clear protective- and risk-factor profiles as well as age-specific clinical courses mainly driven by inflammatory processes, and a comprehensive map of heritability and genetic correlations among these clusters was revealed. Our results can guide the development of personalized treatments for MDD based on the unique genetic, clinical and non-genetic risk-factor profiles of patients.
Collapse
Grants
- This research has been conducted using the UK Biobank Resource under Application Number 1602. Linked health data Copyright © 2019, NHS England. Re-used with the permission of the UK Biobank. All rights reserved. This study was supported by the Hungarian National Research, Development, and Innovation Office 2019-2.1.7-ERA-NET-2020-00005 under the frame of ERA PerMed (ERAPERMED2019-108); the Hungarian National Research, Development, and Innovation Office (K 143391, K 139330, PD 146014, and PD 134449 grants); the Hungarian Brain Research Program 3.0 (NAP2022-I-4/2022); and the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund, under the TKP2021-EGA funding scheme (TKP2021-EGA-25 and TKP2021-EGA-02). Supported by the European Union project RRF-2.3.1-21-2022-00004 within the framework of the Artificial Intelligence National Laboratory.
Collapse
Affiliation(s)
- Andras Gezsi
- Department of Artificial Intelligence and Systems Engineering, Budapest University of Technology and Economics, Budapest, Hungary
| | - Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Hannu Mäkinen
- Department of Public Health and Welfare, Population Health Unit, Public Health Research Team, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nora Eszlari
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gabor Hullam
- Department of Artificial Intelligence and Systems Engineering, Budapest University of Technology and Economics, Budapest, Hungary
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
| | - Tamas Nagy
- Department of Artificial Intelligence and Systems Engineering, Budapest University of Technology and Economics, Budapest, Hungary
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Sarah Bonk
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Rubèn González-Colom
- Clínic Barcelona, Fundació de Recerca Clinic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Xenia Gonda
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Linda Garvert
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Teemu Paajanen
- Department of Public Health and Welfare, Population Health Unit, Public Health Research Team, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Zsofia Gal
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Kevin Kirchner
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | | | - Carsten O Schmidt
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Bence Bolgar
- Department of Artificial Intelligence and Systems Engineering, Budapest University of Technology and Economics, Budapest, Hungary
| | - Josep Roca
- Clínic Barcelona, Fundació de Recerca Clinic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Isaac Cano
- Clínic Barcelona, Fundació de Recerca Clinic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Mikko Kuokkanen
- Department of Public Health and Welfare, Population Health Unit, Public Health Research Team, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine at University of Texas Rio Grande Valley, Brownsville, TX, USA
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Peter Antal
- Department of Artificial Intelligence and Systems Engineering, Budapest University of Technology and Economics, Budapest, Hungary
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, Budapest, Hungary.
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
18
|
Yang X, Li Y, Mei T, Duan J, Yan X, McNaughton LR, He Z. Genome-wide association study of exercise-induced skeletal muscle hypertrophy and the construction of predictive model. Physiol Genomics 2024; 56:578-589. [PMID: 38881426 DOI: 10.1152/physiolgenomics.00019.2024] [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: 02/11/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024] Open
Abstract
The aim of the current study was to investigate interindividual differences in muscle thickness of the rectus femoris (MTRF) following 12 wk of resistance training (RT) or high-intensity interval training (HIIT) to explore the genetic architecture underlying skeletal muscle hypertrophy and to construct predictive models. We conducted musculoskeletal ultrasound assessments of the MTRF response in 440 physically inactive adults after the 12-wk exercise period. A genome-wide association study was used to identify variants associated with the MTRF response, separately for RT and HIIT. Using the polygenic predictor score (PPS), we estimated the genetic contribution to exercise-induced hypertrophy. Predictive models for the MTRF response were constructed using random forest (RF), support vector mac (SVM), and generalized linear model (GLM) in 10 cross-validated approaches. MTRF increased significantly after both RT (8.8%, P < 0.05) and HIIT (5.3%, P < 0.05), but with considerable interindividual differences (RT: -13.5 to 38.4%, HIIT: -14.2 to 30.7%). Eleven lead single-nucleotide polymorphisms in RT and eight lead single-nucleotide polymorphisms in HIIT were identified at a significance level of P < 1 × 10-5. The PPS was associated with the MTRF response, explaining 47.2% of the variation in response to RT and 38.3% of the variation in response to HIIT. Notably, the GLM and SVM predictive models exhibited superior performance compared with RF models (P < 0.05), and the GLM demonstrated optimal performance with an area under curve of 0.809 (95% confidence interval: 0.669-0.949). Factors such as PPS, baseline MTRF, and exercise protocol exerted influence on the MTRF response to exercise, with PPS being the primary contributor. The GLM and SVM predictive model, incorporating both genetic and phenotypic factors, emerged as promising tools for predicting exercise-induced skeletal muscle hypertrophy.NEW & NOTEWORTHY The interindividual variability induced muscle hypertrophy by resistance training (RT) or high-intensity interval training (HIIT) and the associated genetic architecture remain uncertain. We identified genetic variants that underlie RT- or HIIT-induced muscle hypertrophy and established them as pivotal factors influencing the response regardless of the training type. The genetic-phenotype predictive model developed has the potential to identify nonresponders or individuals with low responsiveness before engaging in exercise training.
Collapse
Affiliation(s)
- Xiaolin Yang
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
- Key Laboratory for Performance Training and Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
| | - Yanchun Li
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
- Key Laboratory for Performance Training and Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
| | - Tao Mei
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
- Key Laboratory for Performance Training and Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
| | - Jiayan Duan
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Xu Yan
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
- Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science, St Albans, Victoria, Australia
| | - Lars Robert McNaughton
- Sport Performance, Exercise and Nutrition Research Group, Department of Sport and Physical Activity, Edge Hill University, Ormskirk, United Kingdom
| | - Zihong He
- Biology Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
19
|
Li Y, Yao M, Xie F, Qiu Y, Zhao X, Li R. Gut microbiota as a residual risk factor causally influencing cardiac structure and function: Mendelian randomization analysis and biological annotation. Front Microbiol 2024; 15:1410272. [PMID: 39132134 PMCID: PMC11316272 DOI: 10.3389/fmicb.2024.1410272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/04/2024] [Indexed: 08/13/2024] Open
Abstract
Background The gut microbiota (GM) is widely acknowledged to have a significant impact on cardiovascular health and may act as a residual risk factor affecting cardiac structure and function. However, the causal relationship between GM and cardiac structure and function remains unclear. Objective This study aims to employ a two-sample Mendelian randomization (MR) approach to investigate the causal association between GM and cardiac structure and function. Methods Data on 119 GM genera were sourced from a genome-wide association study (GWAS) meta-analysis (13,266 European participants) conducted by the MiBioGen consortium, while data on 16 parameters of cardiac structure and function were obtained from the UK Biobank's GWAS of cardiac magnetic resonance imaging (up to 41,135 European participants). Inverse variance weighted (IVW), MR-Egger, and weighted median (WM) methods were utilized for causal association assessments, with sensitivity analyses conducted to reinforce the findings. Finally, biological annotation was performed on the GWAS data of GM and cardiac phenotypes with causal associations to explore potential mechanisms. Results The MR analysis, predominantly based on the IVW model, revealed 93 causal associations between the genetically predicted abundance of 44 GM genera and 16 cardiac structure and function parameters. These associations maintained consistent directions in MR-Egger and WM models, with no evidence of pleiotropy detected. Biological annotations suggest that GM may influence cardiac structure and function through pathways involved in myocardial cell development, cardiac contractility, and apoptosis. Conclusion The MR analysis supports a causal association between certain abundances of genetically predicted GM and cardiac structure and function, suggesting that GM could be a residual risk factor impacting cardiac phenotypes.
Collapse
Affiliation(s)
- Yihua Li
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meidan Yao
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- National Key Laboratory of Chinese Medicine Evidence, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fei Xie
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yijun Qiu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinjun Zhao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Bhatt RR, Gadewar SP, Shetty A, Ba Gari I, Haddad E, Javid S, Ramesh A, Nourollahimoghadam E, Zhu AH, de Leeuw C, Thompson PM, Medland SE, Jahanshad N. The Genetic Architecture of the Human Corpus Callosum and its Subregions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.603147. [PMID: 39091796 PMCID: PMC11291056 DOI: 10.1101/2024.07.22.603147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The corpus callosum (CC) is the largest set of white matter fibers connecting the two hemispheres of the brain. In humans, it is essential for coordinating sensorimotor responses, performing associative/executive functions, and representing information in multiple dimensions. Understanding which genetic variants underpin corpus callosum morphometry, and their shared influence on cortical structure and susceptibility to neuropsychiatric disorders, can provide molecular insights into the CC's role in mediating cortical development and its contribution to neuropsychiatric disease. To characterize the morphometry of the midsagittal corpus callosum, we developed a publicly available artificial intelligence based tool to extract, parcellate, and calculate its total and regional area and thickness. Using the UK Biobank (UKB) and the Adolescent Brain Cognitive Development study (ABCD), we extracted measures of midsagittal corpus callosum morphometry and performed a genome-wide association study (GWAS) meta-analysis of European participants (combined N = 46,685). We then examined evidence for generalization to the non-European participants of the UKB and ABCD cohorts (combined N = 7,040). Post-GWAS analyses implicate prenatal intracellular organization and cell growth patterns, and high heritability in regions of open chromatin, suggesting transcriptional activity regulation in early development. Results suggest programmed cell death mediated by the immune system drives the thinning of the posterior body and isthmus. Global and local genetic overlap, along with causal genetic liability, between the corpus callosum, cerebral cortex, and neuropsychiatric disorders such as attention-deficit/hyperactivity and bipolar disorders were identified. These results provide insight into variability of corpus callosum development, its genetic influence on the cerebral cortex, and biological mechanisms related to neuropsychiatric dysfunction.
Collapse
Affiliation(s)
- Ravi R Bhatt
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Shruti P Gadewar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Ankush Shetty
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Iyad Ba Gari
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Elizabeth Haddad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Shayan Javid
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Abhinaav Ramesh
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Elnaz Nourollahimoghadam
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Alyssa H Zhu
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Christiaan de Leeuw
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Sarah E Medland
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| |
Collapse
|
21
|
Giel AS, Bigge J, Schumacher J, Maj C, Dasmeh P. Analysis of Evolutionary Conservation, Expression Level, and Genetic Association at a Genome-wide Scale Reveals Heterogeneity Across Polygenic Phenotypes. Mol Biol Evol 2024; 41:msae115. [PMID: 38865495 PMCID: PMC11247350 DOI: 10.1093/molbev/msae115] [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: 08/04/2023] [Revised: 03/22/2024] [Accepted: 05/03/2024] [Indexed: 06/14/2024] Open
Abstract
Understanding the expression level and evolutionary rate of associated genes with human polygenic diseases provides crucial insights into their disease-contributing roles. In this work, we leveraged genome-wide association studies (GWASs) to investigate the relationship between the genetic association and both the evolutionary rate (dN/dS) and expression level of human genes associated with the two polygenic diseases of schizophrenia and coronary artery disease. Our findings highlight a distinct variation in these relationships between the two diseases. Genes associated with both diseases exhibit a significantly greater variance in evolutionary rate compared to those implicated in monogenic diseases. Expanding our analyses to 4,756 complex traits in the GWAS atlas database, we unraveled distinct trait categories with a unique interplay among the evolutionary rate, expression level, and genetic association of human genes. In most polygenic traits, highly expressed genes were more associated with the polygenic phenotypes compared to lowly expressed genes. About 69% of polygenic traits displayed a negative correlation between genetic association and evolutionary rate, while approximately 30% of these traits showed a positive correlation between genetic association and evolutionary rate. Our results demonstrate the presence of a spectrum among complex traits, shaped by natural selection. Notably, at opposite ends of this spectrum, we find metabolic traits being more likely influenced by purifying selection, and immunological traits that are more likely shaped by positive selection. We further established the polygenic evolution portal (evopolygen.de) as a resource for investigating relationships and generating hypotheses in the field of human polygenic trait evolution.
Collapse
Affiliation(s)
- Ann-Sophie Giel
- Centre for Human Genetics, Marburg University, Marburg, Germany
| | - Jessica Bigge
- Centre for Human Genetics, Marburg University, Marburg, Germany
| | | | - Carlo Maj
- Centre for Human Genetics, Marburg University, Marburg, Germany
| | - Pouria Dasmeh
- Centre for Human Genetics, Marburg University, Marburg, Germany
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Institute for Evolutionary Biology and Environmental Studies, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Yang JC, Hsu TH, Chen CS, Yu JH, Lin KI, Chen YJ. Enhanced Proteomic Coverage in Tissue Microenvironment by Immune Cell Subtype Library-Assisted DIA-MS. Mol Cell Proteomics 2024; 23:100792. [PMID: 38810695 PMCID: PMC11260568 DOI: 10.1016/j.mcpro.2024.100792] [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: 01/23/2024] [Revised: 04/30/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024] Open
Abstract
Immune cells that infiltrate the tumor microenvironment (TME) play crucial roles in shaping cancer development and influencing clinical outcomes and therapeutic responses. However, obtaining a comprehensive proteomic snapshot of tumor-infiltrating immunity in clinical specimens is often hindered by small sample amounts and a low proportion of immune infiltrating cells in the TME. To enable in-depth and highly sensitive profiling of microscale tissues, we established an immune cell-enriched library-assisted strategy for data-independent acquisition mass spectrometry (DIA-MS). Firstly, six immune cell subtype-specific spectral libraries were established from sorted cluster of differentiation markers, CD8+, CD4+ T lymphocytes, B lymphocytes, natural killer cells, dendritic cells, and macrophages in murine mesenteric lymph nodes (MLNs), covering 7815 protein groups with surface markers and immune cell-enriched proteins. The feasibility of microscale immune proteomic profiling was demonstrated on 1 μg tissue protein from the tumor of murine colorectal cancer (CRC) models using single-shot DIA; the immune cell-enriched library increased coverage to quantify 7419 proteins compared to directDIA analysis (6978 proteins). The enhancement enabled the mapping of 841 immune function-related proteins and exclusive identification of many low-abundance immune proteins, such as CD1D1, and CD244, demonstrating high sensitivity for immune landscape profiling. This approach was used to characterize the MLNs in CRC models, aiming to elucidate the mechanism underlying their involvement in cancer development within the TME. Even with a low percentage of immune cell infiltration (0.25-3%) in the tumor, our results illuminate downregulation in the adaptive immune signaling pathways (such as C-type lectin receptor signaling, and chemokine signaling), T cell receptor signaling, and Th1/Th2/Th17 cell differentiation, suggesting an immunosuppressive status in MLNs of CRC model. The DIA approach using the immune cell-enriched libraries showcased deep coverage and high sensitivity that can facilitate illumination of the immune proteomic landscape for microscale samples.
Collapse
Affiliation(s)
- Jhih-Ci Yang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica and National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Tzi-Hui Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Jou-Hui Yu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan; Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica and National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Chemistry, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
23
|
Qi Q, Wang Y, Huang Y, Fan Y, Li X. PredGCN: a Pruning-enabled Gene-Cell Net for automatic cell annotation of single cell transcriptome data. Bioinformatics 2024; 40:btae421. [PMID: 38924517 PMCID: PMC11236098 DOI: 10.1093/bioinformatics/btae421] [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: 02/21/2024] [Revised: 05/27/2024] [Accepted: 06/25/2024] [Indexed: 06/28/2024] Open
Abstract
MOTIVATION The annotation of cell types from single-cell transcriptomics is essential for understanding the biological identity and functionality of cellular populations. Although manual annotation remains the gold standard, the advent of automatic pipelines has become crucial for scalable, unbiased, and cost-effective annotations. Nonetheless, the effectiveness of these automatic methods, particularly those employing deep learning, significantly depends on the architecture of the classifier and the quality and diversity of the training datasets. RESULTS To address these limitations, we present a Pruning-enabled Gene-Cell Net (PredGCN) incorporating a Coupled Gene-Cell Net (CGCN) to enable representation learning and information storage. PredGCN integrates a Gene Splicing Net (GSN) and a Cell Stratification Net (CSN), employing a pruning operation (PrO) to dynamically tackle the complexity of heterogeneous cell identification. Among them, GSN leverages multiple statistical and hypothesis-driven feature extraction methods to selectively assemble genes with specificity for scRNA-seq data while CSN unifies elements based on diverse region demarcation principles, exploiting the representations from GSN and precise identification from different regional homogeneity perspectives. Furthermore, we develop a multi-objective Pareto pruning operation (Pareto PrO) to expand the dynamic capabilities of CGCN, optimizing the sub-network structure for accurate cell type annotation. Multiple comparison experiments on real scRNA-seq datasets from various species have demonstrated that PredGCN surpasses existing state-of-the-art methods, including its scalability to cross-species datasets. Moreover, PredGCN can uncover unknown cell types and provide functional genomic analysis by quantifying the influence of genes on cell clusters, bringing new insights into cell type identification and characterizing scRNA-seq data from different perspectives. AVAILABILITY AND IMPLEMENTATION The source code is available at https://github.com/IrisQi7/PredGCN and test data is available at https://figshare.com/articles/dataset/PredGCN/25251163.
Collapse
Affiliation(s)
- Qi Qi
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| | - Yunhe Wang
- School of Artificial Intelligence, Hebei University of Technology, Tianjin 300401, China
| | - Yujian Huang
- College of Computer Science and Cyber Security, Chengdu University of Technology, Chengdu 610059, China
| | - Yi Fan
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| |
Collapse
|
24
|
Jiang J, Hiron TK, Agbaedeng TA, Malhotra Y, Drydale E, Bancroft J, Ng E, Reschen ME, Davison LJ, O’Callaghan CA. A Novel Macrophage Subpopulation Conveys Increased Genetic Risk of Coronary Artery Disease. Circ Res 2024; 135:6-25. [PMID: 38747151 PMCID: PMC11191562 DOI: 10.1161/circresaha.123.324172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Coronary artery disease (CAD), the leading cause of death worldwide, is influenced by both environmental and genetic factors. Although over 250 genetic risk loci have been identified through genome-wide association studies, the specific causal variants and their regulatory mechanisms are still largely unknown, particularly in disease-relevant cell types such as macrophages. METHODS We utilized single-cell RNA-seq and single-cell multiomics approaches in primary human monocyte-derived macrophages to explore the transcriptional regulatory network involved in a critical pathogenic event of coronary atherosclerosis-the formation of lipid-laden foam cells. The relative genetic contribution to CAD was assessed by partitioning disease heritability across different macrophage subpopulations. Meta-analysis of single-cell RNA-seq data sets from 38 human atherosclerotic samples was conducted to provide high-resolution cross-referencing to macrophage subpopulations in vivo. RESULTS We identified 18 782 cis-regulatory elements by jointly profiling the gene expression and chromatin accessibility of >5000 macrophages. Integration with CAD genome-wide association study data prioritized 121 CAD-related genetic variants and 56 candidate causal genes. We showed that CAD heritability was not uniformly distributed and was particularly enriched in the gene programs of a novel CD52-hi lipid-handling macrophage subpopulation. These CD52-hi macrophages displayed significantly less lipoprotein accumulation and were also found in human atherosclerotic plaques. We investigated the cis-regulatory effect of a risk variant rs10488763 on FDX1, implicating the recruitment of AP-1 and C/EBP-β in the causal mechanisms at this locus. CONCLUSIONS Our results provide genetic evidence of the divergent roles of macrophage subsets in atherogenesis and highlight lipid-handling macrophages as a key subpopulation through which genetic variants operate to influence disease. These findings provide an unbiased framework for functional fine-mapping of genome-wide association study results using single-cell multiomics and offer new insights into the genotype-environment interactions underlying atherosclerotic disease.
Collapse
Affiliation(s)
- Jiahao Jiang
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Thomas K. Hiron
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Thomas A. Agbaedeng
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Yashaswat Malhotra
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Edward Drydale
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - James Bancroft
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| | - Esther Ng
- Nuffield Department of Orthopaedics, Kennedy Institute of Rheumatology, Rheumatology and Musculoskeletal Sciences (E.N.), University of Oxford, United Kingdom
| | - Michael E. Reschen
- Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, United Kingdom (M.E.R.)
| | - Lucy J. Davison
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, United Kingdom (L.J.D.)
| | - Chris A. O’Callaghan
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics (J.J., T.K.H., T.A.A., Y.M., E.D., J.B., L.J.D., C.A.O.), University of Oxford, United Kingdom
| |
Collapse
|
25
|
García-González J, Garcia-Gonzalez S, Liou L, O'Reilly PF. The Gene Expression Landscape of Disease Genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.20.24309121. [PMID: 38947033 PMCID: PMC11213058 DOI: 10.1101/2024.06.20.24309121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Fine-mapping and gene-prioritisation techniques applied to the latest Genome-Wide Association Study (GWAS) results have prioritised hundreds of genes as causally associated with disease. Here we leverage these recently compiled lists of high-confidence causal genes to interrogate where in the body disease genes operate. Specifically, we combine GWAS summary statistics, gene prioritisation results and gene expression RNA-seq data from 46 tissues and 204 cell types in relation to 16 major diseases (including 8 cancers). In tissues and cell types with well-established relevance to the disease, the prioritised genes typically have higher absolute and relative (i.e. tissue/cell specific) expression compared to non-prioritised 'control' genes. Examples include brain tissues in psychiatric disorders (P-value < 1×10-7), microglia cells in Alzheimer's Disease (P-value = 9.8×10-3) and colon mucosa in colorectal cancer (P-value < 1×10-3). We also observe significantly higher expression for disease genes in multiple tissues and cell types with no established links to the corresponding disease. While some of these results may be explained by cell types that span multiple tissues, such as macrophages in brain, blood, lung and spleen in relation to Alzheimer's disease (P-values < 1×10-3), the cause for others is unclear and motivates further investigation that may provide novel insights into disease etiology. For example, mammary tissue in Type 2 Diabetes (P-value < 1×10-7); reproductive tissues such as breast, uterus, vagina, and prostate in Coronary Artery Disease (P-value < 1×10-4); and motor neurons in psychiatric disorders (P-value < 3×10-4). In the GTEx dataset, tissue type is the major predictor of gene expression but the contribution of each predictor (tissue, sample, subject, batch) varies widely among disease-associated genes. Finally, we highlight genes with the highest levels of gene expression in relevant tissues to guide functional follow-up studies. Our results could offer novel insights into the tissues and cells involved in disease initiation, inform drug target and delivery strategies, highlighting potential off-target effects, and exemplify the relative performance of different statistical tests for linking disease genes with tissue and cell type gene expression.
Collapse
Affiliation(s)
- Judit García-González
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| | - Saul Garcia-Gonzalez
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
- Center for Excellence in Youth Education, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| | - Lathan Liou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| | - Paul F O'Reilly
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York City, NY 10029, USA
| |
Collapse
|
26
|
Lv Z, Jiang S, Kong S, Zhang X, Yue J, Zhao W, Li L, Lin S. Advances in Single-Cell Transcriptome Sequencing and Spatial Transcriptome Sequencing in Plants. PLANTS (BASEL, SWITZERLAND) 2024; 13:1679. [PMID: 38931111 PMCID: PMC11207393 DOI: 10.3390/plants13121679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/31/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
"Omics" typically involves exploration of the structure and function of the entire composition of a biological system at a specific level using high-throughput analytical methods to probe and analyze large amounts of data, including genomics, transcriptomics, proteomics, and metabolomics, among other types. Genomics characterizes and quantifies all genes of an organism collectively, studying their interrelationships and their impacts on the organism. However, conventional transcriptomic sequencing techniques target population cells, and their results only reflect the average expression levels of genes in population cells, as they are unable to reveal the gene expression heterogeneity and spatial heterogeneity among individual cells, thus masking the expression specificity between different cells. Single-cell transcriptomic sequencing and spatial transcriptomic sequencing techniques analyze the transcriptome of individual cells in plant or animal tissues, enabling the understanding of each cell's metabolites and expressed genes. Consequently, statistical analysis of the corresponding tissues can be performed, with the purpose of achieving cell classification, evolutionary growth, and physiological and pathological analyses. This article provides an overview of the research progress in plant single-cell and spatial transcriptomics, as well as their applications and challenges in plants. Furthermore, prospects for the development of single-cell and spatial transcriptomics are proposed.
Collapse
Affiliation(s)
- Zhuo Lv
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Shuaijun Jiang
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Shuxin Kong
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Xu Zhang
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Jiahui Yue
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Wanqi Zhao
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Long Li
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
| | - Shuyan Lin
- Co-Innovation Center for Sustainable Forestry in Southern China, Nanjing Forestry University, Nanjing 210037, China; (Z.L.); (S.J.); (S.K.); (X.Z.); (J.Y.); (W.Z.); (L.L.)
- Bamboo Research Institute, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
27
|
Zhukovsky P, Tio ES, Coughlan G, Bennett DA, Wang Y, Hohman TJ, Pizzagalli DA, Mulsant BH, Voineskos AN, Felsky D. Genetic influences on brain and cognitive health and their interactions with cardiovascular conditions and depression. Nat Commun 2024; 15:5207. [PMID: 38890310 PMCID: PMC11189393 DOI: 10.1038/s41467-024-49430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Approximately 40% of dementia cases could be prevented or delayed by modifiable risk factors related to lifestyle and environment. These risk factors, such as depression and vascular disease, do not affect all individuals in the same way, likely due to inter-individual differences in genetics. However, the precise nature of how genetic risk profiles interact with modifiable risk factors to affect brain health is poorly understood. Here we combine multiple data resources, including genotyping and postmortem gene expression, to map the genetic landscape of brain structure and identify 367 loci associated with cortical thickness and 13 loci associated with white matter hyperintensities (P < 5×10-8), with several loci also showing a significant association with cognitive function. We show that among 220 unique genetic loci associated with cortical thickness in our genome-wide association studies (GWAS), 95 also showed evidence of interaction with depression or cardiovascular conditions. Polygenic risk scores based on our GWAS of inferior frontal thickness also interacted with hypertension in predicting executive function in the Canadian Longitudinal Study on Aging. These findings advance our understanding of the genetic underpinning of brain structure and show that genetic risk for brain and cognitive health is in part moderated by treatable mid-life factors.
Collapse
Grants
- P30 AG072975 NIA NIH HHS
- U01 AG046152 NIA NIH HHS
- U01 AG061356 NIA NIH HHS
- R01 AG017917 NIA NIH HHS
- P30 AG010161 NIA NIH HHS
- R01 AG059716 NIA NIH HHS
- Wellcome Trust
- R01 AG015819 NIA NIH HHS
- Gouvernement du Canada | Instituts de Recherche en Santé du Canada | CIHR Skin Research Training Centre (Skin Research Training Centre)
- D.F. is supported by the generous contributions from the Michael and Sonja Koerner Foundation and the Krembil Family Foundation. D.F. is also supported in part by the Centre for Addiction and Mental Health (CAMH) Discovery Fund and CIHR.
- PZ was funded by the Canadian Institute of Health Research Postdoctoral Fellowship.
- Over the past 3 years, D.A.P has received consulting fees from Albright Stonebridge Group, Boehringer Ingelheim, Compass Pathways, Engrail Therapeutics, Neumora Therapeutics (formerly BlackThorn Therapeutics), Neurocrine Biosciences, Neuroscience Software, Otsuka, Sunovion, and Takeda; he has received honoraria from the Psychonomic Society and American Psychological Association (for editorial work) and from Alkermes; he has received research funding from the Brain and Behavior Research Foundation, the Dana Foundation, Millennium Pharmaceuticals, Wellcome Leap MCPsych, and NIMH; he has received stock options from Compass Pathways, Engrail Therapeutics, Neumora Therapeutics, and Neuroscience Software. No funding from these entities was used to support the current work, and all views expressed are solely those of the authors.
- U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- A.N.V. currently receives funding from CIHR, the NIH, the National Sciences and Engineering Research Council (NSERC), the CAMH Foundation, and the University of Toronto. E.S.T. was funded by the Ontario Graduate Scholarship.
Collapse
Affiliation(s)
- Peter Zhukovsky
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Earvin S Tio
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Gillian Coughlan
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - David A Bennett
- Department of Neurological Sciences, RUSH Medical College, Chicago, IL, 60612, USA
| | - Yanling Wang
- Department of Neurological Sciences, RUSH Medical College, Chicago, IL, 60612, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Diego A Pizzagalli
- Department of Psychiatry, Harvard Medical School and Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, MA, 02478, USA
| | - Benoit H Mulsant
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada
| | - Aristotle N Voineskos
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, M5T 1R8, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada.
| | - Daniel Felsky
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada.
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Rotman Research Institute, Baycrest Hospital, Toronto, ON, M6A 2E1, Canada.
| |
Collapse
|
28
|
Ganji-Arjenaki M, Kamali Z, Sardari S, de Borst M, Snieder H, Vaez A. Prioritization of Kidney Cell Types Highlights Myofibroblast Cells in Regulating Human Blood Pressure. Kidney Int Rep 2024; 9:1849-1859. [PMID: 38899223 PMCID: PMC11184402 DOI: 10.1016/j.ekir.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction Blood pressure (BP) is a highly heritable trait with over 2000 underlying genomic loci identified to date. Although the kidney plays a key role, little is known about specific cell types involved in the genetic regulation of BP. Methods Here, we applied stratified linkage disequilibrium score (LDSC) regression to connect BP genome-wide association studies (GWAS) results to specific cell types of the mature human kidney. We used the largest single-stage BP genome-wide analysis to date, including up to 1,028,980 adults of European ancestry, and single-cell transcriptomic data from 14 mature human kidneys, with mean age of 41 years. Results Our analyses prioritized myofibroblasts and endothelial cells, among the total of 33 annotated cell type, as specifically involved in BP regulation (P < 0.05/33, i.e., 0.001515). Enrichment of heritability for systolic BP (SBP) was observed in myofibroblast cells in mature human kidney cortex, and enrichment of heritability for diastolic BP (DBP) was observed in descending vasa recta and peritubular capillary endothelial cells as well as stromal myofibroblast cells. The new finding of myofibroblast, the significant cell type for both BP traits, was consistent in 8 replication efforts using 7 sets of independent data, including in human fetal kidney, in East-Asian (EAS) ancestry, using mouse single-cell RNA sequencing (scRNA-seq) data, and when using another prioritization method. Conclusion Our findings provide a solid basis for follow-up studies to further identify genes and mechanisms in myofibroblast cells that underlie the regulation of BP.
Collapse
Affiliation(s)
- Mahboube Ganji-Arjenaki
- Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zoha Kamali
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Soroush Sardari
- Drug Design and Bioinformatics Unit, Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Martin de Borst
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Ahmad Vaez
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Bioinformatics, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
29
|
Deng YT, Wu BS, Yang L, He XY, Kang JJ, Liu WS, Li ZY, Wu XR, Zhang YR, Chen SD, Ge YJ, Huang YY, Feng JF, Zhu Y, Dong Q, Mao Y, Cheng W, Yu JT. Large-scale whole-exome sequencing of neuropsychiatric diseases and traits in 350,770 adults. Nat Hum Behav 2024; 8:1194-1208. [PMID: 38589703 DOI: 10.1038/s41562-024-01861-4] [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: 10/06/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024]
Abstract
While numerous genomic loci have been identified for neuropsychiatric conditions, the contribution of protein-coding variants has yet to be determined. Here we conducted a large-scale whole-exome-sequencing study to interrogate the impact of protein-coding variants on 46 neuropsychiatric diseases and 23 traits in 350,770 adults from the UK Biobank. Twenty new genes were associated with neuropsychiatric diseases through coding variants, among which 16 genes had impacts on the longitudinal risks of diseases. Thirty new genes were associated with neuropsychiatric traits, with SYNGAP1 showing pleiotropic effects across cognitive function domains. Pairwise estimation of genetic correlations at the coding-variant level highlighted shared genetic associations among pairs of neurodegenerative diseases and mental disorders. Lastly, a comprehensive multi-omics analysis suggested that alterations in brain structures, blood proteins and inflammation potentially contribute to the gene-phenotype linkages. Overall, our findings characterized a compendium of protein-coding variants for future research on the biology and therapeutics of neuropsychiatric phenotypes.
Collapse
Affiliation(s)
- Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ze-Yu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, UK
| | - Ying Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital Fudan University, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Zhejiang, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Toikumo S, Jennings MV, Pham BK, Lee H, Mallard TT, Bianchi SB, Meredith JJ, Vilar-Ribó L, Xu H, Hatoum AS, Johnson EC, Pazdernik VK, Jinwala Z, Pakala SR, Leger BS, Niarchou M, Ehinmowo M, Jenkins GD, Batzler A, Pendegraft R, Palmer AA, Zhou H, Biernacka JM, Coombes BJ, Gelernter J, Xu K, Hancock DB, Cox NJ, Smoller JW, Davis LK, Justice AC, Kranzler HR, Kember RL, Sanchez-Roige S. Multi-ancestry meta-analysis of tobacco use disorder identifies 461 potential risk genes and reveals associations with multiple health outcomes. Nat Hum Behav 2024; 8:1177-1193. [PMID: 38632388 PMCID: PMC11199106 DOI: 10.1038/s41562-024-01851-6] [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: 03/24/2023] [Accepted: 02/21/2024] [Indexed: 04/19/2024]
Abstract
Tobacco use disorder (TUD) is the most prevalent substance use disorder in the world. Genetic factors influence smoking behaviours and although strides have been made using genome-wide association studies to identify risk variants, most variants identified have been for nicotine consumption, rather than TUD. Here we leveraged four US biobanks to perform a multi-ancestral meta-analysis of TUD (derived via electronic health records) in 653,790 individuals (495,005 European, 114,420 African American and 44,365 Latin American) and data from UK Biobank (ncombined = 898,680). We identified 88 independent risk loci; integration with functional genomic tools uncovered 461 potential risk genes, primarily expressed in the brain. TUD was genetically correlated with smoking and psychiatric traits from traditionally ascertained cohorts, externalizing behaviours in children and hundreds of medical outcomes, including HIV infection, heart disease and pain. This work furthers our biological understanding of TUD and establishes electronic health records as a source of phenotypic information for studying the genetics of TUD.
Collapse
Affiliation(s)
- Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mariela V Jennings
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Benjamin K Pham
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Hyunjoon Lee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Travis T Mallard
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Sevim B Bianchi
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - John J Meredith
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - 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
| | - Heng Xu
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Hatoum
- Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Emma C Johnson
- Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Zeal Jinwala
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shreya R Pakala
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Brittany S Leger
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Program in Biomedical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Maria Niarchou
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
| | | | - Greg D Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Richard Pendegraft
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Brandon J Coombes
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Ke Xu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | | | - Nancy J Cox
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Lea K Davis
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amy C Justice
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Public Health, New Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Henry R Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rachel L Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
31
|
Jasper E, Hellwege J, Greene C, Edwards TL, Edwards DV. Genomic Insights into Gestational Weight Gain: Uncovering Tissue-Specific Mechanisms and Pathways. RESEARCH SQUARE 2024:rs.3.rs-4427250. [PMID: 38854080 PMCID: PMC11160900 DOI: 10.21203/rs.3.rs-4427250/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Increasing gestational weight gain (GWG) is linked to adverse outcomes in pregnant persons and their children. The Early Growth Genetics (EGG) Consortium identified previously genetic variants that could contribute to early, late, and total GWG from fetal and maternal genomes. However, the biologic mechanisms and tissue-Specificity of these variants in GWG is unknown. We evaluated the association between genetically predicted gene expression in five relevant maternal (subcutaneous and visceral adipose, breast, uterus, and whole blood) from GTEx (v7) and fetal (placenta) tissues and early, late, and total GWG using S-PrediXcan. We tested enrichment of pre-defined biological pathways for nominally (P < 0.05) significant associations using the GENE2FUNC module from Functional Mapping and Annotation of Genome-Wide Association Studies. After multiple testing correction, we did not find significant associations between maternal and fetal gene expression and early, late, or total GWG. There was significant enrichment of several biological pathways, including metabolic processes, secretion, and intracellular transport, among nominally significant genes from the maternal analyses (false discovery rate p-values: 0.016 to 9.37×10). Enriched biological pathways varied across pregnancy. Though additional research is necessary, these results indicate that diverse biological pathways are likely to impact GWG, with their influence varying by tissue and weeks of gestation.
Collapse
Affiliation(s)
| | | | | | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center
| | | |
Collapse
|
32
|
Davis C, Khan Y, Toikumo S, Jinwala Z, Boomsma D, Levey D, Gelernter J, Kember R, Kranzler H. A Multivariate Genome-Wide Association Study Reveals Neural Correlates and Common Biological Mechanisms of Psychopathology Spectra. RESEARCH SQUARE 2024:rs.3.rs-4228593. [PMID: 38659902 PMCID: PMC11042423 DOI: 10.21203/rs.3.rs-4228593/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
There is considerable comorbidity across externalizing and internalizing behavior dimensions of psychopathology. We applied genomic structural equation modeling (gSEM) to genome-wide association study (GWAS) summary statistics to evaluate the factor structure of externalizing and internalizing psychopathology across 16 traits and disorders among European-ancestry individuals (n's = 16,400 to 1,074,629). We conducted GWAS on factors derived from well-fitting models. Downstream analyses served to identify biological mechanisms, explore drug repurposing targets, estimate genetic overlap between the externalizing and internalizing spectra, and evaluate causal effects of psychopathology liability on physical health. Both a correlated factors model, comprising two factors of externalizing and internalizing risk, and a higher-order single-factor model of genetic effects contributing to both spectra demonstrated acceptable t. GWAS identified 409 lead single nucleotide polymorphisms (SNPs) associated with externalizing and 85 lead SNPs associated with internalizing, while the second-order GWAS identified 256 lead SNPs contributing to broad psychopathology risk. In bivariate causal mixture models, nearly all externalizing and internalizing causal variants overlapped, despite a genetic correlation of only 0.37 (SE = 0.02) between them. Externalizing genes showed cell-type specific expression in GABAergic, cortical, and hippocampal neurons, and internalizing genes were associated with reduced subcallosal cortical volume, providing insight into the neurobiological underpinnings of psychopathology. Genetic liability for externalizing, internalizing, and broad psychopathology exerted causal effects on pain, general health, cardiovascular diseases, and chronic illnesses. These findings underscore the complex genetic architecture of psychopathology, identify potential biological pathways for the externalizing and internalizing spectra, and highlight the physical health burden of psychiatric comorbidity.
Collapse
Affiliation(s)
| | - Yousef Khan
- University of Pennsylvania Perelman School of Medicine
| | | | - Zeal Jinwala
- University of Pennsylvania Perelman School of Medicine
| | - D Boomsma
- Vrije Universiteit Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
33
|
Toikumo S, Vickers-Smith R, Jinwala Z, Xu H, Saini D, Hartwell EE, Pavicic M, Sullivan KA, Xu K, Jacobson DA, Gelernter J, Rentsch CT, Stahl E, Cheatle M, Zhou H, Waxman SG, Justice AC, Kember RL, Kranzler HR. A multi-ancestry genetic study of pain intensity in 598,339 veterans. Nat Med 2024; 30:1075-1084. [PMID: 38429522 DOI: 10.1038/s41591-024-02839-5] [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: 03/08/2023] [Accepted: 01/27/2024] [Indexed: 03/03/2024]
Abstract
Chronic pain is a common problem, with more than one-fifth of adult Americans reporting pain daily or on most days. It adversely affects the quality of life and imposes substantial personal and economic costs. Efforts to treat chronic pain using opioids had a central role in precipitating the opioid crisis. Despite an estimated heritability of 25-50%, the genetic architecture of chronic pain is not well-characterized, in part because studies have largely been limited to samples of European ancestry. To help address this knowledge gap, we conducted a cross-ancestry meta-analysis of pain intensity in 598,339 participants in the Million Veteran Program, which identified 126 independent genetic loci, 69 of which are new. Pain intensity was genetically correlated with other pain phenotypes, level of substance use and substance use disorders, other psychiatric traits, education level and cognitive traits. Integration of the genome-wide association studies findings with functional genomics data shows enrichment for putatively causal genes (n = 142) and proteins (n = 14) expressed in brain tissues, specifically in GABAergic neurons. Drug repurposing analysis identified anticonvulsants, β-blockers and calcium-channel blockers, among other drug groups, as having potential analgesic effects. Our results provide insights into key molecular contributors to the experience of pain and highlight attractive drug targets.
Collapse
Affiliation(s)
- Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rachel Vickers-Smith
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Epidemiology and Environmental Health, University of Kentucky College of Public Health, Lexington, KY, USA
| | - Zeal Jinwala
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Heng Xu
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divya Saini
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Emily E Hartwell
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mirko Pavicic
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Kyle A Sullivan
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Ke Xu
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Daniel A Jacobson
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Joel Gelernter
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Christopher T Rentsch
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- London School of Hygiene & Tropical Medicine, London, UK
| | - Eli Stahl
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Martin Cheatle
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Hang Zhou
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Section of Biomedical Informatics and Data Science, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Amy C Justice
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale University School of Public Health, New Haven, CT, USA
| | - Rachel L Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Henry R Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA.
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Elliott A, Walters RK, Pirinen M, Kurki M, Junna N, Goldstein JI, Reeve MP, Siirtola H, Lemmelä SM, Turley P, Lahtela E, Mehtonen J, Reis K, Elnahas AG, Reigo A, Palta P, Esko T, Mägi R, Palotie A, Daly MJ, Widén E. Distinct and shared genetic architectures of gestational diabetes mellitus and type 2 diabetes. Nat Genet 2024; 56:377-382. [PMID: 38182742 PMCID: PMC10937370 DOI: 10.1038/s41588-023-01607-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 11/07/2023] [Indexed: 01/07/2024]
Abstract
Gestational diabetes mellitus (GDM) is a common metabolic disorder affecting more than 16 million pregnancies annually worldwide1,2. GDM is related to an increased lifetime risk of type 2 diabetes (T2D)1-3, with over a third of women developing T2D within 15 years of their GDM diagnosis. The diseases are hypothesized to share a genetic predisposition1-7, but few studies have sought to uncover the genetic underpinnings of GDM. Most studies have evaluated the impact of T2D loci only8-10, and the three prior genome-wide association studies of GDM11-13 have identified only five loci, limiting the power to assess to what extent variants or biological pathways are specific to GDM. We conducted the largest genome-wide association study of GDM to date in 12,332 cases and 131,109 parous female controls in the FinnGen study and identified 13 GDM-associated loci, including nine new loci. Genetic features distinct from T2D were identified both at the locus and genomic scale. Our results suggest that the genetics of GDM risk falls into the following two distinct categories: one part conventional T2D polygenic risk and one part predominantly influencing mechanisms disrupted in pregnancy. Loci with GDM-predominant effects map to genes related to islet cells, central glucose homeostasis, steroidogenesis and placental expression.
Collapse
Grants
- R00 AG062787 NIA NIH HHS
- R01 MH101244 NIMH NIH HHS
- A.E. was a research Scholar supported by Sarnoff Cardiovascular Research Foundation
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- Academy of Finland (Suomen Akatemia)
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- The FinnGen project is funded by two grants from Business Finland (HUS 4685/31/2016 and UH 4386/31/2016) and by eleven industry partners (AbbVie Inc, AstraZeneca UK Ltd, Biogen MA Inc, Celgene Corporation, Celgene International II Sàrl, Genentech Inc, Merck Sharp & Dohme Corp, Pfizer Inc., GlaxoSmithKline, Sanofi, Maze Therapeutics Inc., Janssen Biotech Inc).
- EstBB GWAS analysis is supported by research funding from the Estonian Research Council: Team grant PRG1291 and PRG1911.
Collapse
Affiliation(s)
- Amanda Elliott
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Mitja Kurki
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Nella Junna
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Jacqueline I Goldstein
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mary Pat Reeve
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Harri Siirtola
- TAUCHI Research Center, Faculty of Information Technology and Communication Sciences (ITC), Tampere University, Tampere, Finland
| | - Susanna M Lemmelä
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Patrick Turley
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
- Department of Economics, University of Southern California, Los Angeles, CA, USA
| | - Elisa Lahtela
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Juha Mehtonen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Kadri Reis
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - Anu Reigo
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Priit Palta
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Aarno Palotie
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland.
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
35
|
Kim H, Ahn Y, Yoon J, Jung K, Kim S, Shim I, Park TH, Ko H, Jung SH, Kim J, Park S, Lee DJ, Choi S, Cha S, Kim B, Cho MY, Cho H, Kim DS, Jang Y, Ihm HK, Park WY, Bakhshi H, O Connell KS, Andreassen OA, Kendler KS, Myung W, Won HH. Genome-wide association analyses using machine learning-based phenotyping reveal genetic architecture of occupational creativity and overlap with psychiatric disorders. Psychiatry Res 2024; 333:115753. [PMID: 38335777 DOI: 10.1016/j.psychres.2024.115753] [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: 09/03/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
Creativity is known to be heritable and exhibits familial aggregation with psychiatric disorders; however, the complex nature of their relationship has not been well-established. In the present study, we demonstrate that using an expanded and validated machine learning (ML)-based phenotyping of occupational creativity (OC) can allow us to further understand the trait of creativity, which was previously difficult to define and study. We conducted the largest genome-wide association study (GWAS) on OC with 241,736 participants from the UK Biobank and identified 25 lead variants that have not yet been reported and three candidate causal genes that were previously associated with educational attainment and psychiatric disorders. We found extensive genetic overlap between OC and psychiatric disorders with mixed effect direction through various post-GWAS analyses, including the bivariate causal mixture model. In addition, we discovered a strongly genetic correlation between our original GWAS and the GWAS adjusted for education years (rg = 0.95). Our GWAS analysis via ML-based phenotyping contributes to the understanding of the genetic architecture of creativity, which may inform genetic discovery and genetic prediction in human cognition and psychiatric disorders.
Collapse
Affiliation(s)
- Hyejin Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Yeeun Ahn
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Joohyun Yoon
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyeongmin Jung
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soyeon Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Injeong Shim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Tae Hwan Park
- Department of Plastic Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwasung, South Korea
| | - Hyunwoong Ko
- Interdisciplinary Program in Cognitive Science, Seoul National University, Seoul, South Korea; Department of Psychiatry, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea; Dental Research Institute, Seoul National University School of Dentistry, Seoul, South Korea
| | - Sang-Hyuk Jung
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Jaeyoung Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Sanghyeon Park
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Dong June Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| | - Sunho Choi
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea
| | - Soojin Cha
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Beomsu Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Min Young Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Hyunbin Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Dan Say Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Yoonjeong Jang
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea; Department of Health Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Hong Kyu Ihm
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hasan Bakhshi
- Creative Industries Policy and Evidence Centre, Nesta, London, United Kingdom
| | - Kevin S O Connell
- Norwegian Center for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Center for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Kenneth S Kendler
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea; Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea; Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
36
|
Chen TT, Kim J, Lam M, Chuang YF, Chiu YL, Lin SC, Jung SH, Kim B, Kim S, Cho C, Shim I, Park S, Ahn Y, Okbay A, Jang H, Kim HJ, Seo SW, Park WY, Ge T, Huang H, Feng YCA, Lin YF, Myung W, Chen CY, Won HH. Shared genetic architectures of educational attainment in East Asian and European populations. Nat Hum Behav 2024; 8:562-575. [PMID: 38182883 PMCID: PMC10963262 DOI: 10.1038/s41562-023-01781-9] [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: 03/19/2023] [Accepted: 11/09/2023] [Indexed: 01/07/2024]
Abstract
Educational attainment (EduYears), a heritable trait often used as a proxy for cognitive ability, is associated with various health and social outcomes. Previous genome-wide association studies (GWASs) on EduYears have been focused on samples of European (EUR) genetic ancestries. Here we present the first large-scale GWAS of EduYears in people of East Asian (EAS) ancestry (n = 176,400) and conduct a cross-ancestry meta-analysis with EduYears GWAS in people of EUR ancestry (n = 766,345). EduYears showed a high genetic correlation and power-adjusted transferability ratio between EAS and EUR. We also found similar functional enrichment, gene expression enrichment and cross-trait genetic correlations between two populations. Cross-ancestry fine-mapping identified refined credible sets with a higher posterior inclusion probability than single population fine-mapping. Polygenic prediction analysis in four independent EAS and EUR cohorts demonstrated transferability between populations. Our study supports the need for further research on diverse ancestries to increase our understanding of the genetic basis of educational attainment.
Collapse
Affiliation(s)
- Tzu-Ting Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jaeyoung Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Max Lam
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
- Division of Psychiatry Research, the Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Research Division Institute of Mental Health Singapore, Singapore, Singapore
| | - Yi-Fang Chuang
- Institute of Public Health and International Health Program, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Ling Chiu
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan City, Taiwan
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Shu-Chin Lin
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beomsu Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Soyeon Kim
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Chamlee Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Injeong Shim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Sanghyeon Park
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Yeeun Ahn
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Aysu Okbay
- Department of Economics, School of Business and Economics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Hyemin Jang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
| | - Hee Jin Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Tian Ge
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yen-Chen Anne Feng
- Institute of Health Data Analytics and Statistics, College of Public Health, National Taiwan University, Taipei City, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei City, Taiwan
| | - Yen-Feng Lin
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan.
- Department of Public Health and Medical Humanities, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea.
| | | | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea.
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
37
|
Buyukcelebi K, Duval AJ, Abdula F, Elkafas H, Seker-Polat F, Adli M. Integrating leiomyoma genetics, epigenomics, and single-cell transcriptomics reveals causal genetic variants, genes, and cell types. Nat Commun 2024; 15:1169. [PMID: 38326302 PMCID: PMC10850163 DOI: 10.1038/s41467-024-45382-0] [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: 02/03/2023] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
Uterine fibroids (UF), that can disrupt normal uterine function and cause significant physical and psychological health problems, are observed in nearly 70% of women of reproductive age. Although heritable genetics is a significant risk factor, specific genetic variations and gene targets causally associated with UF are poorly understood. Here, we performed a meta-analysis on existing fibroid genome-wide association studies (GWAS) and integrated the identified risk loci and potentially causal single nucleotide polymorphisms (SNPs) with epigenomics, transcriptomics, 3D chromatin organization from diverse cell types as well as primary UF patient's samples. This integrative analysis identifies 24 UF-associated risk loci that potentially target 394 genes, of which 168 are differentially expressed in UF tumors. Critically, integrating this data with single-cell gene expression data from UF patients reveales the causal cell types with aberrant expression of these target genes. Lastly, CRISPR-based epigenetic repression (dCas9-KRAB) or activation (dCas9-p300) in a UF disease-relevant cell type further refines and narrows down the potential gene targets. Our findings and the methodological approach indicate the effectiveness of integrating multi-omics data with locus-specific epigenetic editing approaches for identifying gene- and celt type-targets of disease-relevant risk loci.
Collapse
Affiliation(s)
- Kadir Buyukcelebi
- Department of Obstetrics and Gynecology, Robert Lurie Comprehensive Cancer Center, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Alexander J Duval
- Department of Obstetrics and Gynecology, Robert Lurie Comprehensive Cancer Center, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Fatih Abdula
- Department of Obstetrics and Gynecology, Robert Lurie Comprehensive Cancer Center, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Hoda Elkafas
- Department of Obstetrics and Gynecology, Robert Lurie Comprehensive Cancer Center, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Fidan Seker-Polat
- Department of Obstetrics and Gynecology, Robert Lurie Comprehensive Cancer Center, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Mazhar Adli
- Department of Obstetrics and Gynecology, Robert Lurie Comprehensive Cancer Center, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA.
| |
Collapse
|
38
|
Akinyemi RO, Tiwari HK, Srinivasasainagendra V, Akpa O, Sarfo FS, Akpalu A, Wahab K, Obiako R, Komolafe M, Owolabi L, Osaigbovo GO, Mamaeva OA, Halloran BA, Akinyemi J, Lackland D, Obiabo OY, Sunmonu T, Chukwuonye II, Arulogun O, Jenkins C, Adeoye A, Agunloye A, Ogah OS, Ogbole G, Fakunle A, Uvere E, Coker MM, Okekunle A, Asowata O, Diala S, Ogunronbi M, Adeleye O, Laryea R, Tagge R, Adeniyi S, Adusei N, Oguike W, Olowoyo P, Adebajo O, Olalere A, Oladele O, Yaria J, Fawale B, Ibinaye P, Oyinloye O, Mensah Y, Oladimeji O, Akpalu J, Calys-Tagoe B, Dambatta HA, Ogunniyi A, Kalaria R, Arnett D, Rotimi C, Ovbiagele B, Owolabi MO. Novel functional insights into ischemic stroke biology provided by the first genome-wide association study of stroke in indigenous Africans. Genome Med 2024; 16:25. [PMID: 38317187 PMCID: PMC10840175 DOI: 10.1186/s13073-023-01273-5] [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: 06/07/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND African ancestry populations have the highest burden of stroke worldwide, yet the genetic basis of stroke in these populations is obscure. The Stroke Investigative Research and Educational Network (SIREN) is a multicenter study involving 16 sites in West Africa. We conducted the first-ever genome-wide association study (GWAS) of stroke in indigenous Africans. METHODS Cases were consecutively recruited consenting adults (aged > 18 years) with neuroimaging-confirmed ischemic stroke. Stroke-free controls were ascertained using a locally validated Questionnaire for Verifying Stroke-Free Status. DNA genotyping with the H3Africa array was performed, and following initial quality control, GWAS datasets were imputed into the NIH Trans-Omics for Precision Medicine (TOPMed) release2 from BioData Catalyst. Furthermore, we performed fine-mapping, trans-ethnic meta-analysis, and in silico functional characterization to identify likely causal variants with a functional interpretation. RESULTS We observed genome-wide significant (P-value < 5.0E-8) SNPs associations near AADACL2 and miRNA (MIR5186) genes in chromosome 3 after adjusting for hypertension, diabetes, dyslipidemia, and cardiac status in the base model as covariates. SNPs near the miRNA (MIR4458) gene in chromosome 5 were also associated with stroke (P-value < 1.0E-6). The putative genes near AADACL2, MIR5186, and MIR4458 genes were protective and novel. SNPs associations with stroke in chromosome 2 were more than 77 kb from the closest gene LINC01854 and SNPs in chromosome 7 were more than 116 kb to the closest gene LINC01446 (P-value < 1.0E-6). In addition, we observed SNPs in genes STXBP5-AS1 (chromosome 6), GALTN9 (chromosome 12), FANCA (chromosome 16), and DLGAP1 (chromosome 18) (P-value < 1.0E-6). Both genomic regions near genes AADACL2 and MIR4458 remained significant following fine mapping. CONCLUSIONS Our findings identify potential roles of regulatory miRNA, intergenic non-coding DNA, and intronic non-coding RNA in the biology of ischemic stroke. These findings reveal new molecular targets that promise to help close the current gaps in accurate African ancestry-based genetic stroke's risk prediction and development of new targeted interventions to prevent or treat stroke.
Collapse
Affiliation(s)
- Rufus O Akinyemi
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Center for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Onoja Akpa
- Center for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Fred S Sarfo
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Albert Akpalu
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Kolawole Wahab
- Department of Medicine, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Reginald Obiako
- Department of Medicine, Ahmadu Bello University, Zaria, Nigeria
| | - Morenikeji Komolafe
- Department of Medicine, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | - Lukman Owolabi
- Department of Medicine, Aminu Kano Teaching Hospital, Kano, Nigeria
| | | | - Olga A Mamaeva
- Department of Epidemiology, School of Public Health University of Alabama at Birmingham, Birmingham, USA
| | - Brian A Halloran
- Department of Pediatrics, Volker Hall University of Alabama at Birmingham, Birmingham, USA
| | - Joshua Akinyemi
- Department of Epidemiology and Medical Statistics, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Olugbo Y Obiabo
- Delta State University/Delta State University Teaching Hospital, Oghara, Nigeria
| | - Taofik Sunmonu
- Department of Medicine, Federal Medical Centre, Ondo State, Owo, Nigeria
| | - Innocent I Chukwuonye
- Department of Medicine, Federal Medical Centre Umuahia, Abia State, Umuahia, Nigeria
| | - Oyedunni Arulogun
- Department of Health Education, Faculty of Public Health, University of Ibadan, Ibadan, Nigeria
| | | | - Abiodun Adeoye
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Atinuke Agunloye
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Okechukwu S Ogah
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Godwin Ogbole
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adekunle Fakunle
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Public Health, College of Health Sciences, Osun State University, Osogbo, Nigeria
| | - Ezinne Uvere
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Motunrayo M Coker
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Genetics and Cell Biology Unit, Department of Zoology, Faculty of Science, University of Ibadan, Ibadan, Nigeria
| | - Akinkunmi Okekunle
- Department of Food and Nutrition, Seoul National University, Seoul, South Korea
| | - Osahon Asowata
- Department of Epidemiology and Medical Statistics, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Samuel Diala
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Mayowa Ogunronbi
- Department of Medicine, Federal Medical Centre, Abeokuta, Nigeria
| | - Osi Adeleye
- Department of Medicine, Federal Medical Centre, Abeokuta, Nigeria
| | - Ruth Laryea
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Raelle Tagge
- Weill Institute for Neurosciences, School of Medicine, University of California San-Francisco, San Francisco, USA
| | - Sunday Adeniyi
- Department of Medicine, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Nathaniel Adusei
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Wisdom Oguike
- Department of Medicine, Ahmadu Bello University, Zaria, Nigeria
| | - Paul Olowoyo
- Federal Teaching Hospital, Ido-Ekiti, Ekiti State, Nigeria
| | - Olayinka Adebajo
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Abimbola Olalere
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olayinka Oladele
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Joseph Yaria
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Bimbo Fawale
- Department of Medicine, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | - Philip Ibinaye
- Department of Medicine, Ahmadu Bello University, Zaria, Nigeria
| | - Olalekan Oyinloye
- Department of Medicine, Obafemi Awolowo University Teaching Hospital, Ile-Ife, Nigeria
| | - Yaw Mensah
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Omotola Oladimeji
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Josephine Akpalu
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Benedict Calys-Tagoe
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | - Adesola Ogunniyi
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Rajesh Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Donna Arnett
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, USA
| | - Charles Rotimi
- Center for Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, USA
| | - Bruce Ovbiagele
- Genetics and Cell Biology Unit, Department of Zoology, Faculty of Science, University of Ibadan, Ibadan, Nigeria
| | - Mayowa O Owolabi
- Center for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria.
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria.
- University College Hospital, Ibadan, Nigeria.
- Lebanese American University of Beirut, Beirut, Lebanon.
- Blossom Specialist Medical Center, Ibadan, Nigeria.
| |
Collapse
|
39
|
Carvalho NRG, He Y, Smadbeck P, Flannick J, Mercader JM, Udler M, Manrai AK, Moreno J, Patel CJ. Assessing the genetic contribution of cumulative behavioral factors associated with longitudinal type 2 diabetes risk highlights adiposity and the brain-metabolic axis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.30.24302019. [PMID: 38352440 PMCID: PMC10863013 DOI: 10.1101/2024.01.30.24302019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
While genetic factors, behavior, and environmental exposures form a complex web of interrelated associations in type 2 diabetes (T2D), their interaction is poorly understood. Here, using data from ~500K participants of the UK Biobank, we identify the genetic determinants of a "polyexposure risk score" (PXS) a new risk factor that consists of an accumulation of 25 associated individual-level behaviors and environmental risk factors that predict longitudinal T2D incidence. PXS-T2D had a non-zero heritability (h2 = 0.18) extensive shared genetic architecture with established clinical and biological determinants of T2D, most prominently with body mass index (genetic correlation [rg] = 0.57) and Homeostatic Model Assessment for Insulin Resistance (rg = 0.51). Genetic loci associated with PXS-T2D were enriched for expression in the brain. Biobank scale data with genetic information illuminates how complex and cumulative exposures and behaviors as a whole impact T2D risk but whose biology have been elusive in genome-wide studies of T2D.
Collapse
Affiliation(s)
- Nuno R. G. Carvalho
- School of Biological Sciences; Georgia Institute of Technology; Atlanta, GA, 30332, USA
| | - Yixuan He
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Patrick Smadbeck
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Jason Flannick
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Josep M. Mercader
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Miriam Udler
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
| | - Arjun K Manrai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Jordi Moreno
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA, 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Chirag J. Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
40
|
Yao S, Harder A, Darki F, Chang YW, Li A, Nikouei K, Volpe G, Lundström JN, Zeng J, Wray N, Lu Y, Sullivan PF, Leffler JH. Connecting genomic results for psychiatric disorders to human brain cell types and regions reveals convergence with functional connectivity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.18.24301478. [PMID: 38410450 PMCID: PMC10896415 DOI: 10.1101/2024.01.18.24301478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Understanding the temporal and spatial brain locations etiological for psychiatric disorders is essential for targeted neurobiological research. Integration of genomic insights from genome-wide association studies with single-cell transcriptomics is a powerful approach although past efforts have necessarily relied on mouse atlases. Leveraging a comprehensive atlas of the adult human brain, we prioritized cell types via the enrichment of SNP-heritabilities for brain diseases, disorders, and traits, progressing from individual cell types to brain regions. Our findings highlight specific neuronal clusters significantly enriched for the SNP-heritabilities for schizophrenia, bipolar disorder, and major depressive disorder along with intelligence, education, and neuroticism. Extrapolation of cell-type results to brain regions reveals important patterns for schizophrenia with distinct subregions in the hippocampus and amygdala exhibiting the highest significance. Cerebral cortical regions display similar enrichments despite the known prefrontal dysfunction in those with schizophrenia highlighting the importance of subcortical connectivity. Using functional MRI connectivity from cases with schizophrenia and neurotypical controls, we identified brain networks that distinguished cases from controls that also confirmed involvement of the central and lateral amygdala, hippocampal body, and prefrontal cortex. Our findings underscore the value of single-cell transcriptomics in decoding the polygenicity of psychiatric disorders and offer a promising convergence of genomic, transcriptomic, and brain imaging modalities toward common biological targets.
Collapse
Affiliation(s)
- Shuyang Yao
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Arvid Harder
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fahimeh Darki
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yu-Wei Chang
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Ang Li
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Kasra Nikouei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Giovanni Volpe
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Johan N Lundström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Jian Zeng
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Naomi Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Jens Hjerling Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Hughes O, Bentley AR, Breeze CE, Aguet F, Xu X, Nadkarni G, Sun Q, Lin BM, Gilliland T, Meyer MC, Du J, Raffield LM, Kramer H, Morton RW, Gouveia MH, Atkinson EG, Valladares-Salgado A, Wacher-Rodarte N, Dueker ND, Guo X, Hai Y, Adeyemo A, Best LG, Cai J, Chen G, Chong M, Doumatey A, Eales J, Goodarzi MO, Ipp E, Irvin MR, Jiang M, Jones AC, Kooperberg C, Krieger JE, Lange EM, Lanktree MB, Lash JP, Lotufo PA, Loos RJF, Ha My VT, Peralta-Romero J, Qi L, Raffel LJ, Rich SS, Rodriquez EJ, Tarazona-Santos E, Taylor KD, Umans JG, Wen J, Young BA, Yu Z, Zhang Y, Ida Chen YD, Rundek T, Rotter JI, Cruz M, Fornage M, Lima-Costa MF, Pereira AC, Paré G, Natarajan P, Cole SA, Carson AP, Lange LA, Li Y, Perez-Stable EJ, Do R, Charchar FJ, Tomaszewski M, Mychaleckyj JC, Rotimi C, Morris AP, Franceschini N. Genome-wide study investigating effector genes and polygenic prediction for kidney function in persons with ancestry from Africa and the Americas. CELL GENOMICS 2024; 4:100468. [PMID: 38190104 PMCID: PMC10794846 DOI: 10.1016/j.xgen.2023.100468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/31/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024]
Abstract
Chronic kidney disease is a leading cause of death and disability globally and impacts individuals of African ancestry (AFR) or with ancestry in the Americas (AMS) who are under-represented in genome-wide association studies (GWASs) of kidney function. To address this bias, we conducted a large meta-analysis of GWASs of estimated glomerular filtration rate (eGFR) in 145,732 AFR and AMS individuals. We identified 41 loci at genome-wide significance (p < 5 × 10-8), of which two have not been previously reported in any ancestry group. We integrated fine-mapped loci with epigenomic and transcriptomic resources to highlight potential effector genes relevant to kidney physiology and disease, and reveal key regulatory elements and pathways involved in renal function and development. We demonstrate the varying but increased predictive power offered by a multi-ancestry polygenic score for eGFR and highlight the importance of population diversity in GWASs and multi-omics resources to enhance opportunities for clinical translation for all.
Collapse
Affiliation(s)
- Odessica Hughes
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles E Breeze
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, MD, USA; UCL Cancer Institute, University College London, London, UK
| | - Francois Aguet
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, UK
| | - Girish Nadkarni
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bridget M Lin
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas Gilliland
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mariah C Meyer
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jiawen Du
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Holly Kramer
- Division of Nephrology and Hypertension, Loyola University Chicago, Maywood, IL, USA
| | - Robert W Morton
- Population Health Research Institute, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Mateus H Gouveia
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth G Atkinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Adan Valladares-Salgado
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Niels Wacher-Rodarte
- Unidad de Investigación Médica en Epidemiologia Clinica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Nicole D Dueker
- John P Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Yang Hai
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lyle G Best
- Missouri Breaks Industries Research Inc., Eagle Butte, SD, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Chong
- Population Health Research Institute, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Ayo Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - James Eales
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, UK
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eli Ipp
- Division of Endocrinology and Metabolism, Department of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marguerite Ryan Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Minzhi Jiang
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alana C Jones
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jose E Krieger
- Laboratório de Genética e Cardiologia Molecular do Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ethan M Lange
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew B Lanktree
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - James P Lash
- Division of Nephrology, Department of Medicine, University of Illinois, Chicago, IL, USA
| | - Paulo A Lotufo
- Center for Clinical and Epidemiological Research, Hospital Universitário, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Ruth J F Loos
- The Charles Bronfman Institute of 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
| | - Vy Thi Ha My
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jesús Peralta-Romero
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Lihong Qi
- Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, CA, USA
| | - Leslie J Raffel
- Department of Pediatrics, Genetic and Genomic Medicine, University of California, Irvine, Irvine, CA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Erik J Rodriquez
- Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eduardo Tarazona-Santos
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Jason G Umans
- MedStar Health Research Institute, Hyattsville MD and Georgetown-Howard Universities Center for Clinical and Translational Science, Washington, DC, USA
| | - Jia Wen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bessie A Young
- University of Washington School of Medicine, Seattle, WA, USA; Office of Healthcare Equity, UW Justice, Equity, Diversity, and Inclusion Center for Transformational Research (UW JEDI-CTR), University of Washington, Seattle, WA, USA; Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA; Kidney Research Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Zhi Yu
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Ying Zhang
- Center for American Indian Health Research, Department of Biostatistics and Epidemiology, Hudson College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma, OK, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Tanja Rundek
- Department of Neurology, Epidemiology and Public Health, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | | | - Alexandre C Pereira
- Laboratório de Genética e Cardiologia Molecular do Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Aging Division, Brigham Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Guillaume Paré
- Population Health Research Institute, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Shelley A Cole
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Leslie A Lange
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eliseo J Perez-Stable
- National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Ron Do
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fadi J Charchar
- School of Science, Psychology and Sport, Federation University, Ballarat, VIC, Australia; Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; Department of Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, UK; Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Josyf C Mychaleckyj
- Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, CA, USA
| | - Charles Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK.
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
42
|
Hamel AR, Yan W, Rouhana JM, Monovarfeshani A, Jiang X, Mehta PA, Advani J, Luo Y, Liang Q, Rajasundaram S, Shrivastava A, Duchinski K, Mantena S, Wang J, van Zyl T, Pasquale LR, Swaroop A, Gharahkhani P, Khawaja AP, MacGregor S, Chen R, Vitart V, Sanes JR, Wiggs JL, Segrè AV. Integrating genetic regulation and single-cell expression with GWAS prioritizes causal genes and cell types for glaucoma. Nat Commun 2024; 15:396. [PMID: 38195602 PMCID: PMC10776627 DOI: 10.1038/s41467-023-44380-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Primary open-angle glaucoma (POAG), characterized by retinal ganglion cell death, is a leading cause of irreversible blindness worldwide. However, its molecular and cellular causes are not well understood. Elevated intraocular pressure (IOP) is a major risk factor, but many patients have normal IOP. Colocalization and Mendelian randomization analysis of >240 POAG and IOP genome-wide association study (GWAS) loci and overlapping expression and splicing quantitative trait loci (e/sQTLs) in 49 GTEx tissues and retina prioritizes causal genes for 60% of loci. These genes are enriched in pathways implicated in extracellular matrix organization, cell adhesion, and vascular development. Analysis of single-nucleus RNA-seq of glaucoma-relevant eye tissues reveals that the POAG and IOP colocalizing genes and genome-wide associations are enriched in specific cell types in the aqueous outflow pathways, retina, optic nerve head, peripapillary sclera, and choroid. This study nominates IOP-dependent and independent regulatory mechanisms, genes, and cell types that may contribute to POAG pathogenesis.
Collapse
Affiliation(s)
- Andrew R Hamel
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Wenjun Yan
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - John M Rouhana
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Aboozar Monovarfeshani
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Xinyi Jiang
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Puja A Mehta
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jayshree Advani
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MA, USA
| | - Yuyang Luo
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Qingnan Liang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Skanda Rajasundaram
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Centre for Evidence-Based Medicine, University of Oxford, Oxford, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Arushi Shrivastava
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Katherine Duchinski
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Bioinformatics and Integrative Genomics (BIG) PhD Program, Harvard Medical School, Boston, MA, USA
| | - Sreekar Mantena
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Jiali Wang
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Tavé van Zyl
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
- Department of Ophthalmology and Visual Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MA, USA
| | - Puya Gharahkhani
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Anthony P Khawaja
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Janey L Wiggs
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ayellet V Segrè
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
43
|
Dai Y, Hsu YC, Fernandes BS, Zhang K, Li X, Enduru N, Liu A, Manuel AM, Jiang X, Zhao Z. Disentangling Accelerated Cognitive Decline from the Normal Aging Process and Unraveling Its Genetic Components: A Neuroimaging-Based Deep Learning Approach. J Alzheimers Dis 2024; 97:1807-1827. [PMID: 38306043 PMCID: PMC11649026 DOI: 10.3233/jad-231020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Background The progressive cognitive decline, an integral component of Alzheimer's disease (AD), unfolds in tandem with the natural aging process. Neuroimaging features have demonstrated the capacity to distinguish cognitive decline changes stemming from typical brain aging and AD between different chronological points. Objective To disentangle the normal aging effect from the AD-related accelerated cognitive decline and unravel its genetic components using a neuroimaging-based deep learning approach. Methods We developed a deep-learning framework based on a dual-loss Siamese ResNet network to extract fine-grained information from the longitudinal structural magnetic resonance imaging (MRI) data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We then conducted genome-wide association studies (GWAS) and post-GWAS analyses to reveal the genetic basis of AD-related accelerated cognitive decline. Results We used our model to process data from 1,313 individuals, training it on 414 cognitively normal people and predicting cognitive assessment for all participants. In our analysis of accelerated cognitive decline GWAS, we identified two genome-wide significant loci: APOE locus (chromosome 19 p13.32) and rs144614292 (chromosome 11 p15.1). Variant rs144614292 (G > T) has not been reported in previous AD GWA studies. It is within the intronic region of NELL1, which is expressed in neurons and plays a role in controlling cell growth and differentiation. The cell-type-specific enrichment analysis and functional enrichment of GWAS signals highlighted the microglia and immune-response pathways. Conclusions Our deep learning model effectively extracted relevant neuroimaging features and predicted individual cognitive decline. We reported a novel variant (rs144614292) within the NELL1 gene.
Collapse
Affiliation(s)
- Yulin Dai
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Yu-Chun Hsu
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Brisa S. Fernandes
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Kai Zhang
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Xiaoyang Li
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Biostatistics and Data Science, School of
Public Health, The University of Texas Health Science Center at Houston, Houston,
TX, USA
| | - Nitesh Enduru
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
| | - Andi Liu
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
| | - Astrid M. Manuel
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence for Healthcare,
School of Biomedical Informatics, The University of Texas Health Science Center at
Houston, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of
Biomedical Informatics, The University of Texas Health Science Center at Houston,
Houston, TX, USA
- Department of Epidemiology, Human Genetics and
Environmental Sciences, School of Public Health, The University of Texas Health
Science Center at Houston, Houston, TX, USA
- Department of Biomedical Informatics, Vanderbilt University
Medical enter, Nashville, TN, USA
| |
Collapse
|
44
|
Kim JJ, Vitale D, Otani DV, Lian MM, Heilbron K, Iwaki H, Lake J, Solsberg CW, Leonard H, Makarious MB, Tan EK, Singleton AB, Bandres-Ciga S, Noyce AJ, Blauwendraat C, Nalls MA, Foo JN, Mata I. Multi-ancestry genome-wide association meta-analysis of Parkinson's disease. Nat Genet 2024; 56:27-36. [PMID: 38155330 PMCID: PMC10786718 DOI: 10.1038/s41588-023-01584-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/20/2023] [Indexed: 12/30/2023]
Abstract
Although over 90 independent risk variants have been identified for Parkinson's disease using genome-wide association studies, most studies have been performed in just one population at a time. Here we performed a large-scale multi-ancestry meta-analysis of Parkinson's disease with 49,049 cases, 18,785 proxy cases and 2,458,063 controls including individuals of European, East Asian, Latin American and African ancestry. In a meta-analysis, we identified 78 independent genome-wide significant loci, including 12 potentially novel loci (MTF2, PIK3CA, ADD1, SYBU, IRS2, USP8, PIGL, FASN, MYLK2, USP25, EP300 and PPP6R2) and fine-mapped 6 putative causal variants at 6 known PD loci. By combining our results with publicly available eQTL data, we identified 25 putative risk genes in these novel loci whose expression is associated with PD risk. This work lays the groundwork for future efforts aimed at identifying PD loci in non-European populations.
Collapse
Affiliation(s)
- Jonggeol Jeffrey Kim
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Preventive Neurology Unit, Centre for Prevention Diagnosis and Detection, Wolfson Institute of Population Health, Queen Mary University of London, London, UK.
| | - Dan Vitale
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Diego Véliz Otani
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru
- Institute for Genome Sciences, University of Maryland, Baltimore, MD, USA
| | - Michelle Mulan Lian
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, A*STAR, Singapore, Singapore
| | | | - Hirotaka Iwaki
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Julie Lake
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Caroline Warly Solsberg
- Pharmaceutical Sciences and Pharmacogenomics, UCSF, San Francisco, CA, USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, UCSF, San Francisco, CA, USA
| | - Hampton Leonard
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Washington, DC, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mary B Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- UCL Movement Disorders Centre, University College London, London, UK
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Duke NUS Medical School, Singapore, Singapore
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sara Bandres-Ciga
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Alastair J Noyce
- Preventive Neurology Unit, Centre for Prevention Diagnosis and Detection, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Data Tecnica International, Washington, DC, USA.
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore.
- Genome Institute of Singapore, Agency for Science, Technology and Research, A*STAR, Singapore, Singapore.
| | - Ignacio Mata
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
45
|
Ho CY, Lee JI, Huang SP, Chen SC, Geng JH. A Genome-Wide Association Study of Metabolic Syndrome in the Taiwanese Population. Nutrients 2023; 16:77. [PMID: 38201907 PMCID: PMC10780952 DOI: 10.3390/nu16010077] [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: 11/08/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The purpose of this study was to investigate genetic factors associated with metabolic syndrome (MetS) by conducting a large-scale genome-wide association study (GWAS) in Taiwan, addressing the limited data on Asian populations compared to Western populations. Using data from the Taiwan Biobank, comprehensive clinical and genetic information from 107,230 Taiwanese individuals was analyzed. Genotyping data from the TWB1.0 and TWB2.0 chips, including over 650,000 single nucleotide polymorphisms (SNPs), were utilized. Genotype imputation using the 1000 Genomes Project was performed, resulting in more than 9 million SNPs. MetS was defined based on a modified version of the Adult Treatment Panel III criteria. Among all participants (mean age: 50 years), 23% met the MetS definition. GWAS analysis identified 549 SNPs significantly associated with MetS, collectively mapping to 10 genomic risk loci. Notable risk loci included rs1004558, rs3812316, rs326, rs4486200, rs2954038, rs10830963, rs662799, rs62033400, rs183130, and rs34342646. Gene-set analysis revealed 22 associated genes: CETP, LPL, APOA5, SIK3, ZPR1, APOC1, BUD13, MLXIPL, TOMM40, GCK, YKT6, RPS6KB1, FTO, VMP1, TUBD1, BCL7B, C19orf80 (ANGPTL8), SIDT2, SENP7, PAFAH1B2, DOCK6, and FOXA2. This study identified genomic risk loci for MetS in a large Taiwanese population through a comprehensive GWAS approach. These associations provide novel insights into the genetic basis of MetS and hold promise for the potential discovery of clinical biomarkers.
Collapse
Affiliation(s)
- Chih-Yi Ho
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jia-In Lee
- Department of Psychiatry, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shu-Pin Huang
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Ph.D. Program in Environmental and Occupational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Institute of Medical Science and Technology, College of Medicine, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Szu-Chia Chen
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jiun-Hung Geng
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Urology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
| |
Collapse
|
46
|
Icick R, Shadrin A, Holen B, Karadag N, Parker N, O'Connell K, Frei O, Bahrami S, Høegh M, Lagerberg T, Cheng W, Seibert T, Djurovic S, Dale A, Zhou H, Edenberg H, Gelernter J, Smeland O, Hindley G, Andreassen O. Identification of Novel Loci and Cross-Disorder Pleiotropy Through Multi-Ancestry Genome-Wide Analysis of Alcohol Use Disorder in Over One Million Individuals. RESEARCH SQUARE 2023:rs.3.rs-3755915. [PMID: 38196616 PMCID: PMC10775504 DOI: 10.21203/rs.3.rs-3755915/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Alcohol use disorder (AUD) is highly heritable and burdensome worldwide. Genome-wide association studies (GWASs) can provide new evidence regarding the aetiology of AUD. We report a multi-ancestry GWASs across diverse ancestries focusing on a narrow AUD phenotype, using novel statistical tools in a total sample of 1,041,450 individuals [102,079 cases; European, 75,583; African, 20,689 (mostly African-American); Hispanic American, 3,449; East Asian, 2,254; South Asian, 104; descent]. Cross-ancestry functional analyses were performed with European and African samples. Thirty-seven genome-wide significant loci were identified, of which seven were novel for AUD and six for other alcohol phenotypes. Loci were mapped to genes enriched for brain regions relevant for AUD (striatum, hypothalamus, and prefrontal cortex) and potential drug targets (GABAergic, dopaminergic and serotonergic neurons). African-specific analysis yielded a unique pattern of immune-related gene sets. Polygenic overlap and positive genetic correlations showed extensive shared genetic architecture between AUD and both mental and general medical phenotypes, suggesting they are not only complications of alcohol use but also share genetic liability with AUD. Leveraging a cross-ancestry approach allowed identification of novel genetic loci for AUD and underscores the value of multi-ancestry genetic studies. These findings advance our understanding of AUD risk and clinically-relevant comorbidities.
Collapse
Affiliation(s)
| | | | - Børge Holen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Naz Karadag
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Nadine Parker
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Kevin O'Connell
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | | | - Shahram Bahrami
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Margrethe Høegh
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Trine Lagerberg
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Weiqiu Cheng
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Tyler Seibert
- Department of Radiation Medicine and Applied Sciences, Department of Radiology, Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo; NORMENT Centre, Department of Clinical Science, University of Bergen, Bergen
| | - Anders Dale
- Department of Neurosciences, University of California San Diego
| | | | | | | | - Olav Smeland
- NORMENT Centre for Mental Disorders Research, University of Oslo and Oslo University Hospital
| | - Guy Hindley
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, 0407 Oslo
| | - Ole Andreassen
- Oslo University Hospital & Institute of Clinical Medicine, University of Oslo
| |
Collapse
|
47
|
Fan H, Liu Z, Wu X, Yu G, Gu X, Kuang N, Zhang K, Liu Y, Jia T, Sahakian BJ, Robbins TW, Schumann G, Cheng W, Feng J, Becker B, Zhang J. Decoding anxiety-impulsivity subtypes in preadolescent internalising disorders: findings from the Adolescent Brain Cognitive Development study. Br J Psychiatry 2023; 223:542-554. [PMID: 37730654 DOI: 10.1192/bjp.2023.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Internalising disorders are highly prevalent emotional dysregulations during preadolescence but clinical decision-making is hampered by high heterogeneity. During this period impulsivity represents a major risk factor for psychopathological trajectories and may act on this heterogeneity given the controversial anxiety-impulsivity relationships. However, how impulsivity contributes to the heterogeneous symptomatology, neurobiology, neurocognition and clinical trajectories in preadolescent internalising disorders remains unclear. AIMS The aim was to determine impulsivity-dependent subtypes in preadolescent internalising disorders that demonstrate distinct anxiety-impulsivity relationships, neurobiological, genetic, cognitive and clinical trajectory signatures. METHOD We applied a data-driven strategy to determine impulsivity-related subtypes in 2430 preadolescents with internalising disorders from the Adolescent Brain Cognitive Development study. Cross-sectional and longitudinal analyses were employed to examine subtype-specific signatures of the anxiety-impulsivity relationship, brain morphology, cognition and clinical trajectory from age 10 to 12 years. RESULTS We identified two distinct subtypes of patients who internalise with comparably high anxiety yet distinguishable levels of impulsivity, i.e. enhanced (subtype 1) or decreased (subtype 2) compared with control participants. The two subtypes exhibited opposing anxiety-impulsivity relationships: higher anxiety at baseline was associated with higher lack of perseverance in subtype 1 but lower sensation seeking in subtype 2 at baseline/follow-up. Subtype 1 demonstrated thicker prefrontal and temporal cortices, and genes enriched in immune-related diseases and glutamatergic and GABAergic neurons. Subtype 1 exhibited cognitive deficits and a detrimental trajectory characterised by increasing emotional/behavioural dysregulations and suicide risks during follow-up. CONCLUSIONS Our results indicate impulsivity-dependent subtypes in preadolescent internalising disorders and unify past controversies about the anxiety-impulsivity interaction. Clinically, individuals with a high-impulsivity subtype exhibit a detrimental trajectory, thus early interventions are warranted.
Collapse
Affiliation(s)
- Huaxin Fan
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Zhaowen Liu
- School of Computer Science, Northwestern Polytechnical University, China
| | - Xinran Wu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Gechang Yu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Xinrui Gu
- Sino-European School of Technology, Shanghai University, China
| | - Nanyu Kuang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Kai Zhang
- School of Computer Science and Technology, East China Normal University, Shanghai, China
| | - Yu Liu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Tianye Jia
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Barbara J Sahakian
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, UK and Department of Psychiatry, University of Cambridge School of Clinical Medicine, UK
| | - Trevor W Robbins
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China; Behavioural and Clinical Neuroscience Institute, University of Cambridge, UK and Department of Psychology, University of Cambridge, UK
| | - Gunter Schumann
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and PONS-Center, Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Germany
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China; Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, China and Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Benjamin Becker
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China; Department of Psychology, The University of Hong Kong, Hong Kong, China
| | - Jie Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, China and Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| |
Collapse
|
48
|
Yang HS, Teng L, Kang D, Menon V, Ge T, Finucane HK, Schultz AP, Properzi M, Klein HU, Chibnik LB, Schneider JA, Bennett DA, Hohman TJ, Mayeux RP, Johnson KA, De Jager PL, Sperling RA. Cell-type-specific Alzheimer's disease polygenic risk scores are associated with distinct disease processes in Alzheimer's disease. Nat Commun 2023; 14:7659. [PMID: 38036535 PMCID: PMC10689816 DOI: 10.1038/s41467-023-43132-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Many of the Alzheimer's disease (AD) risk genes are specifically expressed in microglia and astrocytes, but how and when the genetic risk localizing to these cell types contributes to AD pathophysiology remains unclear. Here, we derive cell-type-specific AD polygenic risk scores (ADPRS) from two extensively characterized datasets and uncover the impact of cell-type-specific genetic risk on AD endophenotypes. In an autopsy dataset spanning all stages of AD (n = 1457), the astrocytic ADPRS affected diffuse and neuritic plaques (amyloid-β), while microglial ADPRS affected neuritic plaques, microglial activation, neurofibrillary tangles (tau), and cognitive decline. In an independent neuroimaging dataset of cognitively unimpaired elderly (n = 2921), astrocytic ADPRS was associated with amyloid-β, and microglial ADPRS was associated with amyloid-β and tau, connecting cell-type-specific genetic risk with AD pathology even before symptom onset. Together, our study provides human genetic evidence implicating multiple glial cell types in AD pathophysiology, starting from the preclinical stage.
Collapse
Affiliation(s)
- Hyun-Sik Yang
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Ling Teng
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel Kang
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Tian Ge
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Hilary K Finucane
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michael Properzi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Lori B Chibnik
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Timothy J Hohman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Richard P Mayeux
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology and the Taub Institute for the Study of Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Reisa A Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Li P, Wei J, Zhu Y. CellGO: a novel deep learning-based framework and webserver for cell-type-specific gene function interpretation. Brief Bioinform 2023; 25:bbad417. [PMID: 37995133 PMCID: PMC10790717 DOI: 10.1093/bib/bbad417] [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: 08/07/2023] [Revised: 10/09/2023] [Accepted: 10/29/2023] [Indexed: 11/25/2023] Open
Abstract
Interpreting the function of genes and gene sets identified from omics experiments remains a challenge, as current pathway analysis tools often fail to consider the critical biological context, such as tissue or cell-type specificity. To address this limitation, we introduced CellGO. CellGO tackles this challenge by leveraging the visible neural network (VNN) and single-cell gene expressions to mimic cell-type-specific signaling propagation along the Gene Ontology tree within a cell. This design enables a novel scoring system to calculate the cell-type-specific gene-pathway paired active scores, based on which, CellGO is able to identify cell-type-specific active pathways associated with single genes. In addition, by aggregating the activities of single genes, CellGO extends its capability to identify cell-type-specific active pathways for a given gene set. To enhance biological interpretation, CellGO offers additional features, including the identification of significantly active cell types and driver genes and community analysis of pathways. To validate its performance, CellGO was assessed using a gene set comprising mixed cell-type markers, confirming its ability to discern active pathways across distinct cell types. Subsequent benchmarking analyses demonstrated CellGO's superiority in effectively identifying cell types and their corresponding cell-type-specific pathways affected by gene knockouts, using either single genes or sets of genes differentially expressed between knockout and control samples. Moreover, CellGO demonstrated its ability to infer cell-type-specific pathogenesis for disease risk genes. Accessible as a Python package, CellGO also provides a user-friendly web interface, making it a versatile and accessible tool for researchers in the field.
Collapse
Affiliation(s)
- Peilong Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Junfeng Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
50
|
Coltell O, Asensio EM, Sorlí JV, Ortega-Azorín C, Fernández-Carrión R, Pascual EC, Barragán R, González JI, Estruch R, Alzate JF, Pérez-Fidalgo A, Portolés O, Ordovas JM, Corella D. Associations between the New DNA-Methylation-Based Telomere Length Estimator, the Mediterranean Diet and Genetics in a Spanish Population at High Cardiovascular Risk. Antioxidants (Basel) 2023; 12:2004. [PMID: 38001857 PMCID: PMC10669035 DOI: 10.3390/antiox12112004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Biological aging is a relevant risk factor for chronic diseases, and several indicators for measuring this factor have been proposed, with telomere length (TL) among the most studied. Oxidative stress may regulate telomere shortening, which is implicated in the increased risk. Using a novel estimator for TL, we examined whether adherence to the Mediterranean diet (MedDiet), a highly antioxidant-rich dietary pattern, is associated with longer TL. We determined TL using DNA methylation algorithms (DNAmTL) in 414 subjects at high cardiovascular risk from Spain. Adherence to the MedDiet was assessed by a validated score, and genetic variants in candidate genes and at the genome-wide level were analyzed. We observed several significant associations (p < 0.05) between DNAmTL and candidate genes (TERT, TERF2, RTEL1, and DCAF4), contributing to the validity of DNAmTL as a biomarker in this population. Higher adherence to the MedDiet was associated with lower odds of having a shorter TL in the whole sample (OR = 0.93; 95% CI: 0.85-0.99; p = 0.049 after fully multivariate adjustment). Nevertheless, this association was stronger in women than in men. Likewise, in women, we observed a direct association between adherence to the MedDiet score and DNAmTL as a continuous variable (beta = 0.015; SE: 0.005; p = 0.003), indicating that a one-point increase in adherence was related to an average increase of 0.015 ± 0.005 kb in TL. Upon examination of specific dietary items within the global score, we found that fruits, fish, "sofrito", and whole grains exhibited the strongest associations in women. The novel score combining these items was significantly associated in the whole population. In the genome-wide association study (GWAS), we identified ten polymorphisms at the suggestive level of significance (p < 1 × 10-5) for DNAmTL (intergenics, in the IQSEC1, NCAPG2, and ABI3BP genes) and detected some gene-MedDiet modulations on DNAmTL. As this is the first study analyzing the DNAmTL estimator, genetics, and modulation by the MedDiet, more studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Oscar Coltell
- Department of Computer Languages and Systems, Universitat Jaume I, 12071 Castellón, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva M Asensio
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - José V Sorlí
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Carolina Ortega-Azorín
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Rebeca Fernández-Carrión
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Eva C Pascual
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Rocío Barragán
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - José I González
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Ramon Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Juan F Alzate
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia, Medellín 050010, Colombia
- Facultad de Medicina, Centro Nacional de Secuenciación Genómica-CNSG, Sede de Investigación Universitaria-SIU, Universidad de Antioquia, Medellín 050010, Colombia
| | - Alejandro Pérez-Fidalgo
- Department of Medical Oncology, University Clinic Hospital of Valencia, 46010 Valencia, Spain
- Biomedical Research Networking Centre on Cancer (CIBERONC), Health Institute Carlos III, 28029 Madrid, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Olga Portolés
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Jose M Ordovas
- Department of Medical Oncology, University Clinic Hospital of Valencia, 46010 Valencia, Spain
- Nutrition and Genomics, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
- Nutritional Control of the Epigenome Group, Precision Nutrition and Obesity Program, IMDEA Food, UAM + CSIC, 28049 Madrid, Spain
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|