1
|
Lu Y, Sun Y, Feng Z, Jia X, Que J, Cui N, Yu L, Zheng YR, Wei YB, Liu JJ. Genetic insights into the role of mitochondria-related genes in mental disorders: An integrative multi-omics analysis. J Affect Disord 2025; 380:685-695. [PMID: 40180044 DOI: 10.1016/j.jad.2025.03.116] [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: 12/04/2024] [Revised: 02/16/2025] [Accepted: 03/19/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Mitochondrial dysfunction has been implicated in the development of mental disorders, yet the underlying mechanisms remain unclear. In this study, we employed summary-data-based Mendelian randomization (SMR) analysis to explore the associations between mitochondrial-related genes and seven common mental disorders across gene expression, DNA methylation, and protein levels. METHOD Summary statistics from genome-wide association studies were used for seven mental disorders, including attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder, anxiety, bipolar disorder, major depressive disorder, post-traumatic stress disorder, and schizophrenia (SCZ). Instrumental variables associated with 1136 mitochondria-related genes were derived from summary statistics for DNA methylation, gene expression, and protein quantitative trait loci. SMR analyses and colocalization analyses were then conducted across these three biological levels to explore the associations with each of the seven mental disorders. RESULTS We identified mitochondria-related genes associated with mental disorders with multi-omics evidence: RMDN1 for ADHD, and ACADVL, ETFA, MMAB, and PPA2 for SCZ. Specifically, an increase of one standard deviation in the level of RMDN1 was linked to a 12 % decrease in the risk of developing ADHD (OR = 0.88, 95 % CI: 0.83-0.94). Increased levels of ETFA (OR = 1.79, 95 % CI: 1.24-2.60) and MMAB (OR = 1.10, 95 % CI: 1.05-1.16) were significantly associated with increased risk of SCZ. Conversely, high levels of ACADVL (OR = 0.50, 95 % CI: 0.33-0.77) and PPA2 (OR = 0.68, 95 % CI: 0.55-0.85) were associated with a reduced risk of SCZ. CONCLUSIONS These findings suggested that dysfunction in mitochondria-related genes may underlie the molecular mechanisms of ADHD and SCZ, providing novel biomarkers for diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Yan'e Lu
- School of Nursing, Peking University, Beijing 100191, China
| | - Yaoyao Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Zhendong Feng
- Beijing Key Laboratory of Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing 100191, China
| | - Xinlei Jia
- School of Nursing, Peking University, Beijing 100191, China
| | - Jianyu Que
- Xiamen Xianyue Hospital, Xianyue Hospital Affiliated with Xiamen Medical College, Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen 361012, Fujian, China
| | - Naixue Cui
- School of Nursing and Rehabilitation, Shandong University, Shandong Province 250012, China
| | - Lulu Yu
- Mental Health Center, the First Hospital of Hebei Medical University, Hebei Technical Innovation Center for Mental Health Assessment and Intervention, Shijiazhuang, Hebei Province 050031, China
| | - Yi-Ran Zheng
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Ya Bin Wei
- Beijing Key Laboratory of Drug Dependence Research, National Institute on Drug Dependence, Peking University, Beijing 100191, China.
| | - Jia Jia Liu
- School of Nursing, Peking University, Beijing 100191, China.
| |
Collapse
|
2
|
Xu Y, Zhang X, Zhang Y, Ma H, Zhou Z, Qin H, Liu H, Han X. Integrated multi-omics insight into the molecular networks of oxidative stress in triggering multiple sclerosis. Neurobiol Dis 2025; 210:106929. [PMID: 40280189 DOI: 10.1016/j.nbd.2025.106929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
Oxidative stress (OS) is a key pathophysiological mechanism in multiple sclerosis (MS). However, the underlying mechanisms by which OS triggered MS remain unknown. To identify potential causal targets of 1216 OS-related genes for MS, a summary-data-based Mendelian randomization (SMR) method was applied. Given that genes can exert their biological functions through different omics levels, the multi-omics SMR integrating expression, methylation, and protein quantitative trait loci (eQTL, mQTL, and pQTL) of OS-related genes from blood and brain tissues was utilized. Bayesian colocalization test was conducted to examine potential regulatory mechanisms of QTL risk variation in MS. To verify the robustness of our results, we validated these findings in FinnGen cohort. Furthermore, the QTL evidence levels, colocalization findings, and replication cohort results were integrated and potential target genes were categorized into three levels. Consequently, three genes (BACH2, TRAF3, and MAPK3) were identified as potential contributors to MS in blood, and four genes (HMGCL, TSFM, TRAF3 and HLA-B) were identified as potential contributors to MS in brain tissue. Additionally, HMGCL and TSFM from brain tissue were supported by first-level evidence related to MS and were validated via in vitro experiments. This research not only contributed to fundamental research of OS in MS but also supported the identification of potential targets for clinical interventions in MS.
Collapse
Affiliation(s)
- Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaowei Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongxuan Ma
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Urology Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongzhuo Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huimin Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
3
|
Kaminska D. Alternative Splicing Regulation in Metabolic Disorders. Obes Rev 2025:e13950. [PMID: 40425033 DOI: 10.1111/obr.13950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/20/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025]
Abstract
Alternative splicing (AS) is a fundamental mechanism for enhancing transcriptome diversity and regulating gene expression, crucial for various cellular processes and the development of complex traits. This review examines the role of AS in metabolic disorders, including obesity, weight loss, dyslipidemias, and metabolic syndrome. We explore the molecular mechanisms underlying AS regulation, focusing on the interplay between cis-acting elements and trans-acting factors, and the influence of RNA-binding proteins (RBPs). Advances in high-throughput sequencing and bioinformatics have unveiled the extensive landscape of AS events across different tissues and conditions, highlighting the importance of tissue-specific splicing in metabolic regulation. We discuss the impact of genetic variants on AS, with a particular emphasis on splicing quantitative trait loci (sQTLs) and their association with cardiometabolic traits. The review also covers the regulation of spliceosome components by phosphorylation, the role of m6A modification in AS, and the interaction between transcription and splicing. Additionally, we address the clinical relevance of AS, illustrating how splicing misregulation contributes to metabolic diseases and the potential for therapeutic interventions targeting splicing mechanisms. This comprehensive overview underscores the significance of AS in metabolic health and disease, advocating for further research to harness its therapeutic potential.
Collapse
Affiliation(s)
- Dorota Kaminska
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
4
|
Ye F, Huang Y, Li N, Hao L, Deng J, Li S, Yue J, Yu F, Hu X. Morphological alterations and gene expression levels in the cerebral cortex causally influence susceptibility to type 2 diabetes: A Mendelian randomization study. Exp Gerontol 2025; 206:112789. [PMID: 40398530 DOI: 10.1016/j.exger.2025.112789] [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: 07/27/2024] [Revised: 04/07/2025] [Accepted: 05/18/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND The associations between type 2 diabetes (T2D) and neurological as well as psychiatric disorders have garnered growing interest. Previous evidence has indicated a correlation between the cerebral cortex and these conditions. However, the causal direction between the cerebral cortex and T2D remains ambiguous. METHODS We conducted a cerebral cortex-focused systematic Mendelian randomization (MR) study based on multiple data sourced from genome-wide association studies and expression quantitative trait locus. RESULTS The surficial area (SA) of Pars Opercularis and the thickness (TH) of the Supramarginal gyrus were found as significant contributors to the risk of T2D. Conversely, thickening in the Precentral area, Caudal Anterior Cingulate cortex, and banks of the Superior Temporal Sulcus, as well as SA amplification of the Precentral area, were associated with a reduced risk of T2D. There was no evidence of reverse causation. These alterations also have an impact on susceptibility to T2D complications. Combining the summary-data-based MR (SMR) analysis and colocalization analysis, we prioritized the expression of three causal genes in the cerebral cortex with genetic evidence for influencing T2D susceptibility. Elevated expression levels of NUDC and PACC1 increased susceptibility to T2D, whereas RAB29 expression exhibits an inverse association with T2D susceptibility. Mediation MR analysis revealed that TH of the Banks of the Superior Temporal Sulcus, SA of Precentral area, SA of Pars Opercularis, and SA of Supramarginal gyrus mediated the effect of RAB29 on T2D. Cross-tissue colocalization analysis demonstrated that the expression pattern of NUDC displayed brain tissue specificity. PACC1 and RAB29 also exhibited colocalization signals in several specific tissues beyond brain tissue. The phenome-wide association study suggested that these genes underscore the shared genetic burden of T2D with a range of disease phenotypes including mental disorders, cardiovascular disease, and malignancies. CONCLUSIONS These findings underscore the novel role of the central nervous system in genetic liability to T2D and provide valuable clues for future mechanism studies.
Collapse
Affiliation(s)
- Fanghang Ye
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yucheng Huang
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Na Li
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Liyuan Hao
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiali Deng
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shenghao Li
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayun Yue
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Yu
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
5
|
Wei C, He J, Li Y, Luo Y, Song L, Han K, Zhang J, Su S, Wang D. Multi-omics identify ribosome related causal genes methylation, splicing, and expression in prostate cancer. Discov Oncol 2025; 16:740. [PMID: 40354008 PMCID: PMC12069195 DOI: 10.1007/s12672-025-02584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Understanding the molecular underpinnings of prostate cancer remains a critical challenge in oncology. Ribosomes, essential cellular organelles responsible for protein synthesis, have emerged as potential regulators in cancer development. Previous studies suggest that dysfunction in ribosomal processes may contribute significantly to prostate cancer progression. We used summary-data-based Mendelian randomization (SMR) and colocalization analysis, as well as single-cell analysis, to investigate the association between ribosome-related genes and prostate cancer by integrating multi-omics. METHOD In this study, we employed a multi-omics approach integrating genomics and transcriptomics data to investigate the role of ribosome-related genes in prostate cancer. Summary-level data for prostate cancer were obtained from The Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome and FinnGen studies. SMR analyses were performed to assess the relevance of ribosomal gene-related molecular signatures to prostate cancer. We further performed colocalization analysis to assess whether the identified signal pairs shared causal genetic variants. Genes were then validated with single-cell sequencing analysis. RESULTS We identified significant causal effects of ribosome gene methylation on prostate cancer. After integrating the multi-omics data of mQTL, sQTL and eQTL, we identified two ribosomal genes, NSUN4 and MPHOSPH6. Methylation and splicing at different sites on the NSUN4 gene showed increased and decreased risks for prostate cancer, indicating complex gene regulation mechanisms. For instance, NSUN4 methylation site of cg10215817 was genetically associated with the increased prostate cancer risk (OR 1.20, 95% CI 1.10,1.30), while NSUN4 methylation site of cg00937489 was genetically associated with the decreased prostate cancer risk (OR 0.84, 95% CI 0.74,0.94); NSUN4 chr1:46341497:46344801 splicing (OR 1.11, 95% CI 1.05-1.17) were positively associated with prostate cancer risk, while NSUN4 chr1:46340919:46344801 splicing (OR 0.95, 95% CI 0.92-0.97) were negatively associated with prostate cancer risk. Expression analysis indicated significant associations between prostate cancer risk and increased expression levels of NSUN4 (OR 1.06, 95% CI 1.03-1.09; PPH4 = 0.79) and MPHOSPH6 (OR 1.07, 95% CI 1.04-1.10; PPH4 = 0.70). In-depth single-cell analysis showed that NSUN4 highly expresses in epithelial cells, while MPHOSPH6 highly expresses in myeloid cells. CONCLUSION The study found that ribosome NSUN4 and MPHOSPH6 genes were associated with prostate cancer risk. This integrative multi-omics study underscores the significance of ribosome-related genes in prostate cancer etiology. By elucidating the molecular mechanisms underlying ribosome dysfunction, our research identifies potential therapeutic targets for mitigating disease progression. These findings not only enhance our understanding of prostate cancer biology but also pave the way for personalized therapeutic strategies targeting ribosomal pathways to improve clinical outcomes.
Collapse
Affiliation(s)
- Chengcheng Wei
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingke He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunfan Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Luo
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangdong Song
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kun Han
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jindong Zhang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Shuai Su
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Delin Wang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Nagura Y, Shimada M, Kuribayashi R, Ikemoto K, Kiyose H, Igarashi A, Kaname T, Unoki M, Fujimoto A. Long-read sequencing reveals novel isoform-specific eQTLs and regulatory mechanisms of isoform expression in human B cells. Genome Biol 2025; 26:110. [PMID: 40336129 PMCID: PMC12060498 DOI: 10.1186/s13059-025-03583-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Genetic variations linked to changes in gene expression are known as expression quantitative loci (eQTLs). The identification of eQTLs helps to understand the mechanisms governing gene expression. However, prior studies have primarily utilized short-read sequencing techniques, and the analysis of eQTLs on isoforms has been relatively limited. RESULTS In this study, we employ long-read sequencing technology (Oxford Nanopore) on B cells from 67 healthy Japanese individuals to explore genetic variations associated with isoform expression levels, referred to as isoform eQTLs (ieQTLs). Our analysis reveals 17,119 ieQTLs, with 70.6% remaining undetected by a gene-level analysis. Additionally, we identify ieQTLs that have significantly different effects on isoform expression levels within a gene. A functional feature analysis demonstrates a significant enrichment of ieQTLs at splice sites and specific histone marks, such as H3K36me3, H3K4me1, H3K4me3, and H3K79me2. Through an experimental validation using genome editing, we observe that a distant genomic region can modulate isoform-specific expression. Moreover, an ieQTL analysis and minigene splicing assays unveils functionally crucial variants in splicing that splicing prediction software did not assign a high prediction score. A comparison with GWAS data reveals a higher number of colocalizations between ieQTLs and GWAS findings compared to gene eQTLs. CONCLUSIONS These findings highlight the substantial contribution of ieQTLs identified through long-read analysis in our understanding of the functional implications of genetic variations and the regulatory mechanisms governing isoforms.
Collapse
Affiliation(s)
- Yuya Nagura
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mihoko Shimada
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryoji Kuribayashi
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ko Ikemoto
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki Kiyose
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Arisa Igarashi
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Genome Medicine, National Centre for Child Health and Development, Tokyo, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Centre for Child Health and Development, Tokyo, Japan
| | - Motoko Unoki
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Fujimoto
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
7
|
Bao W, Bi H, Chao L, Jiang Y, Yu X, Ruan F, Wu D, Chen Z, Le K. Identifying the mediating role of brain atrophy on the relationship between DNA damage repair pathway and Alzheimer's disease: A Mendelian randomization analysis and mediation analysis. J Alzheimers Dis 2025:13872877251333811. [PMID: 40313062 DOI: 10.1177/13872877251333811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
BackgroundDNA damage and repair (DDR) and structural atrophies in different brain regions were recognized as critical factors in the onset of Alzheimer's disease (AD).ObjectiveWe utilized Mendelian randomization (MR) to examine the causal effects of the DDR-related molecular traits on AD and the potential mediating roles of different brain region volumes.MethodsIn primary analysis, we utilized public genome-wide association studies of AD and summary data from existing molecular traits datasets, including gene expression, DNA methylation, and protein levels quantitative trait loci (eQTL, mQTL, and pQTL) in both blood and brain to examine their causal associations by summary-data-based MR analysis and additional five two-sample MR methods. Subsequently, mediation analysis explored the potential mediate roles of 13 imaging-derived brain volume phenotypes in the associations between the DDR pathways and AD through a network MR design.ResultsWe found that the volumes of the right thalamus proper and global cerebral white matter mediated the causal pathways from EGFR to AD and relatively weak mediation effects of the right lateral ventricle volume in the causal pathways involving CHRNE, DNTT, and AD.ConclusionsWe identified causal relationships among DDR pathways, specific brain region volumes, and AD. Monitoring the molecular traits of these DDR-related genes and developing targeted drugs may help detect and interrupt the early progression of AD.
Collapse
Affiliation(s)
- Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang, Jiangxi Province, China
| | - Haidi Bi
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Lishuo Chao
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang, Jiangxi Province, China
| | - Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong S.A.R., China
| |
Collapse
|
8
|
Xu C, Zhu Z, Chen X, Lu M, Wang C, Zhang S, Shi L, Cheng L, Zhang X. Integrating a multi-omics strategy framework to screen potential targets in cognitive impairment-related epilepsy. Methods 2025; 237:34-44. [PMID: 40049431 DOI: 10.1016/j.ymeth.2025.03.003] [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: 01/08/2025] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025] Open
Abstract
Epilepsy is a prevalent neurological disorder that affects over 70 million individuals worldwide and is often associated with cognitive impairments. Despite the widespread impact of epilepsy and cognitive impairments, the genetic basis and causal relationships underlying these conditions remain uncertain, prompting us to conduct a comprehensive investigation into the molecular mechanisms involved. In this study, we utilized statistical data from the third National Health and Nutrition Examination Survey (NHANES III) to evaluate correlation and large-scale pan-phenotype genome-wide association study (GWAS) data to establish genetic correlation and causality. Leveraging multi-omics datasets, we performed a comprehensive post-analysis that included variant prioritization, gene analysis, tissue and cell type enrichment, and pathway annotation. An integrated strategy-multi-trait analysis of GWAS (MTAG), transcriptome-wide association study (TWAS), summary-data-based Mendelian Randomization (SMR), and protein quantitative trait locus (pQTL)-MR-was performed to investigate the shared genetic architecture. Based on multiple orthogonal lines of evidence, we thereby identified 40 single nucleotide polymorphisms (SNPs) and 85 genes common to both conditions. Additionally, we optimized candidate genes such as GNAQ, FADS1, and PTK2 by single-cell expression analysis and molecular pathway mechanisms, thereby highlighting potential shared genetic pathways. These findings elucidate the genetic interplay and co-occurring mechanisms between epilepsy and cognitive impairments, providing crucial insights for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Chao Xu
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150028, China; Department of Pediatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Zijun Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China.
| | - Xinyu Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China
| | - Minke Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China
| | - Chao Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China.
| | - Sainan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China
| | - Lei Shi
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150028, China.
| | - Liang Cheng
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150028, China; College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150001, China.
| | - Xue Zhang
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin 150028, China; Department of Pediatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin 150081, China; McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
9
|
Fan Z, Su H, Qiao T, Shi S, Shi P, Zhang A. TEX10: A Novel Drug Target and Potential Therapeutic Direction for Sleep Apnea Syndrome. Nat Sci Sleep 2025; 17:731-746. [PMID: 40330585 PMCID: PMC12053781 DOI: 10.2147/nss.s499895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Background Sleep apnea syndrome (SAS) is a prevalent sleep disorder strongly associated with obesity, metabolic dysregulation, and cardiovascular diseases. While its underlying pathophysiological mechanisms remain incompletely understood, genetic factors likely play a pivotal role in SAS pathogenesis. This study investigates the causal relationships between potential drug target genes and SAS using multiple statistical approaches, aiming to provide novel insights for targeted therapeutic development. Methods We conducted a comprehensive genetic analysis integrating multiple methodologies to investigate gene-SAS relationships. Using publicly available GWAS and eQTL databases, we performed Mendelian Randomization (MR) analysis with the inverse variance weighted (IVW) method, validated by weighted median and MR-Egger approaches. Summary-data-based MR (SMR) analysis, coupled with HEIDI testing, assessed direct gene expression-SAS associations while controlling for linkage disequilibrium (LD). Colocalization analysis evaluated the probability of shared causal variants between SNPs, gene expression, and SAS. Statistical significance was determined using Benjamini-Hochberg multiple testing correction (FDR < 0.05). Additionally, mediation analysis explored TEX10's influence on SAS through metabolic intermediates including BMI, waist circumference, and HDL cholesterol. Results We identified 18 candidate drug target genes significantly associated with SAS, with MAPKAPK3, TNXB, MPHOSPH8, and TEX10 showing consistent associations across multiple analyses. TEX10, in particular, exhibited significant associations with SAS risk in blood, cerebral cortex, hippocampus, and basal ganglia (PP.H4 > 0.9). Mediation analysis suggested that TEX10 might influence SAS risk indirectly through BMI, waist circumference, and HDL cholesterol levels. Conclusion Our study identified multiple potential therapeutic targets causally linked to SAS, with TEX10 emerging as a key candidate gene. These findings advance our understanding of SAS pathogenesis and offer promising directions for personalized diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Zhitao Fan
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Hui Su
- Department of Neurosurgery, Xingtai People’s Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Tong Qiao
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Sunan Shi
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Pengfei Shi
- Department of Ophthalmology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Anqi Zhang
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| |
Collapse
|
10
|
Li Z, Li Y, Zhao J, Zhang F, Dang W, Jia Y, Guo F, Guo L. Association among blood pressure, antihypertensive drugs, and amyotrophic lateral sclerosis. ARQUIVOS DE NEURO-PSIQUIATRIA 2025; 83:1-8. [PMID: 40360159 DOI: 10.1055/s-0045-1804922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disease. The impacts of antihypertensive drugs and blood pressure (BP) on ALS are currently debatable. OBJECTIVE To evaluate the causal relationship involving antihypertensive drugs, BP, and ALS through a Mendelian randomization (MR) analysis. METHODS The causal relationship between BP and ALS was evaluated by a bidirectional two-sample MR analysis. Then, a sensitivity analysis was performed using a secondary BP genome-wide association study. The drug-target MR was employed to evaluate the impact of antihypertensive drugs on ALS. Furthermore, we used cis-expression quantitative trait loci (cis-eQTLs) data from brain tissue and blood to validate the positive results by a summary-based MR method. RESULTS We found that an increment in systolic BP (SBP) could elevate the risk of ALS (inverse-variance weighted [IVW] odds ratio [OR] = 1.003; 95% confidence interval [95%CI]: 1.001-1.006; per 10-mmHg increment) and ALS might be protected by angiotensin-converting enzyme inhibitors (ACEIs; OR = 0.970; 95%CI: 0.956-0.984; p = 1.96 × 10-5; per 10-mmHg decrement). A causal relationship was not observed between diastolic BP and other antihypertensive drugs in ALS. CONCLUSION In the present study, genetic support for elevated SBP serves as a risk factor for ALS. Besides, ACEIs hold promise as a candidate for ALS.
Collapse
Affiliation(s)
- Zhiguang Li
- Xingtai Central Hospital, Department of Neurology, Xingtai Hebei, People's Republic of China
- Xingtai Medical College, Department of Basic Medicine, Xingtai Hebei, People's Republic of China
| | - Yan Li
- Xingtai Central Hospital, Department of Neurology, Xingtai Hebei, People's Republic of China
| | - Jiankai Zhao
- Xingtai Central Hospital, Department of Neurology, Xingtai Hebei, People's Republic of China
- Xingtai Medical College, Department of Basic Medicine, Xingtai Hebei, People's Republic of China
| | - Feifei Zhang
- Xingtai Medical College, Department of Basic Medicine, Xingtai Hebei, People's Republic of China
| | - Wei Dang
- Xingtai Medical College, Department of Basic Medicine, Xingtai Hebei, People's Republic of China
| | - Yanan Jia
- Xingtai Central Hospital, Department of Science and Education, Xingtai Hebei, People's Republic of China
| | - Fei Guo
- Xingtai Medical College, Department of Basic Medicine, Xingtai Hebei, People's Republic of China
| | - Lixin Guo
- Xingtai Medical College, Department of Basic Medicine, Xingtai Hebei, People's Republic of China
- Xingtai Central Hospital, Department of Cardiac Surgery, Xingtai Hebei, People's Republic of China
| |
Collapse
|
11
|
Guan D, Bai Z, Zhu X, Zhong C, Hou Y, Zhu D, Li H, Lan F, Diao S, Yao Y, Zhao B, Li X, Pan Z, Gao Y, Wang Y, Zou D, Wang R, Xu T, Sun C, Yin H, Teng J, Xu Z, Lin Q, Shi S, Shao D, Degalez F, Lagarrigue S, Wang Y, Wang M, Peng M, Rocha D, Charles M, Smith J, Watson K, Buitenhuis AJ, Sahana G, Lund MS, Warren W, Frantz L, Larson G, Lamont SJ, Si W, Zhao X, Li B, Zhang H, Luo C, Shu D, Qu H, Luo W, Li Z, Nie Q, Zhang X, Xiang R, Liu S, Zhang Z, Zhang Z, Liu GE, Cheng H, Yang N, Hu X, Zhou H, Fang L. Genetic regulation of gene expression across multiple tissues in chickens. Nat Genet 2025; 57:1298-1308. [PMID: 40200121 DOI: 10.1038/s41588-025-02155-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/05/2025] [Indexed: 04/10/2025]
Abstract
The chicken is a valuable model for understanding fundamental biology and vertebrate evolution and is a major global source of nutrient-dense and lean protein. Despite being the first non-mammalian amniote to have its genome sequenced, a systematic characterization of functional variation on the chicken genome remains lacking. Here, we integrated bulk RNA sequencing (RNA-seq) data from 7,015 samples, single-cell RNA-seq data from 127,598 cells and 2,869 whole-genome sequences to present a pilot atlas of regulatory variants across 28 chicken tissues. This atlas reveals millions of regulatory effects on primary expression (protein-coding genes, long non-coding RNA and exons) and post-transcriptional modifications (alternative splicing and 3'-untranslated region alternative polyadenylation). We highlighted distinct molecular mechanisms underlying these regulatory variants, their context-dependent behavior and their utility in interpreting genome-wide associations for 39 chicken complex traits. Finally, our comparative analyses of gene regulation between chickens and mammals demonstrate how this resource can facilitate cross-species gene mapping of complex traits.
Collapse
Affiliation(s)
- Dailu Guan
- Department of Animal Science, University of California-Davis, Davis, CA, USA
| | - Zhonghao Bai
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Xiaoning Zhu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Conghao Zhong
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yali Hou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Di Zhu
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Houcheng Li
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Fangren Lan
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuqi Diao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yuelin Yao
- MRC Human Genetics Unit at the Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
- School of Informatics, The University of Edinburgh, Edinburgh, UK
| | - Bingru Zhao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhangyuan Pan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yahui Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Yuzhe Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Dong Zou
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Ruizhen Wang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianyi Xu
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hongwei Yin
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Jinyan Teng
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhiting Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qing Lin
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shourong Shi
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Dan Shao
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | | | | | - Ying Wang
- Department of Animal Science, University of California-Davis, Davis, CA, USA
| | - Mingshan Wang
- State Key Laboratory of Genetic Evolution & Animal Models and Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Minsheng Peng
- State Key Laboratory of Genetic Evolution & Animal Models and Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Dominique Rocha
- INRAE, GABI, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Mathieu Charles
- INRAE, GABI, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jacqueline Smith
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Kellie Watson
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | | | - Goutam Sahana
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Mogens Sandø Lund
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark
| | - Wesley Warren
- Department of Animal Sciences, Data Science and Informatics Institute, University of Missouri, Columbia, MO, USA
| | - Laurent Frantz
- Palaeogenomics Group, Department of Veterinary Sciences, Ludwig Maximilian University, Munich, Germany
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Greger Larson
- The Palaeogenomics & Bio-Archaeology Research Network, School of Archaeology, University of Oxford, Oxford, UK
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Wei Si
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of Animal Science, McGill University, Montreal, Quebec, Canada
| | - Xin Zhao
- Department of Animal Science, McGill University, Montreal, Quebec, Canada
| | - Bingjie Li
- Scotland's Rural College (SRUC), Roslin Institute Building, Midlothian, UK
| | - Haihan Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Chenglong Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Dingming Shu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hao Qu
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Wei Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zhenhui Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qinghua Nie
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiquan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ruidong Xiang
- Agriculture Victoria, Agribio, Centre for AgriBiosciences, Bundoora, Victoria, Australia
- Cambridge-Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- School of Agriculture, Food and Ecosystem Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Shuli Liu
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Zhe Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhang Zhang
- Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD, USA
| | - Hans Cheng
- Avian Disease and Oncology Laboratory, USDA, ARS, USNPRC, East Lansing, MI, USA
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| | - Xiaoxiang Hu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| | - Huaijun Zhou
- Department of Animal Science, University of California-Davis, Davis, CA, USA.
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark.
| |
Collapse
|
12
|
Zhou M, Ling C, Xiao H, Zhang Z. Identification of Gene Expression and Splicing QTLs in Porcine Muscle Associated with Meat Quality Traits. Animals (Basel) 2025; 15:1209. [PMID: 40362025 PMCID: PMC12071002 DOI: 10.3390/ani15091209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/10/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Understanding the genetic regulation of gene expression and splicing in muscle tissues is critical for elucidating the molecular mechanisms of meat quality traits. In this study, we integrated large-scale whole-genome sequencing and strand-specific RNA-seq data from 582 F2 hybrid pigs (White Duroc × Erhualian) to characterize the expression and splicing quantitative trait loci (eQTLs/sQTL) in longissimus dorsi muscle. We identified 11,058 cis-eQTL-associated genes (eGenes) and 5139 cis-sQTL-associated genes (sGenes), of which 29% of eGenes and 80% of sGenes were previously unreported in the PigGTEx database. Functional analyses revealed distinct genomic features: eQTLs were enriched near transcription start sites (TSSs) and associated with active TSS-proximal transcribed regions and enhancers, whereas sQTLs clustered at splice junctions, underscoring their distinct roles in gene expression and splicing regulation. Colocalization analysis of e/sQTLs with GWAS signals prioritized PHKG1 as a key candidate gene (PPH4 > 0.9) for glycogen metabolism. Notably, we confirmed that an sQTL-driven alternative splicing event in exon 10 of PHKG1 was significantly correlated with phenotypic variation (R = -0.39, p = 9.5 × 10-21). Collectively, this study provides novel insights into the genetic regulation of gene expression and alternative splicing in porcine muscle tissue, advancing our understanding of the molecular mechanisms underlying economically important meat quality traits.
Collapse
Affiliation(s)
- Meng Zhou
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation Technology, Jiangxi Agricultural University, Nanchang 330045, China; (C.L.); (H.X.); (Z.Z.)
| | | | | | | |
Collapse
|
13
|
Charles M, Gaiani N, Sanchez MP, Boussaha M, Hozé C, Boichard D, Rocha D, Boulling A. Functional impact of splicing variants in the elaboration of complex traits in cattle. Nat Commun 2025; 16:3893. [PMID: 40274775 PMCID: PMC12022281 DOI: 10.1038/s41467-025-58970-5] [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: 04/16/2024] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
GWAS conducted directly on imputed whole genome sequence have led to the identification of numerous genetic variants associated with agronomic traits in cattle. However, such variants are often simply markers in linkage disequilibrium with the actual causal variants, which is a limiting factor for the development of accurate genomic predictions. It is possible to identify causal variants by integrating information on how variants impact gene expression into GWAS output. RNA splicing plays a major role in regulating gene expression. Thus, assessing the effect of variants on RNA splicing may explain their function. Here, we use a high-throughput strategy to functionally analyse putative splice-disrupting variants in the bovine genome. Using GWAS, massively parallel reporter assay and deep learning algorithms designed to predict splice-disrupting variants, we identify 38 splice-disrupting variants associated with complex traits in cattle, three of which could be classified as causal. Our results indicate that splice-disrupting variants are widely found in the quantitative trait loci related to these phenotypes. Using our combined approach, we also assess the validity of splicing predictors originally developed to analyse human variants in the context of the bovine genome.
Collapse
Affiliation(s)
- Mathieu Charles
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- INRAE, SIGENAE, 78350, Jouy-en-Josas, France
| | - Nicolas Gaiani
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Marie-Pierre Sanchez
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Mekki Boussaha
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Chris Hozé
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- ELIANCE, 75012, Paris, France
| | - Didier Boichard
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Dominique Rocha
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Arnaud Boulling
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
| |
Collapse
|
14
|
Wang C, Pu Q, Mo X, Han X, Wang F, Li W, Chen C, Xue Y, Xin J, Shen C, Du M, Wu D. A global overview of shared genetic architecture between smoking behaviors and major depressive disorder in European and East Asian ancestry. J Affect Disord 2025; 375:10-21. [PMID: 39842668 DOI: 10.1016/j.jad.2025.01.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/01/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND The co-occurrence of smoking behaviors and major depressive disorder (MDD) has been widely documented in populations. However, the underlying mechanism of this association remains unclear. METHODS Genome-wide association studies of smoking behaviors and MDD, combined with multi-omics datasets, were used to characterise genetic correlations, identify shared loci and genes, and explore underlying biological mechanisms. Mendelian randomization (MR) analyses were conducted to infer causal relationships between smoking behaviors and MDD. Druggability analyses were performed to identify potential drugs with both antidepressant and smoking cessation effects. RESULTS Extensive overall genetic correlations were found between smoking behaviors and MDD. Furthermore, eighteen local regions showed significant genetic correlations, which could be partly explained by gene co-expression patterns. We identified 24 shared loci and 120 genes, which were enriched in limbic system, GABAergic and dopaminergic neurons, as well as in synaptic pathways. Through integrating with tissue specific information, seven key genes (ANKK1, NEGR1, USP4, TCTA, SORCS5, SPPL3, and USP28) were pinpointed. Notably, druggability analyses supported ANKK1 as a potential drug target for the treatment of MDD and tobacco dependence. MR analyses suggested a bidirectional causal relationship between smoking initiation and MDD. Although findings in East Asian ancestry were limited, the shared locus (chr15:47613403-47,685,504) identified in European ancestry remained significant in East Asian ancestry. CONCLUSIONS Our findings suggest the extensive genetic overlap between smoking behaviors and MDD, support the role of limbic system and synapse involved in shared mechanisms, and implicate for prevention, intervention and treatment.
Collapse
Affiliation(s)
- Chao Wang
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiuyi Pu
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoxiao Mo
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xu Han
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Feifan Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wen Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Changying Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yong Xue
- Department of Medical Laboratory, Huai'an No 3 People's Hospital, Huai'an, China
| | - Junyi Xin
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Mulong Du
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Dongmei Wu
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Toikumo S, Davis C, Jinwala Z, Khan Y, Jennings M, Davis L, Sanchez-Roige S, Kember RL, Kranzler HR. Gene discovery and pleiotropic architecture of chronic pain in a genome-wide association study of >1.2 million individuals. RESEARCH SQUARE 2025:rs.3.rs-6173614. [PMID: 40297705 PMCID: PMC12036444 DOI: 10.21203/rs.3.rs-6173614/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Chronic pain is highly prevalent worldwide, and genome-wide association studies (GWAS) have identified a growing number of chronic pain loci. To further elucidate its genetic architecture, we leveraged data from 1,235,695 European ancestry individuals across three biobanks. In a meta-analytic GWAS, we identified 343 independent loci for chronic pain, 92 of which were new. Sex-specific meta-analyses revealed 115 independent loci (12 of which were new) for males (N = 583,066) and 12 loci (two of which were new) for females (N = 241,266). Multi-omics gene prioritization analyses highlighted 490 genes associated with chronic pain through their effects on brain- and blood-specific regulation. Loci associated with increased risk for chronic pain were also associated with increased risk for multiple other traits, with Mendelian randomization analyses showing that chronic pain was causally associated with psychiatric disorders, substance use disorders, and C-reactive protein levels. Chronic pain variants also exhibited pleiotropic associations with cortical area brain structures. This study expands our knowledge of the genetics of chronic pain and its pathogenesis, highlighting the importance of its pleiotropy with multiple disorders and elucidating its multi-omic pathophysiology.
Collapse
Affiliation(s)
- Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Christal Davis
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Zeal Jinwala
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Yousef Khan
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Mariela Jennings
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Lea Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, 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
| | - Rachel L. Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Henry R. Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| |
Collapse
|
16
|
Li Y, Dang X, Chen R, Teng Z, Wang J, Li S, Yue Y, Mitchell BL, Zeng Y, Yao YG, Li M, Liu Z, Yuan Y, Li T, Zhang Z, Luo XJ. Cross-ancestry genome-wide association study and systems-level integrative analyses implicate new risk genes and therapeutic targets for depression. Nat Hum Behav 2025; 9:806-823. [PMID: 39994458 DOI: 10.1038/s41562-024-02073-6] [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: 01/23/2024] [Accepted: 10/23/2024] [Indexed: 02/26/2025]
Abstract
Deciphering the genetic architecture of depression is pivotal for characterizing the associated pathophysiological processes and development of new therapeutics. Here we conducted a cross-ancestry genome-wide meta-analysis on depression (416,437 cases and 1,308,758 controls) and identified 287 risk loci, of which 49 are new. Variant-level fine mapping prioritized potential causal variants and functional genomic analysis identified variants that regulate the binding of transcription factors. We validated that 80% of the identified functional variants are regulatory variants, and expression quantitative trait loci analysis uncovered the potential target genes regulated by the prioritized risk variants. Gene-level analysis, including transcriptome and proteome-wide association studies, colocalization and Mendelian randomization-based analyses, prioritized potential causal genes and drug targets. Gene prioritization analyses highlighted likely causal genes, including TMEM106B, CTNND1, AREL1 and so on. Pathway analysis indicated significant enrichment of depression risk genes in synapse-related pathways. Finally, knockdown of Tmem106b in mice resulted in depression-like behaviours, supporting the involvement of Tmem106b in depression. Our study identified new risk loci, likely causal variants and genes for depression, providing important insights into the genetic architecture of depression and potential therapeutic targets.
Collapse
Affiliation(s)
- Yifan Li
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Xinglun Dang
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yingying Yue
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China
| | - Brittany L Mitchell
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Yong Zeng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yonggui Yuan
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
| | - Tao Li
- Affiliated Mental Health Center, Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhijun Zhang
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Xiong-Jian Luo
- Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
17
|
Strom NI, Halvorsen MW, Grove J, Ásbjörnsdóttir B, Luðvígsson P, Thorarensen Ó, de Schipper E, Bäckmann J, Andrén P, Tian C, Als TD, Nissen JB, Meier SM, Bybjerg-Grauholm J, Hougaard DM, Werge T, Børglum AD, Hinds DA, Rück C, Mataix-Cols D, Stefánsson H, Stefansson K, Crowley JJ, Mattheisen M. Genome-Wide Association Study Meta-Analysis of 9619 Cases With Tic Disorders. Biol Psychiatry 2025; 97:743-752. [PMID: 39389409 PMCID: PMC12036792 DOI: 10.1016/j.biopsych.2024.07.025] [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: 12/05/2023] [Revised: 06/11/2024] [Accepted: 07/05/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Despite the significant personal and societal burden of tic disorders (TDs), treatment outcomes remain modest, necessitating a deeper understanding of their etiology. Family history is the biggest known risk factor, and identifying risk genes could accelerate progress in the field. METHODS Expanding upon previous sample size limitations, we added 4800 new TD cases and 971,560 controls and conducted a genome-wide association study (GWAS) meta-analysis with 9619 cases and 981,048 controls of European ancestry. We attempted to replicate the results in an independent deCODE genetics GWAS (885 TD cases and 310,367 controls). To characterize GWAS findings, we conducted several post-GWAS gene-based and enrichment analyses. RESULTS A genome-wide significant hit (rs79244681, p = 2.27 × 10-8) within MCHR2-AS1 was identified, although it was not replicated. Post-GWAS analyses revealed a 13.8% single nucleotide polymorphism heritability and 3 significant genes: BCL11B, NDFIP2, and RBM26. Common variant risk for TD was enriched within genes preferentially expressed in the cortico-striato-thalamo-cortical circuit (including the putamen, caudate, nucleus accumbens, and Brodmann area 9) and 5 brain cell types (excitatory and inhibitory telencephalon neurons, inhibitory diencephalon and mesencephalon neurons, and hindbrain and medium spiny neurons). TD polygenic risk was enriched within loss-of-function intolerant genes (p = .0017) and high-confidence neurodevelopmental disorder genes (p = .0108). Of 112 genetic correlations, 43 were statistically significant, showing high positive correlations with most psychiatric disorders. Of the 2 single nucleotide polymorphisms previously associated with TDs, one (rs2453763) replicated in an independent subsample of our GWAS (p = .00018). CONCLUSIONS This GWAS was still underpowered to identify high-confidence, replicable loci, but the results suggest imminent discovery of common genetic variants for TDs.
Collapse
Affiliation(s)
- Nora I Strom
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany; Institute of Psychiatric Phenomics and Genomics, University Hospital, LMU Munich, Munich, Germany; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden; Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Matthew W Halvorsen
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jakob Grove
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark; Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | | | - Pétur Luðvígsson
- Department of Pediatrics, Landspitali University Hospital, Reykjavik, Iceland
| | - Ólafur Thorarensen
- Department of Pediatrics, Landspitali University Hospital, Reykjavik, Iceland
| | - Elles de Schipper
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden
| | - Julia Bäckmann
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden
| | - Per Andrén
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden
| | - Chao Tian
- 23andMe, Inc., Sunnyvale, California
| | - Thomas Damm Als
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Judith Becker Nissen
- Department of Child and Adolescent Psychiatry, Aarhus University Hospital, Aarhus, Denmark; Institute of Clinical Medicine, Institute of Health, Aarhus University, Aarhus, Denmark
| | - Sandra M Meier
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - David M Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Institute of Biological Psychiatry, Mental Health Services Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark; GLOBE Institute, Center for GeoGenetics, University of Copenhagen, Copenhagen, Denmark
| | - Anders D Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | | | - Christian Rück
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden
| | - David Mataix-Cols
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden
| | | | | | - James J Crowley
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Sweden; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Manuel Mattheisen
- Institute of Psychiatric Phenomics and Genomics, University Hospital, LMU Munich, Munich, Germany; Department of Community Health and Epidemiology & Faculty of Computer Science, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
18
|
Hu B, Yin MY, Zhang CY, Shi Z, Wang L, Lei X, Li M, Li SW, Tuo QH. The INO80E at 16p11.2 locus increases risk of schizophrenia in humans and induces schizophrenia-like phenotypes in mice. EBioMedicine 2025; 114:105645. [PMID: 40088626 PMCID: PMC11957503 DOI: 10.1016/j.ebiom.2025.105645] [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: 05/21/2024] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Chromosome 16p11.2 is one of the most significant loci in the genome-wide association studies (GWAS) of schizophrenia. Despite several integrative analyses and functional genomics studies having been carried out to identify possible risk genes, their impacts in the pathogenesis of schizophrenia remain to be fully characterized. METHODS We performed expression quantitative trait loci (eQTL) and summary-data-based Mendelian randomization (SMR) analyses to identify schizophrenia risk genes in the 16p11.2 GWAS locus. We constructed a murine model with dysregulated expression of risk gene in the medial prefrontal cortex (mPFC) using stereotaxic injection of adeno-associated virus (AAV), followed by behavioural assessments, dendritic spine analyses and RNA sequencing. FINDINGS We identified significant associations between elevated INO80E mRNA expression in the frontal cortex and risk of schizophrenia. The mice overexpressing Ino80e in mPFC (Ino80e-OE) exhibited schizophrenia-like behaviours, including increased anxiety behaviour, anhedonia, and impaired prepulse inhibition (PPI) when compared with control group. The neuronal sparse labelling assay showed that the density of stubby spines in the pyramidal neurons of mPFC was significantly increased in Ino80e-OE mice compared with control mice. Transcriptomic analysis in the mPFC revealed significant alterations in the mRNA levels of schizophrenia-related genes and processes related to synapses upon overexpressing Ino80e. INTERPRETATION Our results suggest that upregulation of the Ino80e gene in mPFC may induce schizophrenia-like behaviours in mice, further supporting the hypothesis that INO80E is an authentic risk gene. FUNDING This project received support from the National Key Research and Development Program of China, National Natural Science Foundation of China, Key Research and Development Projects of Hunan Provincial Science and Technology Department, Science and Technology Innovation team of Hunan Province, etc.
Collapse
Affiliation(s)
- Bo Hu
- Hunan Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Mei-Yu Yin
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhe Shi
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Pharmacy of School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lu Wang
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaoming Lei
- Hunan Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ming Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shi-Wu Li
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China; Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Qin-Hui Tuo
- Hunan Key Laboratory of Vascular Biology and Translational Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
19
|
Wang D, Li D, Dang X, Mu C, Liu C, Zeng Y, Yuan Y, Teng Z, Li Y, Luo XJ. Mendelian Randomization Reveals Causalities Between DNA Methylation and Schizophrenia. Biol Psychiatry 2025:S0006-3223(25)01100-X. [PMID: 40157589 DOI: 10.1016/j.biopsych.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/06/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Epigenetic factors (such as DNA methylation) have been widely reported to be associated with schizophrenia (SCZ). However, the causal relationships between epigenetic factors and SCZ remain largely unknown. METHODS Here, we conducted a Mendelian randomization (MR) study to investigate the causal relationships between DNA methylation and SCZ. Brain methylation quantitative trait loci (mQTL) (N = 1160) and blood mQTL (N = 27,750) data were used as exposures, and genome-wide association data of SCZ (53,386 cases and 77,258 controls) were used as the outcome. RESULTS We identified 172 (mapped to 160 genes) and 157 (mapped to 155 genes) methylation sites whose methylation levels in brain and blood are causally associated with SCZ, respectively. Among the mapped genes, 36 overlapping genes were identified. Interestingly, 3 methylation sites (near BRD2, CNNM2, and RERE) showed significant associations in both brain and blood, with the same direction of effect. We also performed MR analysis using brain expression quantitative trait loci (eQTLs) as exposures and identified 123 genes whose expression levels were causally associated with SCZ. Comparing the significant genes from eQTLs and brain mQTLs prioritized 15 overlapping genes, suggesting that both epigenetic modification and expression of these genes confer risk of SCZ. Finally, we validated our findings with genome editing and animal model experiments. CONCLUSIONS Our study identified methylation sites whose methylation levels are causally associated with SCZ and demonstrated the important roles of epigenetic factors in SCZ. Our findings also reveal pivotal risk genes whose expression and epigenetic regulation are causally associated with SCZ.
Collapse
Affiliation(s)
- Danni Wang
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Danyang Li
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Xinglun Dang
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Changgai Mu
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Chang Liu
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Yong Zeng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Yonggui Yuan
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Zhaowei Teng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China.
| | - Yifan Li
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China.
| | - Xiong-Jian Luo
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
20
|
Baranova A, Liu D, Chandhoke V, Cao H, Zhang F. Unraveling the genetic links between depression and type 2 diabetes. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111258. [PMID: 39837361 DOI: 10.1016/j.pnpbp.2025.111258] [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: 11/18/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Type 2 diabetes (T2D) is a chronic metabolic disorder that has high comorbidity with mental disorders. The genetic relationships between T2D and depression are far from being well understood. METHODS We performed genetic correlation, polygenic overlap, Mendelian randomization (MR) analyses, cross-trait meta-analysis, and Bayesian colocalization analysis to assess genetic relationships between T2D and depression, in the forms of major depressive disorder (MDD) and depressed affect (DAF). Then, the summary data-based MR (SMR) analysis was performed to prioritize genes contributing to MDD and to T2D from functional perspective. MDD-driven signaling pathways were constructed to understand the influence of MDD on T2D at the molecular level. RESULTS T2D has positive genetic correlations both with MDD (rg = 0.14) and with DAF (rg = 0.19). The polygenic overlap analysis showed that about 60 % of causal variants for T2D are shared with MDD and DAF. The MR analysis indicated that genetic liabilities to both MDD (OR: 1.24, 95 % CI: 1.11-1.38) and DAF (OR: 1.48, 95 % CI: 1.23-1.78) are associated with an increased risk for T2D, while genetic liability to T2D is not associated with the risk for MDD (OR: 1.00, 95 % CI: 0.99-1.01) or DAF (OR: 1.01, 95 % CI: 1.00-1.02). The cross-trait meta-analysis identified 271 genomic loci, of which 29 were novel. Genetic predisposition to MDD and T2D shares six overlapping loci, involving some well-characterized genes, such as TCF4 and NEGR1. Colocalization analysis revealed three shared chromosome regions between MDD and T2D, which covers mediator genes including SCYL1, DENND1A, and MAD1L1. Molecular pathway analysis suggests mechanisms that promote the development of T2D through inflammatory pathways overactive in patients with MDD. The SMR analysis and the meta-analysis highlighted seven genes with functional implications for both MDD and T2D, including TNKS2, CCDC92, FADS1, ERI1, THUMPD3, NUCKS1, and PM20D1. CONCLUSIONS Our study points out that depression, in the forms of MDD and DAF, may increase the risk of T2D. Analysis of underlying genetic variation and the molecular pathways, connecting depression and T2D, indicate that the pathophysiological foundations of these two conditions have a notable overlap.
Collapse
Affiliation(s)
- Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax 22030, USA; Research Centre for Medical Genetics, Moscow 115478, Russia
| | - Dongming Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Vikas Chandhoke
- School of Systems Biology, George Mason University, Fairfax 22030, USA
| | - Hongbao Cao
- School of Systems Biology, George Mason University, Fairfax 22030, USA
| | - Fuquan Zhang
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China; Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
21
|
Zhang Y, Zhang Z, Yu Q, Jiang Y, Fei C, Wu F, Li F. Mapping fatigue: discovering brain regions and genes linked to fatigue susceptibility. J Transl Med 2025; 23:293. [PMID: 40055680 PMCID: PMC11887381 DOI: 10.1186/s12967-025-06284-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/20/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND The relationship between the brain and fatigue is gaining increasing attention, with numerous studies indicating that certain specific brain regions may be closely linked to fatigue. Our study aimed to identify brain regions exhibiting significant causal relationships to fatigue and discover potential neurotherapeutic targets associated with fatigue, in the pursuit of seeking new approaches for fatigue treatment. METHODS A bidirectional two-sample Mendelian randomization (TSMR) method was employed to investigate causal relationships between cortical and subcortical gray matter volumes in 83 regions and fatigue. Then, we utilized frontal cortex expression Quantitative Trait Loci data, employing the methods of Summary-data-based Mendelian Randomization (SMR) and Bayesian colocalization to identify genes that exhibit significant association with fatigue. Finally, the transcription levels of candidate genes were assessed in a central fatigue rat model using RT-qPCR. RESULTS The results of the TSMR analysis revealed that an increased in the volume of the right lateral orbitofrontal, left caudal middle frontal, right caudal middle frontal, and right rostral middle frontal cortices may be correlated with a diminished susceptibility to fatigue. The SMR and Bayesian colocalization analysis identified ECE2, GPX1, METTL21EP, RP11-665J16.1, and SNF8 as candidate genes associated with fatigue. RT-qPCR results confirmed significantly elevated transcription levels of Ece2, Gpx1, and Snf8 in the frontal cortex of central fatigue model rats compared to controls. CONCLUSIONS Our findings afford substantial theoretical support for the connection between the brain and fatigue, while also providing novel insights into the genetic mechanisms and therapeutic targets for fatigue, particularly central fatigue.
Collapse
Affiliation(s)
- Yifei Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Fangshan District, Beijing, China
| | - Zehan Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Fangshan District, Beijing, China
| | - Qingqian Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Fangshan District, Beijing, China
| | - Yutong Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Fangshan District, Beijing, China
| | - Chenyu Fei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Fangshan District, Beijing, China
| | - Fengzhi Wu
- Periodical Center, Beijing University of Chinese Medicine, Fangshan District, Beijing, China.
| | - Feng Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Fangshan District, Beijing, China.
| |
Collapse
|
22
|
Zhang L, Ge Q, Sun Z, Zhang R, Li X, Luo X, Tian R, Cao Y, Pu C, Li L, Wu D, Jiang P, Yu C, Nosarti C, Xiao C, Liu Z. Association and shared biological bases between birth weight and cortical structure. Transl Psychiatry 2025; 15:74. [PMID: 40044659 PMCID: PMC11882966 DOI: 10.1038/s41398-025-03294-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/14/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Associations between birth weight and cortical structural phenotypes have been detected; however, the understanding is incomprehensive, and the potential biological bases are not well defined. Leveraging data from genome-wide association studies, we investigated the associations and the shared transcriptomic, proteomic and cellular bases of birth weight and 13 cortical structural phenotypes. Mendelian randomization analyses were performed to examine associations between birth weight and cortical structure. Downstream transcriptome-wide association study (TWAS), proteome-wide association study (PWAS) and summary-based Mendelian randomization (SMR) analyses were utilized to identify the shared cis-regulated gene expressions and proteins. Finally, cell-type expression-specific integration for complex traits (CELLECT) analyses were conducted to explore the enriched cell types. The Mendelian randomization analyses found positive associations between birth weight and global cortical folding index, intrinsic curvature index, local gyrification index, surface area and volume. Downstream transcriptomic-level TWAS and SMR identified three gene expressions both linked to birth weight and at least one cortical structural phenotype (CNNM2, RABGAP1 and CENPW). Parallel PWAS and SMR analyses at the proteomic level identified four proteins linked to both phenotypes (CNNM2, RAB7L1, RAB5B and PPA2), of which CNNM2 was replicated. CELLECT analyses revealed brain cell types enriched in birth weight, including pericytes, inhibitory GABAergic neurons and cerebrovascular cells. These findings support the importance of early life growth to cortical structure, and suggest underlying transcriptomic, proteomic and cellular bases. These results provide intriguing targets for further research into the mechanisms of cortical development.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Qiaoyue Ge
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zeyuan Sun
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Rui Zhang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xinxi Li
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xiaoli Luo
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Run Tian
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuheng Cao
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunyan Pu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lin Li
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Dongsheng Wu
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ping Jiang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- West China Medical Publishers, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Chuan Yu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chiara Nosarti
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Chenghan Xiao
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Zhenmi Liu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Toikumo S, Davis C, Jinwala Z, Khan Y, Jennings M, Davis L, Sanchez-Roige S, Kember RL, Kranzler HR. Gene discovery and pleiotropic architecture of Chronic Pain in a Genome-wide Association Study of >1.2 million Individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.28.25323112. [PMID: 40093235 PMCID: PMC11908286 DOI: 10.1101/2025.02.28.25323112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Chronic pain is highly prevalent worldwide, and genome-wide association studies (GWAS) have identified a growing number of chronic pain loci. To further elucidate its genetic architecture, we leveraged data from 1,235,695 European ancestry individuals across three biobanks. In a meta-analytic GWAS, we identified 343 independent loci for chronic pain, 92 of which were new. Sex-specific meta-analyses revealed 115 independent loci (12 of which were new) for males (N = 583,066) and 12 loci (two of which were new) for females (N = 241,266). Multi-omics gene prioritization analyses highlighted 490 genes associated with chronic pain through their effects on brain- and blood-specific regulation. Loci associated with increased risk for chronic pain were also associated with increased risk for multiple other traits, with Mendelian randomization analyses showing that chronic pain was causally associated with psychiatric disorders, substance use disorders, and C-reactive protein levels. Chronic pain variants also exhibited pleiotropic associations with cortical area brain structures. This study expands our knowledge of the genetics of chronic pain and its pathogenesis, highlighting the importance of its pleiotropy with multiple disorders and elucidating its multi-omic pathophysiology.
Collapse
Affiliation(s)
- Sylvanus Toikumo
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Christal Davis
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Zeal Jinwala
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Yousef Khan
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Mariela Jennings
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Lea Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, 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
| | - Rachel L. Kember
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| | - Henry R. Kranzler
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104, USA
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, 3535 Market Street, Philadelphia, PA 19104
| |
Collapse
|
24
|
Dos Santos TCF, Silva EN, Frezarim GB, Salatta BM, Baldi F, Fonseca LFS, Albuquerque LGD, Muniz MMM, Silva DBDS. Identification of cis-sQTL demonstrates genetic associations and functional implications of inflammatory processes in Nelore cattle muscle tissue. Mamm Genome 2025; 36:106-117. [PMID: 39825903 DOI: 10.1007/s00335-024-10100-0] [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: 09/24/2024] [Accepted: 12/22/2024] [Indexed: 01/20/2025]
Abstract
This study aimed to identify splicing quantitative trait loci (cis-sQTL) in Nelore cattle muscle tissue and explore the involvement of spliced genes (sGenes) in immune system-related biological processes. Genotypic data from 80 intact male Nelore cattle were obtained using SNP-Chip technology, while RNA-Seq analysis was performed to measure gene expression levels, enabling the integration of genomic and transcriptomic datasets. The normalized expression levels of spliced transcripts were associated with single nucleotide polymorphisms (SNPs) through an analysis of variance using an additive linear model with the MatrixEQTL package. A permutation analysis then assessed the significance of the best SNPs for each spliced transcript. Functional enrichment analysis was performed on the sGenes to investigate their roles in the immune system. In total, 3,187 variants were linked to 3,202 spliced transcripts, with 83 sGenes involved in immune system processes. Of these, 31 sGenes were enriched for five transcription factors. Most cis-sQTL effects were found in intronic regions, with 27 sQTL variants associated with disease susceptibility and resistance in cattle. Key sGenes identified, such as GSDMA, NLRP6, CASP6, GZMA, CASP4, CASP1, TREM2, NLRP1, and NAIP, were related to inflammasome formation and pyroptosis. Additionally, genes like PIDD1, OPTN, NFKBIB, STAT1, TNIP3, and TREM2 were involved in regulating the NF-kB pathway. These findings lay the groundwork for breeding disease-resistant cattle and enhance our understanding of genetic mechanisms in immune responses.
Collapse
Affiliation(s)
- Thaís Cristina Ferreira Dos Santos
- Universidade Professor Edson Antônio Velano (UNIFENAS), Rodovia 179, Km 0, Alfenas, MG, 37132440, Brasil.
- Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, Brasil.
| | - Evandro Neves Silva
- Universidade Professor Edson Antônio Velano (UNIFENAS), Rodovia 179, Km 0, Alfenas, MG, 37132440, Brasil
- Universidade Federal de Alfenas (UNIFAL), Alfenas, MG, Brasil
| | | | - Bruna Maria Salatta
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, SP, Brasil
| | - Fernando Baldi
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, SP, Brasil
| | | | - Lucia Galvão De Albuquerque
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, SP, Brasil
- Conselho Nacional de Desenvolvimento Científico e Tecnológico, Brasília, DF, Brasil
| | - Maria Malane Magalhães Muniz
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, SP, Brasil
- University of Guelph, UOGELPH, Guelph, Canada
| | - Danielly Beraldo Dos Santos Silva
- Universidade Professor Edson Antônio Velano (UNIFENAS), Rodovia 179, Km 0, Alfenas, MG, 37132440, Brasil.
- Faculdade de Ciências Agrárias e Veterinárias (FCAV-UNESP), Jaboticabal, SP, Brasil.
| |
Collapse
|
25
|
Dang X, Teng Z, Yang Y, Li W, Liu J, Hui L, Zhou D, Gong D, Dai SS, Li Y, Li X, Lv L, Zeng Y, Yuan Y, Ma X, Liu Z, Li T, Luo XJ. Gene-level analysis reveals the genetic aetiology and therapeutic targets of schizophrenia. Nat Hum Behav 2025; 9:609-624. [PMID: 39753749 DOI: 10.1038/s41562-024-02091-4] [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: 04/18/2024] [Accepted: 11/18/2024] [Indexed: 03/27/2025]
Abstract
Genome-wide association studies (GWASs) have reported multiple risk loci for schizophrenia (SCZ). However, the majority of the associations were from populations of European ancestry. Here we conducted a large-scale GWAS in Eastern Asian populations (29,519 cases and 44,392 controls) and identified ten Eastern Asian-specific risk loci, two of which have not been previously reported. A further cross-ancestry GWAS meta-analysis (96,806 cases and 492,818 controls) including populations from diverse ancestries identified 61 previously unreported risk loci. Systematic variant-level analysis, including fine mapping, functional genomics and expression quantitative trait loci, prioritized potential causal variants. Gene-level analyses, including transcriptome-wide association study, proteome-wide association study and Mendelian randomization, nominated the potential causal genes. By integrating evidence from layers of different analyses, we prioritized the most plausible causal genes for SCZ, such as ACE, CNNM2, SNAP91, ABCB9 and GATAD2A. Finally, drug repurposing showed that ACE, CA14, MAPK3 and MAPT are potential therapeutic targets for SCZ. Our study not only showed the power of cross-ancestry GWAS in deciphering the genetic aetiology of SCZ, but also uncovered new genetic risk loci, potential causal variants and genes and therapeutic targets for SCZ.
Collapse
Affiliation(s)
- Xinglun Dang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Zhaowei Teng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Kunming, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Jiewei Liu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, China
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou, China
| | - Dongsheng Zhou
- Department of Psychiatry, Affiliated Kangning Hospital of Ningbo University (Ningbo Kangning Hospital), Ningbo, China
| | - Daohua Gong
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Shan-Shan Dai
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yifan Li
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Xingxing Li
- Department of Psychiatry, Affiliated Kangning Hospital of Ningbo University (Ningbo Kangning Hospital), Ningbo, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Yong Zeng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Kunming, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Xiancang Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Tao Li
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xiong-Jian Luo
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China.
| |
Collapse
|
26
|
Zheng H, Wang W, Qiu W, Feng Y. Association of antihypertensive drug target genes with stroke subtypes: A Mendelian randomization study. J Stroke Cerebrovasc Dis 2025; 34:108244. [PMID: 39826584 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108244] [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: 09/10/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Epidemiological and genetic studies have elucidated the effect of antihypertensive medication (AHM) on stroke subtypes varying upon drug classes, but which drug target genes, how, and where mediated this association remains unknown. We aimed to investigate the impact of AHM on stroke subtypes. METHODS Genetic instruments for the expression of AHM target genes were identified with expression quantitative trait loci in blood, which should be associated with systolic blood pressure (SBP) to proxy for the effect of AHM. Sensitivity analysis, including reverse causality detection, horizontal pleiotropy, phenotype scanning, tissue enrichment analyses, Bayesian colocalization, and linkage disequilibrium check, were utilized to validate our findings. RESULTS A 1-standard deviation (SD) decrease of KCNJ11 gene expression (acting on arteriolar smooth muscle) was associated with a decrease of 2.19 (95 % confidence interval (CI), 1.67-2.71) mmHg of SBP, and a decreased risk of stroke subtypes (Any stroke: odds ratio (OR): 0.80, 95 % CI: 0.70-0.90; Ischemic stroke: OR, 0.79; 95 % CI, 0.69-0.90), respectively. Similarly, a negative association was found between the gene expression of ADRB1 and the risk of small vessel stroke (SVS) (OR, 0.61; 95 % CI, 0.49-0.75). Colocalization supported the probability of shared causal variants for the KCNJ11 and ADRB1 genes in different stroke subtypes. NHLRC2, the nearby gene of ADRB1, was also associated with a higher risk of SVS. CONCLUSION Our study implies that changes in expression of KCNJ11 and ADRB1 mediated possibly via AHM may decrease stroke subtypes' risk and NHLRC2 is a potential therapy target gene of SVS.
Collapse
Affiliation(s)
- He Zheng
- School of Medicine, South China University of Technology, Guangzhou, China; Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wenbin Wang
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Weida Qiu
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yingqing Feng
- School of Medicine, South China University of Technology, Guangzhou, China; Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
27
|
Humphrey J, Brophy E, Kosoy R, Zeng B, Coccia E, Mattei D, Ravi A, Naito T, Efthymiou AG, Navarro E, De Sanctis C, Flores-Almazan V, Muller BZ, Snijders GJLJ, Allan A, Münch A, Kitata RB, Kleopoulos SP, Argyriou S, Malakates P, Psychogyiou K, Shao Z, Francoeur N, Tsai CF, Gritsenko MA, Monroe ME, Paurus VL, Weitz KK, Shi T, Sebra R, Liu T, de Witte LD, Goate AM, Bennett DA, Haroutunian V, Hoffman GE, Fullard JF, Roussos P, Raj T. Long-read RNA sequencing atlas of human microglia isoforms elucidates disease-associated genetic regulation of splicing. Nat Genet 2025; 57:604-615. [PMID: 40033057 DOI: 10.1038/s41588-025-02099-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/23/2025] [Indexed: 03/05/2025]
Abstract
Microglia, the innate immune cells of the central nervous system, have been genetically implicated in multiple neurodegenerative diseases. Mapping the genetics of gene expression in human microglia has identified several loci associated with disease-associated genetic variants in microglia-specific regulatory elements. However, identifying genetic effects on splicing is challenging because of the use of short sequencing reads. Here, we present the isoform-centric microglia genomic atlas (isoMiGA), which leverages long-read RNA sequencing to identify 35,879 novel microglia isoforms. We show that these isoforms are involved in stimulation response and brain region specificity. We then quantified the expression of both known and novel isoforms in a multi-ancestry meta-analysis of 555 human microglia short-read RNA sequencing samples from 391 donors, and found associations with genetic risk loci in Alzheimer's and Parkinson's disease. We nominate several loci that may act through complex changes in isoform and splice-site usage.
Collapse
Affiliation(s)
- Jack Humphrey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erica Brophy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roman Kosoy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Biao Zeng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elena Coccia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniele Mattei
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashvin Ravi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tatsuhiko Naito
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anastasia G Efthymiou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elisa Navarro
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biochemistry and Molecular Biology, Universidad Complutense de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto Ramon y Cajal de Investigacion Sanitaria (IRYCIS), Madrid, Spain
| | - Claudia De Sanctis
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Department of Artificial Intelligence & Human Health, Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victoria Flores-Almazan
- Department of Pathology, Department of Artificial Intelligence & Human Health, Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Z Muller
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gijsje J L J Snijders
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda Allan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra Münch
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reta Birhanu Kitata
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Steven P Kleopoulos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stathis Argyriou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Periklis Malakates
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Konstantina Psychogyiou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhiping Shao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nancy Francoeur
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Matthew E Monroe
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Vanessa L Paurus
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Tujin Shi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lot D de Witte
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Vahram Haroutunian
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA.
| | - Towfique Raj
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
28
|
O'Connell KS, Koromina M, van der Veen T, Boltz T, David FS, Yang JMK, Lin KH, Wang X, Coleman JRI, Mitchell BL, McGrouther CC, Rangan AV, Lind PA, Koch E, Harder A, Parker N, Bendl J, Adorjan K, Agerbo E, Albani D, Alemany S, Alliey-Rodriguez N, Als TD, Andlauer TFM, Antoniou A, Ask H, Bass N, Bauer M, Beins EC, Bigdeli TB, Pedersen CB, Boks MP, Børte S, Bosch R, Brum M, Brumpton BM, Brunkhorst-Kanaan N, Budde M, Bybjerg-Grauholm J, Byerley W, Cabana-Domínguez J, Cairns MJ, Carpiniello B, Casas M, Cervantes P, Chatzinakos C, Chen HC, Clarence T, Clarke TK, Claus I, Coombes B, Corfield EC, Cruceanu C, Cuellar-Barboza A, Czerski PM, Dafnas K, Dale AM, Dalkner N, Degenhardt F, DePaulo JR, Djurovic S, Drange OK, Escott-Price V, Fanous AH, Fellendorf FT, Ferrier IN, Forty L, Frank J, Frei O, Freimer NB, Fullard JF, Garnham J, Gizer IR, Gordon SD, Gordon-Smith K, Greenwood TA, Grove J, Guzman-Parra J, Ha TH, Hahn T, Haraldsson M, Hautzinger M, Havdahl A, Heilbronner U, Hellgren D, Herms S, Hickie IB, Hoffmann P, Holmans PA, Huang MC, Ikeda M, Jamain S, Johnson JS, Jonsson L, Kalman JL, Kamatani Y, Kennedy JL, Kim E, Kim J, Kittel-Schneider S, et alO'Connell KS, Koromina M, van der Veen T, Boltz T, David FS, Yang JMK, Lin KH, Wang X, Coleman JRI, Mitchell BL, McGrouther CC, Rangan AV, Lind PA, Koch E, Harder A, Parker N, Bendl J, Adorjan K, Agerbo E, Albani D, Alemany S, Alliey-Rodriguez N, Als TD, Andlauer TFM, Antoniou A, Ask H, Bass N, Bauer M, Beins EC, Bigdeli TB, Pedersen CB, Boks MP, Børte S, Bosch R, Brum M, Brumpton BM, Brunkhorst-Kanaan N, Budde M, Bybjerg-Grauholm J, Byerley W, Cabana-Domínguez J, Cairns MJ, Carpiniello B, Casas M, Cervantes P, Chatzinakos C, Chen HC, Clarence T, Clarke TK, Claus I, Coombes B, Corfield EC, Cruceanu C, Cuellar-Barboza A, Czerski PM, Dafnas K, Dale AM, Dalkner N, Degenhardt F, DePaulo JR, Djurovic S, Drange OK, Escott-Price V, Fanous AH, Fellendorf FT, Ferrier IN, Forty L, Frank J, Frei O, Freimer NB, Fullard JF, Garnham J, Gizer IR, Gordon SD, Gordon-Smith K, Greenwood TA, Grove J, Guzman-Parra J, Ha TH, Hahn T, Haraldsson M, Hautzinger M, Havdahl A, Heilbronner U, Hellgren D, Herms S, Hickie IB, Hoffmann P, Holmans PA, Huang MC, Ikeda M, Jamain S, Johnson JS, Jonsson L, Kalman JL, Kamatani Y, Kennedy JL, Kim E, Kim J, Kittel-Schneider S, Knowles JA, Kogevinas M, Kranz TM, Krebs K, Kushner SA, Lavebratt C, Lawrence J, Leber M, Lee HJ, Liao C, Lucae S, Lundberg M, MacIntyre DJ, Maier W, Maihofer AX, Malaspina D, Manchia M, Maratou E, Martinsson L, Mattheisen M, McGregor NW, McInnis MG, McKay JD, Medeiros H, Meyer-Lindenberg A, Millischer V, Morris DW, Moutsatsou P, Mühleisen TW, O'Donovan C, Olsen CM, Panagiotaropoulou G, Papiol S, Pardiñas AF, Park HY, Perry A, Pfennig A, Pisanu C, Potash JB, Quested D, Rapaport MH, Regeer EJ, Rice JP, Rivera M, Schulte EC, Senner F, Shadrin A, Shilling PD, Sigurdsson E, Sindermann L, Sirignano L, Siskind D, Slaney C, Sloofman LG, Smeland OB, Smith DJ, Sobell JL, Soler Artigas M, Stein DJ, Stein F, Su MH, Sung H, Świątkowska B, Terao C, Tesfaye M, Tesli M, Thorgeirsson TE, Thorp JG, Toma C, Tondo L, Tooney PA, Tsai SJ, Tsermpini EE, Vawter MP, Vedder H, Vreeker A, Walters JTR, Winsvold BS, Witt SH, Won HH, Ye R, Young AH, Zandi PP, Zillich L, 23andMe Research Team, Adolfsson R, Alda M, Alfredsson L, Backlund L, Baune BT, Bellivier F, Bengesser S, Berrettini WH, Biernacka JM, Boehnke M, Børglum AD, Breen G, Carr VJ, Catts S, Cichon S, Corvin A, Craddock N, Dannlowski U, Dikeos D, Etain B, Ferentinos P, Frye M, Fullerton JM, Gawlik M, Gershon ES, Goes FS, Green MJ, Grigoroiu-Serbanescu M, Hauser J, Henskens FA, Hjerling-Leffler J, Hougaard DM, Hveem K, Iwata N, Jones I, Jones LA, Kahn RS, Kelsoe JR, Kircher T, Kirov G, Kuo PH, Landén M, Leboyer M, Li QS, Lissowska J, Lochner C, Loughland C, Luykx JJ, Martin NG, Mathews CA, Mayoral F, McElroy SL, McIntosh AM, McMahon FJ, Medland SE, Melle I, Milani L, Mitchell PB, Morken G, Mors O, Mortensen PB, Müller-Myhsok B, Myers RM, Myung W, Neale BM, Nievergelt CM, Nordentoft M, Nöthen MM, Nurnberger JI, O'Donovan MC, Oedegaard KJ, Olsson T, Owen MJ, Paciga SA, Pantelis C, Pato CN, Pato MT, Patrinos GP, Pawlak JM, Ramos-Quiroga JA, Reif A, Reininghaus EZ, Ribasés M, Rietschel M, Ripke S, Rouleau GA, Roussos P, Saito T, Schall U, Schalling M, Schofield PR, Schulze TG, Scott LJ, Scott RJ, Serretti A, Smoller JW, Squassina A, Stahl EA, Stefansson H, Stefansson K, Stordal E, Streit F, Sullivan PF, Turecki G, Vaaler AE, Vieta E, Vincent JB, Waldman ID, Weickert CS, Weickert TW, Werge T, Whiteman DC, Zwart JA, Edenberg HJ, McQuillin A, Forstner AJ, Mullins N, Di Florio A, Ophoff RA, Andreassen OA, Bipolar Disorder Working Group of the Psychiatric Genomics Consortium. Genomics yields biological and phenotypic insights into bipolar disorder. Nature 2025; 639:968-975. [PMID: 39843750 DOI: 10.1038/s41586-024-08468-9] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 11/28/2024] [Indexed: 01/24/2025]
Abstract
Bipolar disorder is a leading contributor to the global burden of disease1. Despite high heritability (60-80%), the majority of the underlying genetic determinants remain unknown2. We analysed data from participants of European, East Asian, African American and Latino ancestries (n = 158,036 cases with bipolar disorder, 2.8 million controls), combining clinical, community and self-reported samples. We identified 298 genome-wide significant loci in the multi-ancestry meta-analysis, a fourfold increase over previous findings3, and identified an ancestry-specific association in the East Asian cohort. Integrating results from fine-mapping and other variant-to-gene mapping approaches identified 36 credible genes in the aetiology of bipolar disorder. Genes prioritized through fine-mapping were enriched for ultra-rare damaging missense and protein-truncating variations in cases with bipolar disorder4, highlighting convergence of common and rare variant signals. We report differences in the genetic architecture of bipolar disorder depending on the source of patient ascertainment and on bipolar disorder subtype (type I or type II). Several analyses implicate specific cell types in the pathophysiology of bipolar disorder, including GABAergic interneurons and medium spiny neurons. Together, these analyses provide additional insights into the genetic architecture and biological underpinnings of bipolar disorder.
Collapse
Affiliation(s)
- Kevin S O'Connell
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway.
| | - Maria Koromina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- 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
| | | | - Toni Boltz
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Friederike S David
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Jessica Mei Kay Yang
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | | | - Xin Wang
- 23andMe Inc., Sunnyvale, CA, USA
| | - Jonathan R I Coleman
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
- NIHR Maudsley BRC, King's College London, London, UK
| | - Brittany L Mitchell
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Aaditya V Rangan
- New York University, New York, NY, USA
- Flatiron Institute, New York, NY, USA
| | - Penelope A Lind
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences and Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Elise Koch
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway
| | - Arvid Harder
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Nadine Parker
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway
| | - Jaroslav Bendl
- Department of Psychiatry, 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
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristina Adorjan
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Esben Agerbo
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Diego Albani
- Department of Neuroscience, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Silvia Alemany
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d'Hebron Research Institut (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ney Alliey-Rodriguez
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
- Northwestern University, Chicago, IL, USA
| | - Thomas D Als
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- iSEQ, Center for Integrative Sequencing, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | - Till F M Andlauer
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Anastasia Antoniou
- National and Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
| | - Helga Ask
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- PROMENTA Research Centre, Department of Psychology, University of Oslo, Oslo, Norway
| | - Nicholas Bass
- Division of Psychiatry, University College London, London, UK
| | - Michael Bauer
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Eva C Beins
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Tim B Bigdeli
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- VA NY Harbor Healthcare System, Brooklyn, NY, USA
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Carsten Bøcker Pedersen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Marco P Boks
- Psychiatry, Brain Center UMC Utrecht, Utrecht, The Netherlands
| | - Sigrid Børte
- Research and Communication Unit for Musculoskeletal Health, Division of Clinical Neuroscience, Oslo University Hospital, Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rosa Bosch
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Programa SJD MIND Escoles, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Murielle Brum
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ben M Brumpton
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nathalie Brunkhorst-Kanaan
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Monika Budde
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
| | - Jonas Bybjerg-Grauholm
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - William Byerley
- Psychiatry, University of California San Francisco, San Francisco, CA, USA
| | - Judit Cabana-Domínguez
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d'Hebron Research Institut (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Bernardo Carpiniello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Miquel Casas
- Programa SJD MIND Escoles, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Fundació Privada d'Investigació Sant Pau (FISP), Barcelona, Spain
| | - Pablo Cervantes
- Department of Psychiatry, Mood Disorders Program, McGill University Health Center, Montreal, Québec, Canada
| | - Chris Chatzinakos
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Hsi-Chung Chen
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
- Department of Psychiatry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tereza Clarence
- Department of Psychiatry, 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
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Toni-Kim Clarke
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Isabelle Claus
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Brandon Coombes
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth C Corfield
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Nic Waals Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Oslo, Norway
| | - Cristiana Cruceanu
- Department of Psychiatry, Mood Disorders Program, McGill University Health Center, Montreal, Québec, Canada
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Alfredo Cuellar-Barboza
- Department of Psychiatry, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Piotr M Czerski
- Department of Psychiatry, Laboratory of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Konstantinos Dafnas
- National and Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, Departments of Neurosciences, Radiology, and Psychiatry, University of California, San Diego, CA, USA
| | - Nina Dalkner
- Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - J Raymond DePaulo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital Ullevål, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole Kristian Drange
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway
- Department of Psychiatry, Sørlandet Hospital, Kristiansand, Norway
| | - Valentina Escott-Price
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Ayman H Fanous
- Department of Psychiatry, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
- Carl T. Hayden Veterans Affairs Medical Center, Phoenix, AZ, USA
- Banner-University Medical Center, Phoenix, AZ, USA
| | - Frederike T Fellendorf
- Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - I Nicol Ferrier
- Academic Psychiatry, Newcastle University, Newcastle upon Tyne, UK
| | - Liz Forty
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Oleksandr Frei
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Science, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - John F Fullard
- Department of Psychiatry, 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
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie Garnham
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Scott D Gordon
- Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Tiffany A Greenwood
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jakob Grove
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Department of Biomedicine and the iSEQ Center, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, CGPM, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - José Guzman-Parra
- Mental Health Department, University Regional Hospital, Biomedicine Institute (IBIMA), Málaga, Spain
| | - Tae Hyon Ha
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Neuropsychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Magnus Haraldsson
- Faculty of Medicine, Department of Psychiatry, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Landspitali University Hospital, Reykjavik, Iceland
| | - Martin Hautzinger
- Department of Psychology, Eberhard Karls Universität Tübingen, Tubingen, Germany
| | - Alexandra Havdahl
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- PROMENTA Research Centre, Department of Psychology, University of Oslo, Oslo, Norway
- Nic Waals Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
| | - Dennis Hellgren
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Ian B Hickie
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Peter A Holmans
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Ming-Chyi Huang
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan
| | - Masashi Ikeda
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Stéphane Jamain
- Université Paris Est Créteil, INSERM, IMRB, Translational Neuropsychiatry, Créteil, France
| | - Jessica S Johnson
- Department of Psychiatry, 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
- Department of Psychiatry, UNC Chapel Hill School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lina Jonsson
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Janos L Kalman
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - James L Kennedy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Neurogenetics Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Euitae Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Neuropsychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| | - Jaeyoung Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Sarah Kittel-Schneider
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| | - James A Knowles
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | | | - Thorsten M Kranz
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Steven A Kushner
- Department of Psychiatry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Catharina Lavebratt
- Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jacob Lawrence
- Psychiatry, North East London NHS Foundation Trust, Ilford, UK
| | - Markus Leber
- Clinic for Psychiatry and Psychotherapy, University Hospital Cologne, Cologne, Germany
| | - Heon-Jeong Lee
- Department of Psychiatry, Korea University College of Medicine, Seoul, Republic of Korea
| | - Calwing Liao
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Susanne Lucae
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Martin Lundberg
- Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Donald J MacIntyre
- Division of Psychiatry, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Adam X Maihofer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Research/Psychiatry, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Dolores Malaspina
- Department of Psychiatry, 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
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Eirini Maratou
- National and Kapodistrian University of Athens, Medical School, Clinical Biochemistry Laboratory, Attikon General Hospital, Athens, Greece
| | - Lina Martinsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Psychiatry Research, SLSO Region Stockholm, Stockholm, Sweden
| | - Manuel Mattheisen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- iSEQ, Center for Integrative Sequencing, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Nathaniel W McGregor
- Human and Systems Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Melvin G McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - James D McKay
- Genetic Cancer Susceptibility Group, International Agency for Research on Cancer, Lyon, France
| | - Helena Medeiros
- Institute for Genomic Health, SUNY Downstate Medical Center College of Medicine, Brooklyn, NY, USA
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Centre for Mental Health (DZPG), partner site Mannheim-Heidelberg-Ulm, Mannheim, Germany
| | - Vincent Millischer
- Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Derek W Morris
- Centre for Neuroimaging and Cognitive Genomics (NICOG), School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Paraskevi Moutsatsou
- National and Kapodistrian University of Athens, Medical School, Clinical Biochemistry Laboratory, Attikon General Hospital, Athens, Greece
| | - Thomas W Mühleisen
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Claire O'Donovan
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Catherine M Olsen
- Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Sergi Papiol
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Antonio F Pardiñas
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Hye Youn Park
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Neuropsychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Amy Perry
- Psychological Medicine, University of Worcester, Worcester, UK
| | - Andrea Pfennig
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Claudia Pisanu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - James B Potash
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Digby Quested
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Mark H Rapaport
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Eline J Regeer
- Outpatient Clinic for Bipolar Disorder, Altrecht, Utrecht, The Netherlands
| | - John P Rice
- Department of Psychiatry, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Margarita Rivera
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
- Institute of Neurosciences 'Federico Olóriz', Biomedical Research Center (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Eva C Schulte
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Fanny Senner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Alexey Shadrin
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Paul D Shilling
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Engilbert Sigurdsson
- Faculty of Medicine, Department of Psychiatry, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Landspitali University Hospital, Reykjavik, Iceland
| | - Lisa Sindermann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Lea Sirignano
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dan Siskind
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Claire Slaney
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Laura G Sloofman
- Department of Psychiatry, 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
| | - Olav B Smeland
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway
| | - Daniel J Smith
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Janet L Sobell
- Psychiatry and the Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - Maria Soler Artigas
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d'Hebron Research Institut (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Dan J Stein
- SAMRC Unit on Risk and Resilience in Mental Disorders, Dept of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Mei-Hsin Su
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Heejong Sung
- Human Genetics Branch, Intramural Research Program, National Institute of Mental Health, Bethesda, MD, USA
| | - Beata Świątkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Markos Tesfaye
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Martin Tesli
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway
- Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Jackson G Thorp
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Claudio Toma
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid and CSIC, Madrid, Spain
| | - Leonardo Tondo
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Paul A Tooney
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Marquis P Vawter
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, CA, USA
| | - Helmut Vedder
- Psychiatry, Psychiatrisches Zentrum Nordbaden, Wiesloch, Germany
| | - Annabel Vreeker
- Psychiatry, Brain Center UMC Utrecht, Utrecht, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC Sophia Children Hospital, Erasmus University, Rotterdam, The Netherlands
- Department of Psychology Education and Child Studies, Erasmus School of Social and Behavioral Sciences, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Bendik S Winsvold
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hong-Hee Won
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Robert Ye
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Allan H Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Kent, UK
| | - Peter P Zandi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Rolf Adolfsson
- Department of Clinical Sciences, Psychiatry, Umeå University Medical Faculty, Umeå, Sweden
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
- National Institute of Mental Health, Klecany, Czech Republic
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Backlund
- Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Frank Bellivier
- Université Paris Cité, INSERM, Optimisation Thérapeutique en Neuropsychopharmacologie, UMRS-1144, Paris, France
- APHP Nord, DMU Neurosciences, GHU Saint Louis-Lariboisière-Fernand Widal, Département de Psychiatrie et de Médecine Addictologique, Paris, France
| | - Susanne Bengesser
- Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | | | - Joanna M Biernacka
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Michael Boehnke
- Center for Statistical Genetics and Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Anders D Børglum
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Department of Biomedicine and the iSEQ Center, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, CGPM, Aarhus, Denmark
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
- NIHR Maudsley BRC, King's College London, London, UK
| | - Vaughan J Carr
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Stanley Catts
- University of Queensland, Brisbane, Queensland, Australia
| | - Sven Cichon
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Nicholas Craddock
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dimitris Dikeos
- National and Kapodistrian University of Athens, 1st Department of Psychiatry, Eginition Hospital, Athens, Greece
| | - Bruno Etain
- Université Paris Cité, INSERM, Optimisation Thérapeutique en Neuropsychopharmacologie, UMRS-1144, Paris, France
- APHP Nord, DMU Neurosciences, GHU Saint Louis-Lariboisière-Fernand Widal, Département de Psychiatrie et de Médecine Addictologique, Paris, France
| | - Panagiotis Ferentinos
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
- National and Kapodistrian University of Athens, 2nd Department of Psychiatry, Attikon General Hospital, Athens, Greece
| | - Mark Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Micha Gawlik
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| | - Elliot S Gershon
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Melissa J Green
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Maria Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Clinical Psychiatric Hospital, Bucharest, Romania
| | - Joanna Hauser
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Frans A Henskens
- School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - David M Hougaard
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Kristian Hveem
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Ian Jones
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Lisa A Jones
- Psychological Medicine, University of Worcester, Worcester, UK
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Psychiatry, Brain Center UMC Utrecht, Utrecht, The Netherlands
| | - John R Kelsoe
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - George Kirov
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Po-Hsiu Kuo
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
- Department of Public Health and Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Marion Leboyer
- Université Paris Est Créteil, INSERM, IMRB, Translational Neuropsychiatry, Créteil, France
| | - Qingqin S Li
- Neuroscience Therapeutic Area, Janssen Research and Development, Titusville, NJ, USA
- JRD Data Science, Janssen Research and Development, Titusville, NJ, USA
| | - Jolanta Lissowska
- Cancer Epidemiology and Prevention, M. Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Christine Lochner
- SA MRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | | | - Jurjen J Luykx
- Department of Psychiatry, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nicholas G Martin
- Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Carol A Mathews
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Fermin Mayoral
- Mental Health Department, University Regional Hospital, Biomedicine Institute (IBIMA), Málaga, Spain
| | | | - Andrew M McIntosh
- Division of Psychiatry, Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Francis J McMahon
- Human Genetics Branch, Intramural Research Program, National Institute of Mental Health, Bethesda, MD, USA
| | - Sarah E Medland
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ingrid Melle
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Division of Mental Health and Addiction, University of Oslo, Institute of Clinical Medicine, Oslo, Norway
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Philip B Mitchell
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Gunnar Morken
- Department of Mental Health, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Psychiatry, St Olavs University Hospital, Trondheim, Norway
| | - Ole Mors
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital-Psychiatry, Risskov, Denmark
| | - Preben Bo Mortensen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- NCRR and CIRRAU, Aarhus BSS, Aarhus University, Aarhus, Denmark
| | - Bertram Müller-Myhsok
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- University of Liverpool, Liverpool, UK
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Neuropsychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Research/Psychiatry, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Merete Nordentoft
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Mental Health Services in the Capital Region of Denmark, Mental Health Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - John I Nurnberger
- Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Ketil J Oedegaard
- Division of Psychiatry, Haukeland Universitetssjukehus, Bergen, Norway
- Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Stockholm, Sweden
| | - Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Sara A Paciga
- Human Genetics and Computational Biomedicine, Pfizer Global Research and Development, Groton, CT, USA
| | - Christos Pantelis
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, Victoria, Australia
- Monash Institute of Pharmaceutical Sciences (MIPS), Monash University, Parkville, Victoria, Australia
| | - Carlos N Pato
- Rutgers Health, Rutgers University, Piscataway, NJ, USA
| | | | - George P Patrinos
- University of Patras, School of Health Sciences, Department of Pharmacy, Laboratory of Pharmacogenomics and Individualized Therapy, Patras, Greece
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Department of Pathology, Faculty of Medicine and Health Sciences, Clinical Bioinformatics Unit, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joanna M Pawlak
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Josep Antoni Ramos-Quiroga
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d'Hebron Research Institut (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Eva Z Reininghaus
- Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Marta Ribasés
- Instituto de Salud Carlos III, Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Group of Psychiatry Mental Health and Addictions, Vall d'Hebron Research Institut (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
- Montreal Neurological Institute and Hospital, McGill University, Montréal, Québec, Canada
| | - Panos Roussos
- Department of Psychiatry, 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
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Takeo Saito
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Ulrich Schall
- Centre for Brain and Mental Health Research, The University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambtion Heights, New South Wales, Australia
| | - Martin Schalling
- Translational Psychiatry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Laura J Scott
- Center for Statistical Genetics and Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Rodney J Scott
- The School of Biomedical Sciences and Pharmacy, Faculty of Medicine, Health and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
- Cancer Detection and Therapies Program, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Alessandro Serretti
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| | - Jordan W Smoller
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Massachusetts General Hospital, Boston, MA, USA
| | - Alessio Squassina
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Eli A Stahl
- Department of Psychiatry, 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
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | | | - Kari Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eystein Stordal
- Department of Psychiatry, Hospital Namsos, Namsos, Norway
- Department of Neuroscience, Norges Teknisk Naturvitenskapelige Universitet Fakultet for naturvitenskap og teknologi, Trondheim, Norway
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gustavo Turecki
- Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Arne E Vaaler
- Department of Psychiatry, Sankt Olavs Hospital Universitetssykehuset i Trondheim, Trondheim, Norway
| | - Eduard Vieta
- Clinical Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Spain
| | - John B Vincent
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Irwin D Waldman
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Cynthia S Weickert
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Thomas W Weickert
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Thomas Werge
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Center for GeoGenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - David C Whiteman
- Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - John-Anker Zwart
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| | - Howard J Edenberg
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Centre for Human Genetics, University of Marburg, Marburg, Germany
| | - Niamh Mullins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- 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
| | - Arianna Di Florio
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Roel A Ophoff
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Science, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.
- Center for Precision Psychiatry, University of Oslo, Oslo, Norway.
| | | |
Collapse
Collaborators
Byung-Chul Lee, Ji-Woong Kim, Young Kee Lee, Joon Ho Kang, Myeong Jae Cheon, Dong Jun Kim, Mihaela Aslan, Philip D Harvey, Grant D Huang,
Collapse
|
29
|
Deng MG, Wang K, Liu F, Zhou X, Nie JQ, Zhao ZH, Liu J. Shared genetic architecture and causal relationship between frailty and schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2025; 11:24. [PMID: 39984493 PMCID: PMC11845589 DOI: 10.1038/s41537-024-00550-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/20/2024] [Indexed: 02/23/2025]
Abstract
The complex relationship between frailty and schizophrenia has yet to be fully understood. This study aims to clarify their relationship by investigating their genetic links. We hypothesize a shared genetic architecture and a bidirectional causal relationship between the two conditions. Utilizing summary genetic data from European genome-wide association studies, we analyzed genetic associations through global and local correlations, shared genomic loci, tissue enrichments, and functional genes. Bidirectional Mendelian Randomization (MR) was employed to infer causality. Our findings show a positive genetic correlation between frailty and schizophrenia (LDSC: rg = 0.117, p = 6.686 × 10-7; HDL: rg = 0.101, p = 5.63 × 10-13) and local correlations in three genomic regions (chr9: 94167203-96671698, p = 2.21 × 10-6; chr11: 112459488-114257728, p = 1.01 × 10-5; and chr18: 77149991-78017158, p = 9.57 × 10-6). We identified 111 genomic loci associated with both conditions and demonstrated that genetic variants for frailty and schizophrenia share tissue enrichments and functional genes in brain. MR analysis suggests that frailty increases the likelihood of schizophrenia (OR: 1.763, 95% CI: 1.259-2.468, p = 0.001) and vice versa (β: 0.012, 95% CI: 0.006-0.018, p < 0.001). Our research supports the presence of a shared genetic basis and bidirectional causality between frailty and schizophrenia. These findings necessitate further investigation in diverse populations to confirm and expand on this genetic understanding.
Collapse
Affiliation(s)
- Ming-Gang Deng
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, China.
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, China.
| | - Kai Wang
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, 430000, Hubei, China
| | - Fang Liu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - Xiuxiu Zhou
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, China
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, China
| | - Jia-Qi Nie
- Xiaogan Center for Disease Control and Prevention, Xiaogan, 432000, Hubei, China
| | - Zhi-Hui Zhao
- School of Nursing, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jiewei Liu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, China.
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, China.
| |
Collapse
|
30
|
Zhang Y, Zhang CY, Yuan J, Jiang H, Sun P, Hui L, Xu L, Yu L, Guo Z, Wang L, Yang Y, Li M, Li SW, Yang J, Li W, Teng Z, Xiao X. Human mood disorder risk gene Synaptotagmin-14 contributes to mania-like behaviors in mice. Mol Psychiatry 2025:10.1038/s41380-025-02933-1. [PMID: 39966626 DOI: 10.1038/s41380-025-02933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Bipolar disorder (BD) and major depressive disorder (MDD) are the most prevalent mood disorders and cause considerable burden worldwide. Compelling evidence suggests a pronounced overlap between these two disorders in clinical symptoms, treatment strategies, and genetic etiology. Here we leverage a BD GWAS (1822 cases and 4650 controls) and a MDD GWAS (5303 cases and 5337 controls), followed by independent replications, to investigate their shared genetic basis among Han Chinese. We have herein identified a lead SNP rs126277 at the 1q32.2 locus, which also exhibited nominal associations with mood disorders and several relevant sub-clinical phenotypes (e.g., mania) in European populations. Bulk tissue and single-cell eQTL analyses suggest that the risk G-allele of rs126277 predicted lower SYT14 mRNA expression in human brains. We generated mice lacking Syt14 (Syt14-/-) and mice with insufficient expression of Syt14 in the hippocampus (Syt14-KD), and found that depletion of Syt14 resulted in mania-like behaviors including hyperactivity and anti-depressive behaviors, resembling aspects of mood disorders. We also confirmed that deficiency of this gene in the hippocampus was sufficient to induce hyperactivity in mice. RNA-sequencing analyses of the hippocampus of Syt14-/- mice revealed significant upregulation of Per1 as well as downregulation of Slc7a11 and Ptprb. Ultrastructural analyses showed significant alteration of the number of vesicles within 50 nm to the active zone and the width of synaptic cleft in the ventral hippocampus of Syt14-/- mice compared with the control mice. Overall, we have identified a novel mood disorder risk gene SYT14, and confirmed its impact on mania-like behaviors. While the current study identifies an essential mood disorder risk gene, further investigations elucidating the detailed mechanisms by which SYT14 contributes to the pathogenesis of the illnesses are needed.
Collapse
Affiliation(s)
- Yue Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jing Yuan
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongyan Jiang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ping Sun
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Li Hui
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li Xu
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ling Yu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zeyi Guo
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Lu Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yi Yang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shi-Wu Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jianzhong Yang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wei Li
- Department of Blood Transfusion, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiao Xiao
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
31
|
Gong B, Xiao C, Feng Y, Shen J. NEK4: prediction of available drug targets and common genetic linkages in bipolar disorder and major depressive disorder. Front Psychiatry 2025; 16:1414015. [PMID: 39950180 PMCID: PMC11821612 DOI: 10.3389/fpsyt.2025.1414015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Background Bipolar disorder (BD) is a mental illness characterized by alternating episodes of elevated mood and depression, while major depressive disorder (MDD) is a debilitating condition that ranks second globally in terms of disease burden. Pharmacotherapy plays a crucial role in managing both BD and MDD. We investigated the genetic differences in populations of individuals with MDD and BD, and from a genetic perspective, we offered new insights into potential drug targets. This will provide clues to potential drug targets. Methods This study employed genome-wide association studies (GWAS) and summary-data-based Mendelian randomization (SMR) methods to investigate the genetic underpinnings of patients with bipolar disorder (BD) and major depressive disorder (MDD) and to predict potential drug target genes. Genetic variants associated with BD and MDD were identified through large-scale GWAS datasets. For BD, the study utilized a comprehensive meta-analysis comprising 57 BD cohorts from Europe, North America, and Australia, including 41,917 BD cases and 371,549 controls of European ancestry. This dataset included both type 1 and type 2 BD cases diagnosed based on DSM-IV, ICD-9, or ICD-10 criteria through standardized assessments. For MDD, we used data from a meta-analysis by Howard DM et al., which integrated the largest GWAS studies of MDD, totaling 246,363 cases and 561,190 controls. The SMR approach, combined with expression quantitative trait loci (eQTL) data, was then applied to assess causal associations between these genetic variants and gene expression, aiming to identify genetic markers and potential drug targets associated with BD and MDD. Furthermore, two-sample Mendelian randomization (TSMR) analyses were performed to explore causal links between protein quantitative trait loci (pQTL) and these disorders. Results The SMR analysis revealed 41 druggable genes associated with BD, of which five genes appeared in both brain tissue and blood eQTL datasets and were significantly associated with BD risk. Furthermore, 45 druggable genes were found to be associated with MDD by SMR analysis, of which three genes appeared simultaneously in both datasets and were significantly associated with MDD risk. NEK4, a common drug candidate gene for BD and MDD, was also significantly associated with a high risk of both diseases and may help differentiate between type 1 and type 2 BD. Specifically, NEK4 showed a strong association with BD (β brain=0.126, P FDR=0.001; βblood=1.158, P FDR=0.003) and MDD (β brain=0.0316, P FDR=0.022; βblood=0.254, P FDR=0.045). Additionally, NEK4 was notably linked to BD type 1 (βbrain=0.123, P FDR=2.97E-05; βblood=1.018, P FDR=0.002), but showed no significant association with BD type 2.Moreover, TSMR analysis identified four proteins (BMP1, F9, ITIH3, and SIGIRR) affecting the risk of BD, and PSMB4 affecting the risk of MDD. Conclusion Our study identified NEK4 as a key gene linked to both bipolar disorder (BD) and major depressive disorder (MDD), suggesting its potential as a drug target and a biomarker for differentiating BD subtypes. Using GWAS, SMR, and TSMR approaches, we revealed multiple druggable genes and protein associations with BD and MDD risk, providing new insights into the genetic basis of these disorders. These findings offer promising directions for precision medicine and novel therapeutic strategies in mental health treatment.
Collapse
Affiliation(s)
- Bin Gong
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, China
| | - Chenxu Xiao
- Department of Clinical Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Yu Feng
- Department of Clinical Medicine, The University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Medicine, The University of New South Wales, Sydney, NSW, Australia
| | - Jing Shen
- Department of Clinical Medicine, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
32
|
Zhao S, Sinson JC, Li S, Rosenfeld JA, Zapata G, Macakova K, Pena M, Maywald B, Worley KC, Burrage L, Hubshman MW, Ketkar S, Craigen W, Emrick L, Clark T, Lithwick GY, Shipony Z, Eng C, Lee B, Liu P. The Utility of Ultra-Deep RNA sequencing in Mendelian Disorder Diagnostics. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.28.25321295. [PMID: 39974001 PMCID: PMC11838946 DOI: 10.1101/2025.01.28.25321295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Clinical RNA-seq has become an essential tool for resolving variants of uncertain significance (VUS), particularly those affecting gene expression and splicing. However, most reference data and diagnostic protocols employ relatively modest sequencing depths (∼50-150 million reads), which may fail to capture low-abundance transcripts and rare splicing events critical for accurate diagnoses. We evaluated the diagnostic and translational utility of ultra-high-depth (up to ∼one billion unique reads) RNA-seq in four clinically accessible tissues (blood, fibroblast, LCL, and iPSC) using Ultima sequencing platform. After validating the performance of Ultima RNA-seq, we investigated how increasing depth affects gene and isoform detection, splicing variant discovery, and clinical interpretation of VUS. Deep RNA-seq substantially improved sensitivity for detecting lowly-expressed genes and isoforms. At ∼1 billion reads, near-saturation was achieved for gene-level detection, although isoform-level coverage continued to benefit from even deeper sequencing. In two clinical cases with VUS, pathogenic splicing abnormalities were undetected at ∼50 million reads but emerged at 200 million reads, becoming even more pronounced at ∼one billion reads. Using deep RNA-seq data, we constructed a novel resource, MRSD-deep, to estimate the minimum required sequencing depth to achieve desired coverage thresholds. MRSD-deep provided gene- and junction-level guidelines, aiding labs in selecting suitable coverage targets for specific applications. Leveraging deep RNA-seq data on fibroblast, we also built an expanded splicing-variation reference that successfully identified rare splicing events missed by standard-depth data. Our findings underscore the diagnostic and research benefits of deep RNA-seq for Mendelian disease investigations. By capturing rare transcripts and splicing events, ultra-high-depth RNA-seq can facilitate more definitive variant interpretations and enrich splicing-reference databases. We anticipate that cost-effective deep sequencing technologies and robust reference cohorts will further advance RNA-based diagnostics in precision medicine.
Collapse
|
33
|
Gao Y, Wang D, Wang Q, Wang J, Li S, Wang T, Hu X, Wan C. Causal Impacts of Psychiatric Disorders on Cognition and the Mediating Effect of Oxidative Stress: A Mendelian Randomization Study. Antioxidants (Basel) 2025; 14:162. [PMID: 40002349 PMCID: PMC11852177 DOI: 10.3390/antiox14020162] [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: 12/31/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Many psychiatric disorders are associated with major cognitive deficits. However, it is uncertain whether these deficits develop as a result of psychiatric disorders and what shared risk factors might mediate this relationship. Here, we utilized the Mendelian randomization (MR) analysis to investigate the complex causal relationship between nine major psychiatric disorders and three cognitive phenotypes, while also examining the potential mediating role of oxidative stress as a shared biological underpinning. Schizophrenia (SZ), major depressive disorder (MDD), and attention deficit hyperactivity disorder (ADHD) showed a decreasing effect on cognitive performance, intelligence, and education, while bipolar disorder (BPD) increased educational attainment. MR-Clust results exhibit the shared genetic basis between SZ and other psychiatric disorders in relation to cognitive function. Furthermore, when oxidative stress was considered as a potential mediating factor, the associations between SZ and the three dimensions of cognition, as well as between MDD and intelligence and ADHD and intelligence, exhibited larger effect sizes than the overall. Mediation MR analysis also supported the causal effects between psychiatric disorders and cognition via oxidative stress traits, including carotene, vitamin E, bilirubin, and uric acid. Finally, summary-based MR identified 29 potential causal associations of oxidative stress genes with both cognitive performance and psychiatric disorders. Our findings highlight the importance of considering oxidative stress in understanding and potentially treating cognitive impairments associated with psychiatric conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for The Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.G.); (D.W.); (Q.W.); (J.W.); (S.L.); (T.W.)
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for The Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.G.); (D.W.); (Q.W.); (J.W.); (S.L.); (T.W.)
| |
Collapse
|
34
|
Dou L, Xu Z, Xu J, Zang C, Su C, Pieper AA, Leverenz JB, Wang F, Zhu X, Cummings J, Cheng F. A network-based systems genetics framework identifies pathobiology and drug repurposing in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:22. [PMID: 39837893 PMCID: PMC11751448 DOI: 10.1038/s41531-025-00870-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/06/2025] [Indexed: 01/23/2025] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder. However, current treatments only manage symptoms and lack the ability to slow or prevent disease progression. We utilized a systems genetics approach to identify potential risk genes and repurposable drugs for PD. First, we leveraged non-coding genome-wide association studies (GWAS) loci effects on five types of brain-specific quantitative trait loci (xQTLs, including expression, protein, splicing, methylation and histone acetylation) under the protein-protein interactome (PPI) network. We then prioritized 175 PD likely risk genes (pdRGs), such as SNCA, CTSB, LRRK2, DGKQ, and CD44, which are enriched in druggable targets and differentially expressed genes across multiple human brain-specific cell types. Integrating network proximity-based drug repurposing and patient electronic health record (EHR) data observations, we identified Simvastatin as being significantly associated with reduced incidence of PD (hazard ratio (HR) = 0.91 for fall outcome, 95% confidence interval (CI): 0.87-0.94; HR = 0.88 for dementia outcome, 95% CI: 0.86-0.89) after adjusting for 267 covariates. In summary, our network-based systems genetics framework identifies potential risk genes and repurposable drugs for PD and other neurodegenerative diseases if broadly applied.
Collapse
Affiliation(s)
- Lijun Dou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Zhenxing Xu
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Chengxi Zang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Chang Su
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - James B Leverenz
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, UNLV, Las Vegas, NV, 89154, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
35
|
Song Q, Zhang C, Wang W, Wang C, Yi C. Exploring the genetic landscape of the brain-heart axis: A comprehensive analysis of pleiotropic effects between heart disease and psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111172. [PMID: 39423935 DOI: 10.1016/j.pnpbp.2024.111172] [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: 06/27/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The genetic links between heart disease and psychiatric disorders are complex and not well understood. This study uses genome-wide association studies (GWAS) and advanced multilevel analyses to explore these connections. METHODS We analyzed GWAS data from seven psychiatric disorders and five types of heart disease. Genetic correlations and overlaps were examined using linkage disequilibrium score regression (LDSC), high-definition likelihood (HDL), and Genetic analysis incorporating Pleiotropy and Annotation (GPA). Pleiotropic single-nucleotide variations (SNVs) were identified with pleiotropic analysis under the composite null hypothesis (PLACO) and annotated via Functional mapping and annotation of genetic associations (FUMA). Potential pleiotropic genes were identified using Multi-marker Analysis of GenoMic Annotation (MAGMA) and Summary data-based Mendelian Randomization (SMR). RESULTS Among 35 trait pairs, 32 showed significant genetic correlations or overlaps. PLACO identified 15,077 SNVs, with 287 recognized as pleiotropic loci and 20 colocalization sites. MAGMA and SMR revealed 75 potential pleiotropic genes involved in diverse pathways, including cancer, neurodevelopment, and cellular organization. Mouse Genome Informatics (MGI) queries provided evidence linking multiple genes to heart or psychiatric disorders. CONCLUSIONS This analysis reveals loci and genes with pleiotropic effects between heart disease and psychiatric disorders, highlighting shared biological pathways. These findings illuminate the genetic mechanisms underlying the brain-heart axis and suggest shared biological foundations for these conditions, offering potential targets for future prevention and treatment strategies.
Collapse
Affiliation(s)
- Qifeng Song
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Cheng Zhang
- Nanjing Vocational Health College, Nanjing, Jiangsu 210000, China
| | - Wei Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Cihan Wang
- Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Chenlong Yi
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Zheng H, Chen C, Feng Y. Association of antihypertensive drug target genes with alzheimer's disease: a mendelian randomization study. Alzheimers Res Ther 2025; 17:18. [PMID: 39794838 PMCID: PMC11720623 DOI: 10.1186/s13195-025-01671-4] [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: 01/12/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Epidemiological and genetic studies have elucidated associations between antihypertensive medication and Alzheimer's disease (AD), with the directionality of these associations varying upon the specific class of antihypertensive agents. METHODS Genetic instruments for the expression of antihypertensive drug target genes were identified using expression quantitative trait loci (eQTL) in blood, which are associated with systolic blood pressure (SBP). Exposure was derived from existing eQTL data in blood from the eQTLGen consortium and in the brain from the PsychENCODE and subsequently replicated in GTEx V8 and BrainMeta V2. We performed two-sample Mendelian randomization (MR) to estimate the potential effect of different antihypertensive drug classes on AD using summary statistics from a meta-analysis (111,326 cases and 677,663 controls) and further replicated in FinnGen cohorts (9301 cases and 367,976 controls). The reverse causality detection, assessing horizontal pleiotropy, Bayesian co-localization, phenotype scanning, and protein quantitative trait loci (pQTL) analysis were implemented to consolidate the MR findings further. RESULTS A 1-standard deviation (SD) lower expression of the angiotensin-converting enzyme (ACE) gene in blood was associated with a lower SBP of 3.92 (95% confidence interval (CI), 2.69-5.15) mmHg but an increased risk of AD (odds ratio (OR), 2.46; 95% CI, 1.82-3.33). A similar direction of association was also observed between ACE expression in prefrontal cortex (OR, 1.19; 95% CI, 1.10-1.28), frontal cortex (OR, 1.19; 95% CI, 1.11-1.27), cerebellum (OR, 1.13; 95% CI, 1.09-1.17), cortex (OR, 1.59; 95% CI, 1.33-1.28) and ACE protein levels in plasma (OR, 1.13; 95% CI, 1.09-1.17) and AD risk. Colocalization supports these results. Similar results were found in external validation. We found no evidence for an association between genetically estimated blood pressure (BP) and AD risk. CONCLUSIONS There findings suggest an adverse association of lower ACE messenger RNA and protein levels with an elevated risk of AD, irrespective of its BP-lowering effects. These findings warrant greater pharmacovigilance and further investigation into the effect of ACE inhibitors, particularly those that are centrally acting, on neurodegenerative symptoms in patients with AD.
Collapse
Affiliation(s)
- He Zheng
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chaolei Chen
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yingqing Feng
- School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
37
|
Liu S, Cho MY, Huang YN, Park T, Chaudhuri S, Rosewood TJ, Bice PJ, Chung D, Bennett DA, Ertekin-Taner N, Saykin AJ, Nho K. Multi-Omics Analysis for Identifying Cell-Type-Specific Druggable Targets in Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.08.25320199. [PMID: 39830273 PMCID: PMC11741481 DOI: 10.1101/2025.01.08.25320199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background Analyzing disease-linked genetic variants via expression quantitative trait loci (eQTLs) is important for identifying potential disease-causing genes. Previous research prioritized genes by integrating Genome-Wide Association Study (GWAS) results with tissue-level eQTLs. Recent studies have explored brain cell type-specific eQTLs, but they lack a systematic analysis across various Alzheimer's disease (AD) GWAS datasets, nor did they compare effects between tissue and cell type levels or across different cell type-specific eQTL datasets. In this study, we integrated brain cell type-specific eQTL datasets with AD GWAS datasets to identify potential causal genes at the cell type level. Methods To prioritize disease-causing genes, we used Summary Data-Based Mendelian Randomization (SMR) and Bayesian Colocalization (COLOC) to integrate AD GWAS summary statistics with cell-type-specific eQTLs. Combining data from five AD GWAS, three single-cell eQTL datasets, and one bulk tissue eQTL meta-analysis, we identified and confirmed both novel and known candidate causal genes. We investigated gene regulation through enhancer activity using H3K27ac and ATAC-seq data, performed protein-protein interaction and pathway enrichment analyses, and conducted a drug/compound enrichment analysis with the Drug Signatures Database (DSigDB) to support drug repurposing for AD. Results We identified 27 candidate causal genes for AD using cell type-specific eQTL datasets, with the highest numbers in microglia, followed by excitatory neurons, astrocytes, inhibitory neurons, oligodendrocytes, and oligodendrocyte precursor cells (OPCs). PABPC1 emerged as a novel astrocyte-specific gene. Our analysis revealed protein-protein interaction (PPI) networks for these causal genes in microglia and astrocytes. We found the "regulation of aspartic-type peptidase activity" pathway being the most enriched among all the causal genes. AD-risk variants associated with candidate causal gene PABPC1 is located near or within enhancers only active in astrocytes. We classified the genes into three drug tiers and identified druggable interactions, with imatinib mesylate emerging as a key candidate. A drug-target gene network was created to explore potential drug targets for AD. Conclusions We systematically prioritized AD candidate causal genes based on cell type-specific molecular evidence. The integrative approach enhances our understanding of molecular mechanisms of AD-related genetic variants and facilitates the interpretation of AD GWAS results.
Collapse
Affiliation(s)
- Shiwei Liu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Min Young Cho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
- Sungkyunkwan University, Seoul, Republic of Korea
| | - Yen-Ning Huang
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Tamina Park
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Soumilee Chaudhuri
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Thea Jacobson Rosewood
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Paula J Bice
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, OH, 43210, USA
| | - David A. Bennett
- Department of Neurological Science, Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 340 West 10th Street, Fairbanks Hall, Suite 6200 Indianapolis, Indiana, 46202, USA
| |
Collapse
|
38
|
Jia Y, Dong H, Li L, Wang F, Juan L, Wang Y, Guo H, Zhao T. xQTLatlas: a comprehensive resource for human cellular-resolution multi-omics genetic regulatory landscape. Nucleic Acids Res 2025; 53:D1270-D1277. [PMID: 39351883 PMCID: PMC11701524 DOI: 10.1093/nar/gkae837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/26/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
Understanding how genetic variants influence molecular phenotypes in different cellular contexts is crucial for elucidating the molecular and cellular mechanisms behind complex traits, which in turn has spurred significant advances in research into molecular quantitative trait locus (xQTL) at the cellular level. With the rapid proliferation of data, there is a critical need for a comprehensive and accessible platform to integrate this information. To meet this need, we developed xQTLatlas (http://www.hitxqtl.org.cn/), a database that provides a multi-omics genetic regulatory landscape at cellular resolution. xQTLatlas compiles xQTL summary statistics from 151 cell types and 339 cell states across 55 human tissues. It organizes these data into 20 xQTL types, based on four distinct discovery strategies, and spans 13 molecular phenotypes. Each entry in xQTLatlas is meticulously annotated with comprehensive metadata, including the origin of the tissue, cell type, cell state and the QTL discovery strategies utilized. Additionally, xQTLatlas features multiscale data exploration tools and a suite of interactive visualizations, facilitating in-depth analysis of cell-level xQTL. xQTLatlas provides a valuable resource for deepening our understanding of the impact of functional variants on molecular phenotypes in different cellular environments, thereby facilitating extensive research efforts.
Collapse
Affiliation(s)
- Yuran Jia
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Hongchao Dong
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Linhao Li
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Fang Wang
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
| | - Liran Juan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yadong Wang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| | - Hongzhe Guo
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| | - Tianyi Zhao
- Faculty of Computing, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Harbin 450000, China
| |
Collapse
|
39
|
Guo X, Feng Y, Ji X, Jia N, Maimaiti A, Lai J, Wang Z, Yang S, Hu S. Shared genetic architecture and bidirectional clinical risks within the psycho-metabolic nexus. EBioMedicine 2025; 111:105530. [PMID: 39731856 PMCID: PMC11743124 DOI: 10.1016/j.ebiom.2024.105530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Increasing evidence suggests a complex interplay between psychiatric disorders and metabolic dysregulations. However, most research has been limited to specific disorder pairs, leaving a significant gap in our understanding of the broader psycho-metabolic nexus. METHODS This study leveraged large-scale cohort data and genome-wide association study (GWAS) summary statistics, covering 8 common psychiatric disorders and 43 metabolic traits. We introduced a comprehensive analytical strategy to identify shared genetic bases sequentially, from key genetic correlation regions to local pleiotropy and pleiotropic genes. Finally, we developed polygenic risk score (PRS) models to translate these findings into clinical applications. FINDINGS We identified significant bidirectional clinical risks between psychiatric disorders and metabolic dysregulations among 310,848 participants from the UK Biobank. Genetic correlation analysis confirmed 104 robust trait pairs, revealing 1088 key genomic regions, including critical hotspots such as chr3: 47588462-50387742. Cross-trait meta-analysis uncovered 388 pleiotropic single nucleotide variants (SNVs) and 126 shared causal variants. Among variants, 45 novel SNVs were associated with psychiatric disorders and 75 novel SNVs were associated with metabolic traits, shedding light on new targets to unravel the mechanism of comorbidity. Notably, RBM6, a gene involved in alternative splicing and cellular stress response regulation, emerged as a key pleiotropic gene. When psychiatric and metabolic genetic information were integrated, PRS models demonstrated enhanced predictive power. INTERPRETATION The study highlights the intertwined genetic and clinical relationships between psychiatric disorders and metabolic dysregulations, emphasising the need for integrated approaches in diagnosis and treatment. FUNDING The National Key Research and Development Program of China (2023YFC2506200, SHH). The National Natural Science Foundation of China (82273741, SY).
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Feng
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne, Carlton South, VIC, Australia
| | - Xiaolong Ji
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ningning Jia
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Aierpati Maimaiti
- Department of Neurosurgery, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Jianbo Lai
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zheng Wang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Sheng Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Nanhu Brain-Computer Interface Institute, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Precision Psychiatry, Hangzhou, 310003, China; Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China; Brain Research Institute of Zhejiang University, Hangzhou, 310058, China; MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058, China; Department of Psychology and Behavioral Sciences, Graduate School, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
40
|
Liu C, Joehanes R, Ma J, Xie J, Yang J, Wang M, Huan T, Hwang SJ, Wen J, Sun Q, Cumhur DY, Heard-Costa NL, Orchard P, Carson AP, Raffield LM, Reiner A, Li Y, O'Connor G, Murabito JM, Munson P, Levy D. Integrating Whole Genome and Transcriptome Sequencing to Characterize the Genetic Architecture of Isoform Variation and its Implications for Health and Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.04.24318434. [PMID: 39677465 PMCID: PMC11643148 DOI: 10.1101/2024.12.04.24318434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
We created a comprehensive whole blood splice variation quantitative trait locus (sQTL) resource by analyzing isoform expression ratio (isoform-to-gene) in Framingham Heart Study (FHS) participants (discovery: n=2,622; validation: n=1,094) with whole genome (WGS) and transcriptome sequencing (RNA-seq) data. External replication was conducted using WGS and RNA-seq from the Jackson Heart Study (JHS, n=1,020). We identified over 3.5 million cis -sQTL-isoform pairs ( p <5e-8), comprising 1,176,624 cis -sQTL variants and 10,883 isoform transcripts from 4,971 sGenes, with significant change in isoform-to-gene ratio due to allelic variation. We validated 61% of these pairs in the FHS validation sample ( p <1e-4). External validation ( p <1e-4) in JHS for the top 10,000 and 100,000 most significant cis -sQTL-isoform pairs was 88% and 69%, respectively, while overall pairs validated at 23%. For 20% of cis -sQTLs in the FHS discovery sample, allelic variation did not significantly correlate with overall gene expression. sQTLs are enriched in splice donor and acceptor sites, as well as in GWAS SNPs, methylation QTLs, and protein QTLs. We detailed several sentinel cis -sQTLs influencing alternative splicing, with potential causal effects on cardiovascular disease risk. Notably, rs12898397 (T>C) affects splicing of ULK3 , lowering levels of the full-length transcript ENST00000440863.7 and increasing levels of the truncated transcript ENST00000569437.5, encoding proteins of different lengths. Mendelian randomization analysis demonstrated that a lower ratio of the full-length isoform is causally associated with lower diastolic blood pressure and reduced lymphocyte percentages. This sQTL resource provides valuable insights into how transcriptomic variation may influence health outcomes.
Collapse
|
41
|
Tian C, Zhang Y, Tong Y, Kock KH, Sim DY, Liu F, Dong J, Jing Z, Wang W, Gao J, Tan LM, Han KY, Tomofuji Y, Nakano M, Buyamin EV, Sonthalia R, Ando Y, Hatano H, Sonehara K, Jin X, Loh M, Chambers J, Hon CC, Choi M, Park JE, Ishigaki K, Okamura T, Fujio K, Okada Y, Park WY, Shin JW, Roca X, Prabhakar S, Liu B. Single-cell RNA sequencing of peripheral blood links cell-type-specific regulation of splicing to autoimmune and inflammatory diseases. Nat Genet 2024; 56:2739-2752. [PMID: 39627432 PMCID: PMC11631754 DOI: 10.1038/s41588-024-02019-8] [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: 12/07/2023] [Accepted: 10/30/2024] [Indexed: 12/12/2024]
Abstract
Alternative splicing contributes to complex traits, but whether this differs in trait-relevant cell types across diverse genetic ancestries is unclear. Here we describe cell-type-specific, sex-biased and ancestry-biased alternative splicing in ~1 M peripheral blood mononuclear cells from 474 healthy donors from the Asian Immune Diversity Atlas. We identify widespread sex-biased and ancestry-biased differential splicing, most of which is cell-type-specific. We identify 11,577 independent cis-splicing quantitative trait loci (sQTLs), 607 trans-sGenes and 107 dynamic sQTLs. Colocalization between cis-eQTLs and trans-sQTLs revealed a cell-type-specific regulatory relationship between HNRNPLL and PTPRC. We observed an enrichment of cis-sQTL effects in autoimmune and inflammatory disease heritability. Specifically, we functionally validated an Asian-specific sQTL disrupting the 5' splice site of TCHP exon 4 that putatively modulates the risk of Graves' disease in East Asian populations. Our work highlights the impact of ancestral diversity on splicing and provides a roadmap to dissect its role in complex diseases at single-cell resolution.
Collapse
Affiliation(s)
- Chi Tian
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yuntian Zhang
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yihan Tong
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Kian Hong Kock
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Donald Yuhui Sim
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Fei Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Jiaqi Dong
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhixuan Jing
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wenjing Wang
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Junbin Gao
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Le Min Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Yoshihiko Tomofuji
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahiro Nakano
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Eliora Violain Buyamin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Radhika Sonthalia
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yoshinari Ando
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Hiroaki Hatano
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Kyuto Sonehara
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Xin Jin
- BGI Research, Shenzhen, China
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
- Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen, China
| | - Marie Loh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - John Chambers
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chung-Chau Hon
- Laboratory for Genome Information Analysis, RIKEN Center for Integrative Medical Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, South Korea
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukinori Okada
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Jay W Shin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
42
|
Lee D, Baek JH, Kim Y, Lee BD, Cho EY, Joo EJ, Ahn YM, Kim SH, Chung YC, Rami FZ, Kim SJ, Kim SW, Myung W, Ha TH, Lee HJ, Oh H, Lee KY, Kim MJ, Kang CY, Jeon S, Jo A, Yu H, Jeong S, Ha K, Kim B, Shim I, Cho C, Huang H, Won HH, Hong KS. Genome-wide association study and polygenic risk score analysis for schizophrenia in a Korean population. Asian J Psychiatr 2024; 102:104203. [PMID: 39293130 DOI: 10.1016/j.ajp.2024.104203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024]
Abstract
Although large-scale genome-wide association studies (GWASs) have revealed the genetic architecture of schizophrenia, these studies have mainly focused on populations of European ancestry. This study aimed to identify common genetic variants associated with schizophrenia in the Korean population and evaluate the performance of polygenic risk scores (PRSs) derived from large-scale GWASs across ancestries. In the Korean psychiatric GWAS project (KPGP), seven academic institutes and their affiliated hospitals across South Korea recruited a cohort of 1670 patients with DSM-IV-defined schizophrenia and 2271 healthy controls. A total of 6690,822 SNPs were tested for association with schizophrenia. We identified one previously unreported genome-wide significant locus rs2423464 (P = 2.83 × 10-11; odds ratio = 1.65; 95 % confidence interval = 1.43-1.91, minor allele frequency = 0.126). This variant was also associated with increased lysosomal-associated membrane protein family member 5 (LAMP5) gene expression. The PRS derived from the meta-analysis results of East Asian and European GWASs explained a larger proportion of the phenotypic variance in the Korean schizophrenia sample than the PRS of an East Asian or European GWAS. (R2 = 0.073 for meta-analysis; 0.028 for East Asian GWAS; 0.037 for European GWAS). GWASs involving diverse ethnic groups will expand our understanding of the genetic architecture of schizophrenia.
Collapse
Affiliation(s)
- Dongbin Lee
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Ji Hyun Baek
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yujin Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Byung Dae Lee
- Pusan National University Research Park, Busan, South Korea
| | - Eun-Young Cho
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Eun-Jeong Joo
- Department of Psychiatry, Euijeongbu Eulji University Hospital, Euijeongbu, South Korea; Department of Neuropsychiatry, Eulji University, School of Medicine, Daejeon, South Korea
| | - Yong Min Ahn
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Se Hyun Kim
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, South Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Fatima Zahra Rami
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, South Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Se Joo Kim
- Department of Psychiatry and Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, South Korea
| | - Woojae Myung
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Tae Hyon Ha
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Heon-Jeong Lee
- Department of Psychiatry, Korea University College of Medicine, Seoul, South Korea
| | - Hayoung Oh
- Pusan National University Research Park, Pusan, South Korea
| | - Kyu Young Lee
- Department of Neuropsychiatry, Eulji University, School of Medicine, Daejeon, South Korea; Department of Psychiatry, Nowon Eulji University Hospital, Seoul, South Korea
| | - Min Ji Kim
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Chae Yeong Kang
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, South Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Sumoa Jeon
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Anna Jo
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, South Korea
| | - Hyeona Yu
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seunghwa Jeong
- Department of Psychiatry, Korea University College of Medicine, Seoul, South Korea
| | - Kyooseob Ha
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Department of Psychiatry, Lions Gate Hospital - Vancouver Coastal Health Authority, British Columbia, Canada
| | - Beomsu Kim
- Department of Precision Medicine, Sungkyunkwan University, School of Medicine, 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
| | - Chamlee Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - 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, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea.
| | - Kyung Sue Hong
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Department of Psychiatry, Lions Gate Hospital - Vancouver Coastal Health Authority, British Columbia, Canada.
| |
Collapse
|
43
|
Grodstein F, Lemos B, Yang J, de Paiva Lopes K, Vialle RA, Seyfried N, Wang Y, Shireby G, Hannon E, Thomas A, Brookes K, Mill J, De Jager PL, Bennett DA. Genetic architecture of epigenetic cortical clock age in brain tissue from older individuals: alterations in CD46 and other loci. Epigenetics 2024; 19:2392050. [PMID: 39169872 PMCID: PMC11346548 DOI: 10.1080/15592294.2024.2392050] [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: 03/07/2024] [Revised: 07/18/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
The cortical epigenetic clock was developed in brain tissue as a biomarker of brain aging. As one way to identify mechanisms underlying aging, we conducted a GWAS of cortical age. We leveraged postmortem cortex tissue and genotyping array data from 694 participants of the Rush Memory and Aging Project and Religious Orders Study (ROSMAP; 11000,000 SNPs), and meta-analysed ROSMAP with 522 participants of Brains for Dementia Research (5,000,000 overlapping SNPs). We confirmed results using eQTL (cortical bulk and single nucleus gene expression), cortical protein levels (ROSMAP), and phenome-wide association studies (clinical/neuropathologic phenotypes, ROSMAP). In the meta-analysis, the strongest association was rs4244620 (p = 1.29 × 10-7), which also exhibited FDR-significant cis-eQTL effects for CD46 in bulk and single nucleus (microglia, astrocyte, oligodendrocyte, neuron) cortical gene expression. Additionally, rs4244620 was nominally associated with lower cognition, faster slopes of cognitive decline, and greater Parkinsonian signs (n ~ 1700 ROSMAP with SNP/phenotypic data; all p ≤ 0.04). In ROSMAP alone, the top SNP was rs4721030 (p = 8.64 × 10-8) annotated to TMEM106B and THSD7A. Further, in ROSMAP (n = 849), TMEM106B and THSD7A protein levels in cortex were related to many phenotypes, including greater AD pathology and lower cognition (all p ≤ 0.0007). Overall, we identified converging evidence of CD46 and possibly TMEM106B/THSD7A for potential roles in cortical epigenetic clock age.
Collapse
Affiliation(s)
- Francine Grodstein
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Bernardo Lemos
- Coit Center for Longevity and Neurotherapeutics, Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Jingyun Yang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Katia de Paiva Lopes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Ricardo A. Vialle
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas Seyfried
- Department of Biochemistry, and Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, USA
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Gemma Shireby
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Eilis Hannon
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Alan Thomas
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Keeley Brookes
- Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
44
|
Wang L, Zhang A, Hu Y, Yang W, Zhong L, Shi J, Wang Z, Tao Q, Liang Q, Yao X. Landscape of multiple tissues' gene expression pattern associated with severe sepsis: Genetic insights from Mendelian randomization and trans-omics analysis. Life Sci 2024; 358:123181. [PMID: 39471899 DOI: 10.1016/j.lfs.2024.123181] [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: 06/25/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Sepsis, a systemic syndrome often culminating in multiple organ failure (MOF), poses a substantial global health threat. However, the gene expression pattern of various tissues associated with severe sepsis remains elusive. METHODS Applying the summary data-based Mendelian randomization (SMR) method, we integrated sepsis genome-wide association study (GWAS) data and expression quantitative trait loci (eQTLs) summaries. This facilitated the investigation of gene causality across 12 tissue types within 26 cohorts linked to adverse sepsis outcomes, including critical care and 28-day mortality. Additionally, trans-omics analyses, including blood transcriptome and single-cell RNA sequencing, were conducted to examine cellular origins and gene functions. The effects of ST7L on sepsis were validated in vivo and in vitro. RESULTS We identified 127 genes associated with severe sepsis across diverse tissues. Cross-tissue analysis highlighted ST7L as a significant pan-tissue risk factor for severe sepsis, displaying significance across 11 tissues for both critical care sepsis (meta OR 1.19, 95 % CI: 1.14-1.25, meta p < 0.0001) and 28-day-death sepsis (meta OR: 1.22, 95 % CI: 1.17-1.27, meta p < 0.0001). Notably, independent blood single-cell RNA sequencing data showed specific expression of ST7L in dendritic cells (DCs). ST7L+ DCs were elevated in non-surviving sepsis patients and exhibited an augmented inflammatory molecular pattern compared to ST7L- DCs. Both transcription and translation level of ST7L in DCs exhibited a dose-dependent pattern with LPS. Knocking down ST7L by siRNA was sufficient to alleviate the inflammation phenotype of DCs, including inhibiting p65/NF-kB pathway and inflammatory factors. CONCLUSION Our findings underscore ST7L as a pan-tissue risk factor for severe sepsis, specifically manifested in DCs and associated with an inflammatory phenotype. These results offer essential insights into the gene expression profiles across multiple tissues in severe sepsis, potentially identifying therapeutic targets for effective sepsis management.
Collapse
Affiliation(s)
- Lei Wang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Aiping Zhang
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China; Critical Care Medicine Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Yehong Hu
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China; Critical Care Medicine Department, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Wanwei Yang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Li Zhong
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Jianfeng Shi
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Zhiguo Wang
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, Jiangsu, China
| | - Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, Jiangsu, China.
| | - Xiaoming Yao
- Department of Clinical Laboratory, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
45
|
Munro D, Ehsan N, Esmaeili-Fard SM, Gusev A, Palmer AA, Mohammadi P. Multimodal analysis of RNA sequencing data powers discovery of complex trait genetics. Nat Commun 2024; 15:10387. [PMID: 39613793 PMCID: PMC11607376 DOI: 10.1038/s41467-024-54840-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024] Open
Abstract
RNA sequencing has the potential to reveal many modalities of transcriptional regulation, such as various splicing phenotypes, but studies on gene regulation are often limited to gene expression due to the complexity of extracting and analyzing multiple RNA phenotypes. Here, we present Pantry, a framework to efficiently generate diverse RNA phenotypes from RNA sequencing data and perform downstream integrative analyses with genetic data. Pantry generates phenotypes from six modalities of transcriptional regulation (gene expression, isoform ratios, splice junction usage, alternative TSS/polyA usage, and RNA stability) and integrates them with genetic data via QTL mapping, TWAS, and colocalization testing. We apply Pantry to Geuvadis and GTEx data, finding that 4768 of the genes with no identified eQTL in Geuvadis have QTL in at least one other transcriptional modality, resulting in a 66% increase in genes over eQTL mapping. We further found that the QTL exhibit modality-specific functional properties that are further reinforced by joint analysis of different RNA modalities. We also show that generalizing TWAS to multiple RNA modalities approximately doubles the discovery of unique gene-trait associations, and enhances identification of regulatory mechanisms underlying GWAS signal in 42% of previously associated gene-trait pairs.
Collapse
Affiliation(s)
- Daniel Munro
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Nava Ehsan
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | | | - Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| | - Abraham A Palmer
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, UC San Diego, La Jolla, CA, USA.
| | - Pejman Mohammadi
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA.
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
46
|
Lou J, Tu M, Xu M, Cao Z, Song W. Plasma pQTL and brain eQTL integration identifies PNKP as a therapeutic target and reveals mechanistic insights into migraine pathophysiology. J Headache Pain 2024; 25:202. [PMID: 39578729 PMCID: PMC11585170 DOI: 10.1186/s10194-024-01922-z] [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: 10/06/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Migraine is a prevalent neurological disorder affecting 14.1% of the global population. Despite advances in genetic research, further investigation is needed to identify therapeutic targets and better understand its mechanisms. In this study, we aimed to identify drug targets and explore the relationships between gene expression, protein levels, and migraine pathophysiology. METHODS We utilized cis-pQTL data from deCODE Genetics, combined with migraine GWAS data from the GERA + UKB cohort as the discovery cohort and the FinnGen R10 cohort as the replication cohort. SMR and MR analyses identified migraine-associated protein loci. Brain eQTL data from GTEx v8 and BrainMeta v2 were used to explore causal relationships between gene expression, protein levels, and migraine risk. Mediation analysis assessed the role of metabolites, and PheWAS evaluated potential side effects. RESULTS Four loci were identified: PNKP, MRVI1, CALCB, and INPP5B. PNKP and MRVI1 showed a high level of evidence and opposing effects at the gene and protein levels. PNKP gene expression in certain brain regions was protective against migraine, while its plasma protein levels were positively associated with migraine risk. MRVI1 showed protective effects at the protein level but had the opposite effect at the gene expression level. Mediation analysis revealed that the glutamate to pyruvate ratio and 3-CMPFP mediated PNKP's effects on migraine. PheWAS indicated associations between PNKP and body composition traits, suggesting drug safety considerations. CONCLUSION PNKP and MRVI1 exhibit dual mechanisms of action at the gene and protein levels, potentially involving distinct mechanistic pathways. Among them, PNKP emerges as a promising drug target for migraine treatment, supported by multi-layered validation.
Collapse
Affiliation(s)
- Jiafei Lou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Miaoqian Tu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijian Cao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Wenwen Song
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
47
|
Byun S, Coryell P, Kramer N, D’Costa S, Thulson E, Shine J, Parkus S, Chubinskaya S, Loeser RF, Diekman BO, Phanstiel DH. Response splicing QTLs in primary human chondrocytes identifies putative osteoarthritis risk genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622754. [PMID: 39605710 PMCID: PMC11601258 DOI: 10.1101/2024.11.11.622754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Osteoarthritis affects millions worldwide, yet effective treatments remain elusive due to poorly understood molecular mechanisms. While genome-wide association studies (GWAS) have identified over 100 OA-associated loci, identifying the genes impacted at each locus remains challenging. Several studies have mapped expression quantitative trait loci (eQTL) in chondrocytes and colocalized them with OA GWAS variants to identify putative OA risk genes; however, the degree to which genetic variants influence OA risk via alternative splicing has not been explored. We investigated the role of alternative splicing in OA pathogenesis using RNA-seq data from 101 human chondrocyte samples treated with PBS (control) or fibronectin fragment (FN-f), an OA trigger. We identified 590 differentially spliced genes between conditions, with FN-f inducing splicing events similar to those in primary OA tissue. We used CRISPR/Cas9 to mimic an SNRNP70 splicing event observed in OA and FN-f-treated chondrocytes and found that it induced an OA-like expression pattern. Integration with genotyping data revealed 7,188 splicing quantitative trait loci (sQTL) affecting 3,056 genes. While many sQTLs were shared, we identified 738 and 343 condition-specific sQTLs for control and FN-f, respectively. We identified 15 RNA binding proteins whose binding sites were enriched at sQTL splice junctions and found that expression of those RNA binding proteins correlated with exon inclusion. Colocalization with OA GWAS identified 6 putative risk genes, including a novel candidate, PBRM1. Our study highlights the significant impact of alternative splicing in OA and provides potential therapeutic targets for future research.
Collapse
Affiliation(s)
- Seyoun Byun
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Philip Coryell
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nicole Kramer
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan D’Costa
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eliza Thulson
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sylvie Parkus
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan Chubinskaya
- Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Dvision of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27695, USA
| | - Douglas H Phanstiel
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
48
|
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
|
49
|
Dou L, Xu Z, Xu J, Su C, Pieper AA, Zhu X, Leverenz JB, Wang F, Cummings J, Cheng F. A network-based systems genetics framework identifies pathobiology and drug repurposing in Parkinson's disease. RESEARCH SQUARE 2024:rs.3.rs-4869009. [PMID: 39483867 PMCID: PMC11527220 DOI: 10.21203/rs.3.rs-4869009/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: 11/03/2024]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder. However, current treatments are directed at symptoms and lack ability to slow or prevent disease progression. Large-scale genome-wide association studies (GWAS) have identified numerous genomic loci associated with PD, which may guide the development of disease-modifying treatments. We presented a systems genetics approach to identify potential risk genes and repurposable drugs for PD. First, we leveraged non-coding GWAS loci effects on multiple human brain-specific quantitative trait loci (xQTLs) under the protein-protein interactome (PPI) network. We then prioritized a set of PD likely risk genes (pdRGs) by integrating five types of molecular xQTLs: expression (eQTLs), protein (pQTLs), splicing (sQTLs), methylation (meQTLs), and histone acetylation (haQTLs). We also integrated network proximity-based drug repurposing and patient electronic health record (EHR) data observations to propose potential drug candidates for PD treatments. We identified 175 pdRGs from QTL-regulated GWAS findings, such as SNCA, CTSB, LRRK2, DGKQ, CD38 and CD44. Multi-omics data validation revealed that the identified pdRGs are likely to be druggable targets, differentially expressed in multiple cell types and impact both the parkin ubiquitin-proteasome and alpha-synuclein (a-syn) pathways. Based on the network proximity-based drug repurposing followed by EHR data validation, we identified usage of simvastatin as being significantly associated with reduced incidence of PD (fall outcome: hazard ratio (HR) = 0.91, 95% confidence interval (CI): 0.87-0.94; for dementia outcome: HR = 0.88, 95% CI: 0.86-0.89), after adjusting for 267 covariates. Our network-based systems genetics framework identifies potential risk genes and repurposable drugs for PD and other neurodegenerative diseases if broadly applied.
Collapse
Affiliation(s)
- Lijun Dou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Zhenxin Xu
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Chang Su
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Andrew A. Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - James B. Leverenz
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, UNLV, Las Vegas, Nevada 89154, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
50
|
Li D, Wang Q, Tian Y, Lyv X, Zhang H, Hong H, Gao H, Li YF, Zhao C, Wang J, Wang R, Yang J, Liu B, Schnable PS, Schnable JC, Li YH, Qiu LJ. TWAS facilitates gene-scale trait genetic dissection through gene expression, structural variations, and alternative splicing in soybean. PLANT COMMUNICATIONS 2024; 5:101010. [PMID: 38918950 PMCID: PMC11573905 DOI: 10.1016/j.xplc.2024.101010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/15/2024] [Accepted: 06/23/2024] [Indexed: 06/27/2024]
Abstract
A genome-wide association study (GWAS) identifies trait-associated loci, but identifying the causal genes can be a bottleneck, due in part to slow decay of linkage disequilibrium (LD). A transcriptome-wide association study (TWAS) addresses this issue by identifying gene expression-phenotype associations or integrating gene expression quantitative trait loci with GWAS results. Here, we used self-pollinated soybean (Glycine max [L.] Merr.) as a model to evaluate the application of TWAS to the genetic dissection of traits in plant species with slow LD decay. We generated RNA sequencing data for a soybean diversity panel and identified the genetic expression regulation of 29 286 soybean genes. Different TWAS solutions were less affected by LD and were robust to the source of expression, identifing known genes related to traits from different tissues and developmental stages. The novel pod-color gene L2 was identified via TWAS and functionally validated by genome editing. By introducing a new exon proportion feature, we significantly improved the detection of expression variations that resulted from structural variations and alternative splicing. As a result, the genes identified through our TWAS approach exhibited a diverse range of causal variations, including SNPs, insertions or deletions, gene fusion, copy number variations, and alternative splicing. Using this approach, we identified genes associated with flowering time, including both previously known genes and novel genes that had not previously been linked to this trait, providing insights complementary to those from GWAS. In summary, this study supports the application of TWAS for candidate gene identification in species with low rates of LD decay.
Collapse
Affiliation(s)
- Delin Li
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Qi Wang
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China; College of Agriculture, Northeast Agricultural University, Harbin 150030, China
| | - Yu Tian
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiangguang Lyv
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Hao Zhang
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Huilong Hong
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Huawei Gao
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya 572024, China
| | - Yan-Fei Li
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Chaosen Zhao
- Crops Research Institute of Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Jiajun Wang
- Soybean Research Institute, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Ruizhen Wang
- Crops Research Institute of Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Jinliang Yang
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Bin Liu
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | | | - James C Schnable
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Ying-Hui Li
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Li-Juan Qiu
- The National Key Facility for Crop Gene Resources and Genetic Improvement (NFCRI)/Key Laboratory of Crop Gene Resource and Germplasm Enhancement (MOA)/State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|