1
|
Cui Y, Chen Y, Hu M, Zhou H, Guo J, Wang Q, Xu Z, Chen L, Zhang W, Tang S. Bidirectional Mendelian randomization and colocalization analysis of gut microbiota on lipid profile. Comput Biol Chem 2025; 117:108422. [PMID: 40080991 DOI: 10.1016/j.compbiolchem.2025.108422] [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: 02/14/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
The gut microbiota plays a crucial role in human health, but its impact on lipid metabolism remains unclear. Understanding the causal relationship between gut bacteria and lipid profiles is essential for developing strategies to prevent and treat dyslipidemia and cardiovascular diseases. This study aimed to assess this relationship using two-sample Mendelian randomization (MR). Data for both exposure and outcomes were obtained from the IEU-GWAS database, with lipid profile data sourced from a publication. Genome-wide significant single nucleotide polymorphisms (SNPs), which were independent of outcome factors but correlated with exposure variables, were identified as instrumental variables. Several MR methods, including weighted analysis, maximum likelihood, inverse variance weighting (IVW), MR-Egger, and weighted median, were applied. Colocalization analysis further validated the findings. The analysis revealed microbial groups with causal relationships to ApoA1, ApoB, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, total cholesterol, and triglycerides. Reverse MR and colocalization analysis provided additional confirmation of these results. This study offers new evidence of the causal link between gut microbiota and lipid profiles, providing insights for improving lipid profiles and reducing cardiovascular disease risk.
Collapse
Affiliation(s)
- Yu Cui
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China
| | - Yanzhu Chen
- Operating Room 1 Area, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Mengting Hu
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China
| | - He Zhou
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Jiarui Guo
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Qijia Wang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Zaihua Xu
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Liyun Chen
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Research Center of Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Wancong Zhang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China.
| | - Shijie Tang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China.
| |
Collapse
|
2
|
Schwartz R, Warwick AN, Khawaja AP, Luben R, Khalid H, Phatak S, Jhingan M, de Vente C, Valmaggia P, Liakopoulos S, Olvera-Barrios A, Sánchez CI, Egan C, Bonelli R, Tufail A. Genetic Distinctions Between Reticular Pseudodrusen and Drusen: A Genome-Wide Association Study. Am J Ophthalmol 2025; 274:286-295. [PMID: 40064387 DOI: 10.1016/j.ajo.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVE To identify genetic determinants specific to reticular pseudodrusen (RPD) compared with drusen. DESIGN Genome-wide association study (GWAS) SUBJECTS: Participants with RPD, drusen, and controls from the UK Biobank (UKBB), a large, multisite, community-based cohort. METHODS Participants with RPD, drusen, and controls from the UK Biobank (UKBB), a large, multisite, community-based cohort, were included. A deep learning framework analyzed 169,370 optical coherence tomography (OCT) volumes to identify cases and controls within the UKBB. Five retina specialists validated the cohorts using OCT and color fundus photographs. Several GWAS were undertaken utilizing the quantity and presence of RPD and drusen. Genome-wide significance was defined as P < 5e-8. MAIN OUTCOMES MEASURES Genetic associations were examined with the number of RPD and drusen within 'pure' cases, where only RPD or drusen were present in either eye. A candidate approach assessed 46 previously known AMD loci. Secondary GWAS were conducted for number of RPD and drusen in mixed cases, and binary case-control analyses for pure RPD and pure drusen. RESULTS The study included 1787 participants: 1037 controls, 361 pure drusen, 66 pure RPD, and 323 mixed cases. The primary pure RPD GWAS identified four genome-wide significant loci: rs11200630 near ARMS2-HTRA1 (P = 1.9e-09), rs79641866 at PARD3B (P = 1.3e-08), rs143184903 near ITPR1 (P = 8.1e-09), and rs76377757 near SLN (P = 4.3e-08). The latter three are uncommon variants (minor allele frequency <5%). A significant association at the CFH locus was also observed using a candidate approach (P = 1.8e-04). For pure drusen, two loci reached genome-wide significance: rs10801555 at CFH (P = 6.0e-33) and rs61871744 at ARMS2-HTRA1 (P = 4.2e-20). CONCLUSIONS The study highlights a clear association between the ARMS2-HTRA1 locus and higher RPD load. Although the CFH locus association did not achieve genome-wide significance, a suggestive link was observed. Three novel associations unique to RPD were identified, albeit for uncommon genetic variants. Further studies with larger sample sizes are needed to explore these findings.
Collapse
Affiliation(s)
- Roy Schwartz
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; Institute of Health Informatics (R.S.), University College London, London, UK.
| | - Alasdair N Warwick
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; University College London Institute of Cardiovascular Science (A.N.W.), London, UK
| | - Anthony P Khawaja
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| | - Robert Luben
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; MRC Epidemiology Unit (R.L.), University of Cambridge, Cambridge, UK
| | - Hagar Khalid
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Sumita Phatak
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Mahima Jhingan
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Coen de Vente
- Quantitative Healthcare Analysis (qurAI) Group (C.D.V., C.I.S.), Informatics Institute, University of Amsterdam, Amsterdam, Netherlands; Biomedical Engineering and Physics (C.D.V., C.I.S.), Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Philippe Valmaggia
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; Department of Biomedical Engineering (P.V.), University of Basel, Basel, Switzerland; Department of Ophthalmology (P.V.), University Hospital Basel, Basel, Switzerland
| | - Sandra Liakopoulos
- Cologne Image Reading Center (S.L.), Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Ophthalmology (S.L.), Goethe University, Frankfurt, Germany
| | - Abraham Olvera-Barrios
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; Institute of Ophthalmology (A.O.B), University College London, London, UK
| | - Clara I Sánchez
- Quantitative Healthcare Analysis (qurAI) Group (C.D.V., C.I.S.), Informatics Institute, University of Amsterdam, Amsterdam, Netherlands; Biomedical Engineering and Physics (C.D.V., C.I.S.), Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Catherine Egan
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| | - Roberto Bonelli
- Lowy Medical Research Institute (R.B.), La Jolla, California, USA
| | - Adnan Tufail
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
3
|
Leyden GM, Sobczyk MK, Richardson TG, Gaunt TR. Distinct pathway-based effects of blood pressure and body mass index on cardiovascular traits: comparison of novel Mendelian randomization approaches. Genome Med 2025; 17:54. [PMID: 40375348 DOI: 10.1186/s13073-025-01472-2] [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: 12/21/2023] [Accepted: 04/11/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Mendelian randomization (MR) leverages trait associated genetic variants as instrumental variables (IVs) to determine causal relationships in epidemiology. However, genetic IVs for complex traits are typically highly heterogeneous and, at a molecular level, exert effects on different biological processes. Exploration of the biological underpinnings of such heterogeneity can enhance our understanding of disease mechanisms and inform therapeutic strategies. Here, we introduce a new approach to instrument partitioning based on enrichment of Mendelian disease categories (pathway-partitioned) and compare it to an existing method based on genetic colocalization in contrasting tissues (tissue-partitioned). METHODS We employed individual- and summary-level MR methodologies using SNPs grouped by pathway informed by proximity to Mendelian disease genes affecting the renal system or vasculature (for blood pressure (BP)), or mental health and metabolic disorders (for body mass index (BMI)). We compared the causal effects of pathway-partitioned SNPs on cardiometabolic outcomes with those derived using tissue-partitioned SNPs informed by colocalization with gene expression in kidney, artery (BP), or adipose and brain tissues (BMI). Additionally, we assessed the likelihood that estimates observed for partitioned exposures could emerge by chance using random SNP sampling. RESULTS Our pathway-partitioned findings suggest the causal relationship between systolic BP and heart disease is predominantly driven by vessel over renal pathways. The stronger effect attributed to kidney over artery tissue in our tissue-partitioned MR hints at a multifaceted interplay between pathways in the disease aetiology. We consistently identified a dominant role for vessel (pathway) and artery (tissue) driving the negative directional effect of diastolic BP on left ventricular stroke volume and positive directional effect of systolic BP on type 2 diabetes. We also found when dissecting the BMI pathway contribution to atrial fibrillation that metabolic-pathway and brain-tissue IVs predominantly drove the causal effects relative to mental health and adipose in pathway- and tissue-partitioned MR analyses, respectively. CONCLUSIONS This study presents a novel approach to dissecting heterogeneity in MR by integrating clinical phenotypes associated with Mendelian disease. Our findings emphasize the importance of understanding pathway-/tissue-specific contributions to complex exposures when interpreting causal relationships in MR. Importantly, we advocate caution and robust validation when interpreting pathway-partitioned effect size differences.
Collapse
Affiliation(s)
- Genevieve M Leyden
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK.
| | - Maria K Sobczyk
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Oakfield House, Bristol, BS8 2BN, UK.
| |
Collapse
|
4
|
Metz S, Belanich JR, Claussnitzer M, Kilpeläinen TO. Variant-to-function approaches for adipose tissue: Insights into cardiometabolic disorders. CELL GENOMICS 2025; 5:100844. [PMID: 40185091 DOI: 10.1016/j.xgen.2025.100844] [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: 10/31/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with cardiometabolic disorders. However, the functional interpretation of these loci remains a daunting challenge. This is particularly true for adipose tissue, a critical organ in systemic metabolism and the pathogenesis of various cardiometabolic diseases. We discuss how variant-to-function (V2F) approaches are used to elucidate the mechanisms by which GWAS loci increase the risk of cardiometabolic disorders by directly influencing adipose tissue. We outline GWAS traits most likely to harbor adipose-related variants and summarize tools to pinpoint the putative causal variants, genes, and cell types for the associated loci. We explain how large-scale perturbation experiments, coupled with imaging and multi-omics, can be used to screen variants' effects on cellular phenotypes and how these phenotypes can be tied to physiological mechanisms. Lastly, we discuss the challenges and opportunities that lie ahead for V2F research and propose a roadmap for future studies.
Collapse
Affiliation(s)
- Sophia Metz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jonathan Robert Belanich
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melina Claussnitzer
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Genomic Medicine, Endocrine Division, Massachusetts General Hospital, Harvard Medical School, Cambridge, MA 02142, USA
| | - Tuomas Oskari Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
5
|
Strom NI, Gerring ZF, Galimberti M, Yu D, Halvorsen MW, Abdellaoui A, Rodriguez-Fontenla C, Sealock JM, Bigdeli T, Coleman JR, Mahjani B, Thorp JG, Bey K, Burton CL, Luykx JJ, Zai G, Alemany S, Andre C, Askland KD, Bäckman J, Banaj N, Barlassina C, Nissen JB, Bienvenu OJ, Black D, Bloch MH, Børte S, Bosch R, Breen M, Brennan BP, Brentani H, Buxbaum JD, Bybjerg-Grauholm J, Byrne EM, Cabana-Dominguez J, Camarena B, Camarena A, Cappi C, Carracedo A, Casas M, Cavallini MC, Ciullo V, Cook EH, Crosby J, Cullen BA, De Schipper EJ, Delorme R, Djurovic S, Elias JA, Estivill X, Falkenstein MJ, Fundin BT, Garner L, Gironda C, Goes FS, Grados MA, Grove J, Guo W, Haavik J, Hagen K, Harrington K, Havdahl A, Höffler KD, Hounie AG, Hucks D, Hultman C, Janecka M, Jenike E, Karlsson EK, Kelley K, Klawohn J, Krasnow JE, Krebs K, Lange C, Lanzagorta N, Levey D, Lindblad-Toh K, Macciardi F, Maher B, Mathes B, McArthur E, McGregor N, McLaughlin NC, Meier S, Miguel EC, Mulhern M, Nestadt PS, Nurmi EL, O'Connell KS, Osiecki L, Ousdal OT, Palviainen T, Pedersen NL, Piras F, Piras F, Potluri S, Rabionet R, Ramirez A, Rauch S, Reichenberg A, et alStrom NI, Gerring ZF, Galimberti M, Yu D, Halvorsen MW, Abdellaoui A, Rodriguez-Fontenla C, Sealock JM, Bigdeli T, Coleman JR, Mahjani B, Thorp JG, Bey K, Burton CL, Luykx JJ, Zai G, Alemany S, Andre C, Askland KD, Bäckman J, Banaj N, Barlassina C, Nissen JB, Bienvenu OJ, Black D, Bloch MH, Børte S, Bosch R, Breen M, Brennan BP, Brentani H, Buxbaum JD, Bybjerg-Grauholm J, Byrne EM, Cabana-Dominguez J, Camarena B, Camarena A, Cappi C, Carracedo A, Casas M, Cavallini MC, Ciullo V, Cook EH, Crosby J, Cullen BA, De Schipper EJ, Delorme R, Djurovic S, Elias JA, Estivill X, Falkenstein MJ, Fundin BT, Garner L, Gironda C, Goes FS, Grados MA, Grove J, Guo W, Haavik J, Hagen K, Harrington K, Havdahl A, Höffler KD, Hounie AG, Hucks D, Hultman C, Janecka M, Jenike E, Karlsson EK, Kelley K, Klawohn J, Krasnow JE, Krebs K, Lange C, Lanzagorta N, Levey D, Lindblad-Toh K, Macciardi F, Maher B, Mathes B, McArthur E, McGregor N, McLaughlin NC, Meier S, Miguel EC, Mulhern M, Nestadt PS, Nurmi EL, O'Connell KS, Osiecki L, Ousdal OT, Palviainen T, Pedersen NL, Piras F, Piras F, Potluri S, Rabionet R, Ramirez A, Rauch S, Reichenberg A, Riddle MA, Ripke S, Rosário MC, Sampaio AS, Schiele MA, Skogholt AH, Sloofman LG, Smit J, Artigas MS, Thomas LF, Tifft E, Vallada H, van Kirk N, Veenstra-VanderWeele J, Vulink NN, Walker CP, Wang Y, Wendland JR, Winsvold BS, Yao Y, Zhou H, Agrawal A, Alonso P, Berberich G, Bucholz KK, Bulik CM, Cath D, Denys D, Eapen V, Edenberg H, Falkai P, Fernandez TV, Fyer AJ, Gaziano JM, Geller DA, Grabe HJ, Greenberg BD, Hanna GL, Hickie IB, Hougaard DM, Kathmann N, Kennedy J, Lai D, Landén M, Hellard SL, Leboyer M, Lochner C, McCracken JT, Medland SE, Mortensen PB, Neale BM, Nicolini H, Nordentoft M, Pato M, Pato C, Pauls DL, Piacentini J, Pittenger C, Posthuma D, Ramos-Quiroga JA, Rasmussen SA, Richter MA, Rosenberg DR, Ruhrmann S, Samuels JF, Sandin S, Sandor P, Spalletta G, Stein DJ, Stewart SE, Storch EA, Stranger BE, Turiel M, Werge T, Andreassen OA, Børglum AD, Walitza S, Hveem K, Hansen BK, Rück C, Martin NG, Milani L, Mors O, Reichborn-Kjennerud T, Ribasés M, Kvale G, Mataix-Cols D, Domschke K, Grünblatt E, Wagner M, Zwart JA, Breen G, Nestadt G, Kaprio J, Arnold PD, Grice DE, Knowles JA, Ask H, Verweij KJ, Davis LK, Smit DJ, Crowley JJ, Scharf JM, Stein MB, Gelernter J, Mathews CA, Derks EM, Mattheisen M. Genome-wide analyses identify 30 loci associated with obsessive-compulsive disorder. Nat Genet 2025:10.1038/s41588-025-02189-z. [PMID: 40360802 DOI: 10.1038/s41588-025-02189-z] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025]
Abstract
Obsessive-compulsive disorder (OCD) affects ~1% of children and adults and is partly caused by genetic factors. We conducted a genome-wide association study (GWAS) meta-analysis combining 53,660 OCD cases and 2,044,417 controls and identified 30 independent genome-wide significant loci. Gene-based approaches identified 249 potential effector genes for OCD, with 25 of these classified as the most likely causal candidates, including WDR6, DALRD3 and CTNND1 and multiple genes in the major histocompatibility complex (MHC) region. We estimated that ~11,500 genetic variants explained 90% of OCD genetic heritability. OCD genetic risk was associated with excitatory neurons in the hippocampus and the cortex, along with D1 and D2 type dopamine receptor-containing medium spiny neurons. OCD genetic risk was shared with 65 of 112 additional phenotypes, including all the psychiatric disorders we examined. In particular, OCD shared genetic risk with anxiety, depression, anorexia nervosa and Tourette syndrome and was negatively associated with inflammatory bowel diseases, educational attainment and body mass index.
Collapse
Affiliation(s)
- Nora I Strom
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany.
- Department of Psychiatric Phenomics and Genomics (IPPG), Ludwig-Maximilians University Munich, Munich, Germany.
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Services, Region Stockholm, Stockholm, Sweden.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland.
| | - Zachary F Gerring
- Department of Mental Health and Neuroscience, Translational Neurogenomics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Department of Population Health and Immunity, Healthy Development and Ageing, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Marco Galimberti
- Department of Psychiatry, Human Genetics, Yale University, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Dongmei Yu
- Department of Center for Genomic Medicine, Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthew W Halvorsen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Abdel Abdellaoui
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Cristina Rodriguez-Fontenla
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Genomics and Bioinformatics, University of Santiago de Compostela, Santiago de Compostela, Spain
- Grupo de Medicina Xenómica, Genetics, Instituto de Investigación Sanitaria de Santiago de Compostela (FIDIS), Santiago de Compostela, Spain
| | - Julia M Sealock
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Tim Bigdeli
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- VA NY Harbor Healthcare System, Brooklyn, NY, USA
| | - Jonathan R Coleman
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- National Institute for Health and Care Research Maudsley Biomedical Research Centre, South London and Maudsley NHS Trust, London, UK
| | - Behrang Mahjani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jackson G Thorp
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Katharina Bey
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Christie L Burton
- Department of Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jurjen J Luykx
- Department of Psychiatry, Brain University Medical Center Utrecht, Utrecht, the Netherlands
- Second Opinion Outpatient Clinic, GGNet, Warnsveld, the Netherlands
| | - Gwyneth Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Silvia Alemany
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Christine Andre
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Kathleen D Askland
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Hamilton, Ontario, Canada
| | - Julia Bäckman
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Services, Region Stockholm, Stockholm, Sweden
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | - Judith Becker Nissen
- Department of Child and Adolescent Psychiatry, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
- Institute of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - O Joseph Bienvenu
- Department of Psychiatry and Behavioral Sciences, General Hospital Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donald Black
- Departments of Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Michael H Bloch
- Department of Child Study Center and Psychiatry, Yale University, New Haven, CT, USA
| | - Sigrid Børte
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, HUNT Center for Molecular and Clinical Epidemiology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Rosa Bosch
- Department of Child and Adolescent Mental Health, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
- Instituto de Salut Carlos III, Centro de Investigación Biomédica en Red de Salut Mental (CIBERSAM), Madrid, Spain
| | - Michael Breen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian P Brennan
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Helena Brentani
- Department of Psychiatry, Universidade de São Paulo, São Paulo, Brazil
| | - Joseph D Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Enda M Byrne
- Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Judit Cabana-Dominguez
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Beatriz Camarena
- Pharmacogenetics Department, Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramon de la Fuente Muñiz, Mexico City, México
| | | | - Carolina Cappi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
- Department of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Angel Carracedo
- CiMUS, Genomics and Bioinformatics Group, University of Santiago de Compostela, Santiago de Compostela, Spain
- Galician Foundation of Genomic Medicine, Grupo de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Medicina Genómica, Centro de Investigación Biomédica en Red, Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | - Miguel Casas
- Programa MIND Escoles, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
- Departamento de Psiquiatría y Medicina Legal, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | - Valentina Ciullo
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Edwin H Cook
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL, USA
| | - Jesse Crosby
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Bernadette A Cullen
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Mental Health, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elles J De Schipper
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Services, Region Stockholm, Stockholm, Sweden
| | - Richard Delorme
- Child and Adolesccent Psychiatry Department, APHP, Paris, France
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jason A Elias
- Psychiatry, McLean Hospital OCDI, Harvard Medical School, Belmont, MA, USA
- Adult Psychological Services, CBTeam LLC, Lexington, MA, USA
| | - Xavier Estivill
- qGenomics (Quantitative Genomics Laboratories), Esplugues de Llobregat, Spain
| | - Martha J Falkenstein
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Bengt T Fundin
- Department of Medical Epidemiology and Biostatistics, Center for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden
| | - Lauryn Garner
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Christina Gironda
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Fernando S Goes
- Department of Psychiatry, Johns Hopkins University, Baltimore, MD, USA
| | - Marco A Grados
- Department of Psychiatry and Behavioral Sciences, Child and Adolescent Psychiatry, Johns Hopkins University, Baltimore, MD, USA
| | - 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, Denmark
| | - Wei Guo
- Genetic Epidemiology Research Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Kristen Hagen
- Department of Psychiatry, Møre og Romsdal Hospital Trust, Molde, Norway
- Bergen Center for Brain Plasticity, Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Mental Health, Norwegian University for Science and Technology, Trondheim, Norway
| | - Kelly Harrington
- Million Veteran Program (MVP) Coordinating Center, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Alexandra Havdahl
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Nic Waals Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Kira D Höffler
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway
- Department of Medical Genetics, Dr. Einar Martens Research Group for Biological Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Ana G Hounie
- Department of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Donald Hucks
- Department of Medicine, Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christina Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Magdalena Janecka
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Eric Jenike
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Elinor K Karlsson
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kara Kelley
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Julia Klawohn
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Medicine, MSB Medical School Berlin, Berlin, Germany
| | - Janice E Krasnow
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Christoph Lange
- Department of Biostatistics, T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Daniel Levey
- Department of Psychiatry, Yale University, West Haven, CT, USA
- Office of Research and Development, United States Department of Veterans Affairs, West Haven, CT, USA
| | - Kerstin Lindblad-Toh
- Department of Vertebrate Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Fabio Macciardi
- Department of Psychiatry, University of California, Irvine, Irvine, CA, USA
| | - Brion Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Brittany Mathes
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Evonne McArthur
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Nicole C McLaughlin
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
- Butler Hospital, Providence, RI, USA
| | - Sandra Meier
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Euripedes C Miguel
- Department of Psychiatry, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Maureen Mulhern
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Paul S Nestadt
- Department of Psychiatry and Behavioral Science, Johns Hopkins University, Baltimore, MD, USA
| | - Erika L Nurmi
- Department of Psychiatry and Biobehavioral Sciences, Division of Child and Adolescent Psychiatry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kevin S O'Connell
- Department of Clinical Medicine, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- NORMENT, University of Oslo, Oslo, Norway
| | - Lisa Osiecki
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Harvard Medical School, Boston, MA, USA
| | - Olga Therese Ousdal
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Biomedicine, Haukeland University Hospital, Bergen, Norway
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Federica Piras
- Department of Clinical Neuroscience and Neurorehabilitation, Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Sriramya Potluri
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Raquel Rabionet
- Department of Genetics, Microbiology and Statistics, IBUB, Universitat de Barcelona, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red, Madrid, Spain
- Department of Human Molecular Genetics, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, Division of Neurogenetics and Molecular Psychiatry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, Medical Faculty, University Hospital Bonn, Bonn, Germany
- DZNE Bonn, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Cologne Excellence Cluster for Stress Responses in Ageing-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Scott Rauch
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Abraham Reichenberg
- Department of Mental Disorders, Norwegian Institute of Public Health, New York, NY, USA
| | - Mark A Riddle
- Department of Psychiatry and Behavioral Sciences, Child and Adolescent, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Site Berlin-Potsdam, German Center for Mental Health (DZPG), Berlin, Germany
| | - Maria C Rosário
- Department of Psychiatry, Child and Adolescent Psychiatry Unit (UPIA), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Aline S Sampaio
- Department of Neurosciences and Mental Health, Medical School, Federal University of Bahia, Salvador, Brazil
| | - Miriam A Schiele
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Freiburg, Medical Center-University of Freiburg, Freiburg, Germany
| | - Anne Heidi Skogholt
- Department of Public Health and Nursing, HUNT Center for Molecular and Clinical Epidemiology, Trondheim, Norway
| | - Laura G Sloofman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jan Smit
- Department of Psychiatry, Faculty of Medicine, Locaion VUmc, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - María Soler Artigas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Laurent F Thomas
- Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU, Trondheim, Norway
- BioCore, Bioinformatics Core Facility, NTNU, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eric Tifft
- Obsessive-Compulsive Disorder Institute, McLean Hospital, Belmont, MA, USA
| | - Homero Vallada
- Department of Psychiatry, Universidade de São Paulo, São Paulo, Brazil
- Department of Molecular Medicine and Surgery, CMM, Karolinska Institutet, Stockholm, Sweden
| | - Nathanial van Kirk
- OCD Institute, Division of Depression and Anxiety, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Division of Child and Adolescent Psychiatry, Columbia University, New York, NY, USA
- Department of Child and Adolescent Psychiatry, New York State Psychiatric Institute, New York, NY, USA
| | - Nienke N Vulink
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Ying Wang
- Department of Neurology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jens R Wendland
- Laboratory of Clinical Science, NIMH Intramural Research Program, Bethesda, MD, USA
| | - Bendik S Winsvold
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
| | - Yin Yao
- Department of Computional Biology, Institute of Life Science, Fudan University, Fudan, China
| | - Hang Zhou
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Pino Alonso
- Department of Psychiatry, OCD Clinical and Research Unit, Bellvitge Hospital, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
- Department of Psychiatry and Mental Health, Bellvitge Biomedical Research Institute IDIBELLL, Barcelona, Spain
- CIBERSAM, Mental Health Network Biomedical Research Center, Madrid, Spain
| | - Götz Berberich
- Psychosomatic Department, Windach Hospital of Neurobehavioural Research and Therapy, Windach, Germany
| | - Kathleen K Bucholz
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Danielle Cath
- Departments of Rijksuniversiteit Groningen and Psychiatry, University Medical Center Groningen, Groningen, the Netherlands
- Department of Specialized Training, Drenthe Mental Health Care Institute, Groningen, the Netherlands
| | - Damiaan Denys
- Department of Psychiatry, Institute of the Royal Netherlands Academy of Arts and Sciences (NIN-KNAW), Amsterdam, the Netherlands
| | - Valsamma Eapen
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, UNSW, Sydney, New South Wales, Australia
- Academic Unit of Child Psychiatry South-West Sydney, South-West Sydney Clinical School, SWSLHD and Ingham Institute, Sydney, New South Wales, Australia
| | - Howard Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital LMU, Munich, Germany
- Department of Psychiatry, Max Planck Institute, Munich, Germany
| | - Thomas V Fernandez
- Child Study Center and Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Abby J Fyer
- Department of Psychiatry, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - J M Gaziano
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Mass General Brigham, Boston, MA, USA
| | - Dan A Geller
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Child Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Benjamin D Greenberg
- COBRE Center on Neuromodulation, Butler Hospital, Providence, RI, USA
- Center for Neurorestoration and Neurotechnology, VA Providence Healthcare System, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
| | - Gregory L Hanna
- Department of Psychiatry, Child and Adolescent Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Ian B Hickie
- Brain and Mind Centre, the University of Sydney, Sydney, New South Wales, Australia
| | - David M Hougaard
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Norbert Kathmann
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - James Kennedy
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Stéphanie Le Hellard
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Marion Leboyer
- Department of Addictology and Psychiatry, Université Paris-Est Créteil, AP-HP, Inserm, Paris, France
| | - Christine Lochner
- Department of Psychiatry, SA MRC Unit on Risk and Resilience in Mental Disorders, Stellenbosch University, Stellenbosch, South Africa
| | - James T McCracken
- Department of Psychiatry and Biobehavioral Sciences, Division of Child and Adolescent Psychiatry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sarah E Medland
- Department of Mental Health, Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Preben B Mortensen
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Benjamin M Neale
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Humberto Nicolini
- Department of Psychiatry, Psychiatry, Carracci Medical Group, Mexico City, México
- Psiquiatría, Instituto Nacional de Medicina Genómica, Mexico City, México
| | - Merete Nordentoft
- Mental Health Center Copenhagen, Copenhagen Research Center for Mental Health, Mental Health Services in the Capital Region of Denmark, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michele Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ, USA
| | - Carlos Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ, USA
| | - David L Pauls
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - John Piacentini
- Department of Psychiatry and Biobehavioral Sciences, Child and Adolescent Psychiatry, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | | | - Danielle Posthuma
- Department of Complex Trait Genetics, Vrije Universiteit Amsterdam, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Child and Adolescent Psychiatric, Section Complex Trait Genetics, VU Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Josep Antoni Ramos-Quiroga
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Group of Psychiatry, Mental Health and Addictions, Psychiatric Genetics Unit, Vall d'Hebron Research Institute, Barcelona, Spain
- CIBERSAM, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Steven A Rasmussen
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University, Providence, RI, USA
| | - Margaret A Richter
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - David R Rosenberg
- Department of Psychiatry and Behavioral Neurosciences, Child and Adolescent Psychiatry, Wayne State University School of Medicine, Detroit, MI, USA
| | - Stephan Ruhrmann
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Jack F Samuels
- Department of Psychiatry and Behavioral Sciences, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sven Sandin
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Paul Sandor
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychiatry and Behavioral Sciences, Division of Neuropsychiatry, Baylor College of Medicine, Houston, TX, USA
| | - Dan J Stein
- Department of Psychiatry and Neuroscience Institute, SAMRC Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| | - S Evelyn Stewart
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada
| | - Eric A Storch
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Barbara E Stranger
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Copenhagen University Hospital, Mental Health Services (RHP), Copenhagen, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole A Andreassen
- Institute of Clinical Medicine, NORMENT Centre, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Center for Precision Psychiatry, Oslo University Hospital, Oslo, Norway
| | - Anders D Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus University, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus University, Aarhus, Denmark
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
- HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
- Department of Research, Innovation and Education, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Bjarne K Hansen
- Bergen Center for Brain Plasticity, Psychiatry, Haukeland University Hospital, Bergen, Norway
- Centre for Crisis Psychology, Psychology, University of Bergen, Bergen, Norway
| | - Christian Rück
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Services, Region Stockholm, Stockholm, Sweden
| | - Nicholas G Martin
- Department of Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Ole Mors
- Psychosis Research Unit, Psychiatry, Aarhus University Hospital, Aarhus, Denmark
| | - Ted Reichborn-Kjennerud
- Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marta Ribasés
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Gerd Kvale
- Department of Mental Health, Norwegian University for Science and Technology, Trondheim, Norway
- Department of Clinical Psychology, Faculty of Psychology, University of Bergen, Bergen, Norway
| | - David Mataix-Cols
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Services, Region Stockholm, Stockholm, Sweden
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Freiburg, Medical Center-University of Freiburg, Freiburg, Germany
- Partner Site Berlin, DZPG, Berlin, Germany
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Michael Wagner
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- DZNE, Bonn, Germany
| | - John-Anker Zwart
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Trondheim, Norway
- Department of Research and Innovation, Clinical Neuroscience, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatric Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Gerald Nestadt
- Department of Psychiatry and Behavioral Science, Johns Hopkins University, Baltimore, MD, USA
| | | | - Paul D Arnold
- Department of Psychiatry, the Mathison Centre for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Dorothy E Grice
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James A Knowles
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Helga Ask
- PsychGen Center for Genetic Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway
| | - Karin J Verweij
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Lea K Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dirk J Smit
- Department of Psychiatry, Amsterdam UMC location AMC, Amsterdam, the Netherlands
| | - James J Crowley
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Services, Region Stockholm, 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
| | - Jeremiah M Scharf
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Murray B Stein
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry and School of Public Health, University of California San Diego, La Jolla, CA, USA
| | - Joel Gelernter
- Department of Psychiatry, Human Genetics (Psychiatry), Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Carol A Mathews
- Psychiatry and Genetics Institute, Evelyn F. and William L. Mc Knight Brain Institute, Center for OCD, Anxiety and Related Disorders, University of Florida, Gainesville, FL, USA
| | - Eske M Derks
- Department of Mental Health and Neuroscience, QIMR Berghofer, Brisbane, Queensland, Australia
| | - Manuel Mattheisen
- Department of Psychiatric Phenomics and Genomics (IPPG), Ludwig-Maximilians University Munich, Munich, Germany.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Department of Community Health and Epidemiology and Faculty of Computer Science, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
6
|
Byun J, Han Y, Choi J, Sun R, Shaw VR, Zhu C, Xiao X, Lusk C, Badr H, Lee HS, Jang HJ, Li Y, Lim H, Long E, Liu Y, Kachuri L, Walsh KM, Wiencke JK, Albanes D, Lam S, Tardon A, Neuhouser ML, Barnett MJ, Chen C, Bojesen S, Brenner H, Landi MT, Johansson M, Risch A, Wichmann HE, Bickeböller H, Christiani DC, Rennert G, Arnold S, Field JK, Shete S, Le Marchand L, Liu G, Andrew AS, Zienolddiny S, Grankvist K, Johansson M, Caporaso N, Taylor F, Lazarus P, Schabath MB, Aldrich MC, Patel A, Lin X, Zanetti KA, Harris CC, Chanock S, McKay J, Schwartz AG, Hung RJ, Amos CI. Genome-wide association study for lung cancer in 6531 African Americans reveals new susceptibility loci. Hum Mol Genet 2025:ddaf059. [PMID: 40341939 DOI: 10.1093/hmg/ddaf059] [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: 08/08/2024] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Despite lung cancer affecting all races and ethnicities, disparities are observed in incidence and mortality rates among different ethnic groups in the United States. Non-Hispanic African Americans had a high incidence rate of lung cancer at 55.8 per 100 000 people, as well as the highest death rate at 37.2 per 100 000 people from 2016 to 2020. While previous genome-wide association studies (GWAS) have identified over 45 susceptibility risk loci that influence lung cancer development, few GWAS have investigated the etiology of lung cancer in African Americans. To address this gap in knowledge, we conducted GWAS of lung cancer focused on studying African Americans, comprising 2267 lung cancer cases and 4264 controls. We identified three loci associated with lung cancer, one with lung adenocarcinoma, and four with lung squamous cell carcinoma in this population at the genomic-wide significance level. Among them, three novel loci were identified near VWF at 12p13.31 for overall lung cancer and GACAT3 at 2p24.3 and LMAN1L at 15q24.1 for lung squamous cell carcinoma. In addition, we confirmed previously reported risk loci with known or new lead variants near CHRNA5 at 15q25.1 and CYP2A6 at 19q13.2 associated with lung cancer and TRIP13 at 5p15.33 and ERC1 at 12p13.33 associated with lung squamous cell carcinoma. Further multi-step functional analyses shed light on biological mechanisms underlying these associations of lung cancer in this population. Our study highlights the importance of ancestry-specific studies for the potential alleviation of lung cancer burden in African Americans.
Collapse
Affiliation(s)
- Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Ryan Sun
- Department of Biostatistics, University of Texas, M.D. Anderson Cancer Center, 7007 Bertner Ave, Houston, TX, 77030, United States
| | - Vikram R Shaw
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Catherine Zhu
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Xiangjun Xiao
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Christine Lusk
- Department of Oncology, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, United States
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, United States
| | - Hoda Badr
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Hyun-Sung Lee
- Systems Onco-Immunology Lab, David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Hee-Jin Jang
- Systems Onco-Immunology Lab, David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Yafang Li
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Hyeyeun Lim
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Erping Long
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yanhong Liu
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University, 300 Pasteur Drive, Stanford, CA, 94305, United States
| | - Kyle M Walsh
- Duke Cancer Institute, Duke University Medical Center, 20 Duke Medicine Cir, Durham, NC, 27701, United States
| | - John K Wiencke
- Department of Neurological Surgery, The University of California, San Francisco, 400 Parnassus Ave, San Francisco, CA, 94143, United States
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Stephen Lam
- Department of Integrative Oncology, University of British Columbia, 675 West 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Adonina Tardon
- Public Health Department, University of Oviedo, and Health Research Institute of Asturias, ISPA, Av. del Hospital Universitario, s/n, 33011 Oviedo, Asturias, Spain
| | - Marian L Neuhouser
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Matt J Barnett
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Chu Chen
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Stig Bojesen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Mattias Johansson
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Angela Risch
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Im Neuenheimer Feld 156, 69120, Heidelberg, Germany
- Division of Cancer Epigenomics, DKFZ-German Cancer Research Center, Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, University of Salzburg and Cancer Cluster Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - H-Erich Wichmann
- Helmholtz-Munich Institute of Epidemiology, Ingolstädter Landstraße 1, Neuherberg, 85764, Germany
| | - Heike Bickeböller
- University Medical Center Göttingen, Institute of Genetic Epidemiology, Humboldtallee 32, 37073 Göttingen, Germany
| | - David C Christiani
- Department of Environmental Health and Epidemiology, Harvard T.H.Chan School of Public Health, 665 Huntington Avenue, Building 1, Boston, MA, 02115, United States
| | - Gad Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Mikhal St 7, Haifa, 3436212, Israel
| | - Susanne Arnold
- University of Kentucky, Markey Cancer Center, 800 Rose Street, Lexington, KY, 40536, United States
| | - John K Field
- Institute of Translational Medicine, University of Liverpool, the Sherrington Building, Ashton St, Liverpool, L69 3GE, United Kingdom
| | - Sanjay Shete
- Department of Biostatistics, University of Texas, M.D. Anderson Cancer Center, 7007 Bertner Ave, Houston, TX, 77030, United States
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, United States
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, United States
| | - Geoffrey Liu
- University Health Network- The Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
| | - Angeline S Andrew
- Departments of Epidemiology and Community and Family Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, United States
| | | | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, 901 87 Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, 901 87 Umeå, Sweden
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Fiona Taylor
- Sheffield Teaching Hospitals Foundation Trust, 8 Beech Hill Road, Sheffield, S10 2SB, United Kingdom
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, 412 East Spokane Falls Blvd, PBS 130, Spokane, WA, 99202, United States
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, United States
| | - Melinda C Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, 1161 21st Ave S, Nashville, TN, 37232, United States
| | - Alpa Patel
- American Cancer Society, Inc., 270 Peachtree Street NW, Atlanta, GA, 30303, United States
| | - Xihong Lin
- Department of Biostatistics, Harvard TH Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, United States
| | - Krista A Zanetti
- Office of Nutrition Research, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, 6705 Rockledge Drive, Bethesda, MD, 20817, United States
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, 37 Convent Dr, Bethesda, MD, 20892, United States
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - James McKay
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Ann G Schwartz
- Department of Oncology, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, United States
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, United States
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, 155 College Street, Toronto, Ontario, M5T 3M7, Canada
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| |
Collapse
|
7
|
Gui A, Hollowell A, Wigdor EM, Morgan MJ, Hannigan LJ, Corfield EC, Odintsova V, Hottenga JJ, Wong A, Pool R, Cullen H, Wilson S, Warrier V, Eilertsen EM, Andreassen OA, Middeldorp CM, St Pourcain B, Bartels M, Boomsma DI, Hartman CA, Robinson EB, Arichi T, Edwards AD, Johnson MH, Dudbridge F, Sanders SJ, Havdahl A, Ronald A. Genome-wide association meta-analysis of age at onset of walking in over 70,000 infants of European ancestry. Nat Hum Behav 2025:10.1038/s41562-025-02145-1. [PMID: 40335706 DOI: 10.1038/s41562-025-02145-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/21/2025] [Indexed: 05/09/2025]
Abstract
Age at onset of walking is an important early childhood milestone which is used clinically and in public health screening. In this genome-wide association study meta-analysis of age at onset of walking (N = 70,560 European-ancestry infants), we identified 11 independent genome-wide significant loci. SNP-based heritability was 24.13% (95% confidence intervals = 21.86-26.40) with ~11,900 variants accounting for about 90% of it, suggesting high polygenicity. One of these loci, in gene RBL2, co-localized with an expression quantitative trait locus (eQTL) in the brain. Age at onset of walking (in months) was negatively genetically correlated with ADHD and body-mass index, and positively genetically correlated with brain gyrification in both infant and adult brains. The polygenic score showed out-of-sample prediction of 3-5.6%, confirmed as largely due to direct effects in sib-pair analyses, and was separately associated with volume of neonatal brain structures involved in motor control. This study offers biological insights into a key behavioural marker of neurodevelopment.
Collapse
Affiliation(s)
- Anna Gui
- Department of Psychology, University of Essex, Wivenhoe Park, Colchester, UK
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK
| | - Anja Hollowell
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK
| | - Emilie M Wigdor
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Morgan J Morgan
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Laurie J Hannigan
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Elizabeth C Corfield
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Veronika Odintsova
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Psychiatry, University Medical Center of Groningen, University of Groningen, Groningen, the Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - René Pool
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Harriet Cullen
- Research Department of Early Life Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Siân Wilson
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, USA
- Division of Newborn Medicine, Harvard Medical School, Boston, MA, USA
| | - Varun Warrier
- Department of Psychiatry and Psychology, University of Cambridge, Cambridge, UK
| | | | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Christel M Middeldorp
- Department of Child and Youth Psychiatry and Psychology, Amsterdam Reproduction and Development Research Institute, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Arkin Mental Health Care, Amsterdam, the Netherlands
- Levvel, Academic Center for Child and Adolescent Psychiatry, Amsterdam, the Netherlands
- Child Health Research Centre, University of Queensland, Brisbane, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, Brisbane, Australia
| | - Beate St Pourcain
- Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Meike Bartels
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands
| | - Catharina A Hartman
- University Medical Center Psychopathology and Emotion Regulation (ICPE), Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Tomoki Arichi
- Research Department of Early Life Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Anthony D Edwards
- Research Department of Early Life Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Mark H Johnson
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Frank Dudbridge
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Stephan J Sanders
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Alexandra Havdahl
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- PROMENTA Research Center, University of Oslo, Oslo, Norway
| | - Angelica Ronald
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK.
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK.
| |
Collapse
|
8
|
Tong Y, Chen YJ, Cui GB. The genetic overlap and causal relationship between attention deficit hyperactivity disorder and obstructive sleep apnea: a large-scale genomewide cross-trait analysis. BMC Psychiatry 2025; 25:454. [PMID: 40329273 PMCID: PMC12057209 DOI: 10.1186/s12888-025-06899-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Attention deficit hyperactivity disorder (ADHD) and Obstructive sleep apnea (OSA) are highly clinically co-occurring, but the mechanisms behind this remain unclear, so this article analyzes the reasons for the co-morbidities from a genetic perspective. METHODS We examined the genetic architecture of ADHD and OSA based on the large genome-wide association studies (GWAS). The global genetic relationship between OSA and ADHD was explored. Cross-trait analysis from single nucleotide polymorphism (SNP) and gene level was performed subsequently to detect the crucial genomic regions. Finally, we revealed the anatomical change on which genetic overlap relies and further explored whether genetic factors exert a causal effect. RESULTS After using both linkage disequilibrium score regression (LDSC) and High-definition likelihood inference (HDL) methods, we identified a significant genetic correlation between OSA and ADHD (PLDSC = 2.45E-28, PHDL = 1.09E-25), demonstrating a consistent direction. Furthermore, through the application of various cross-trait methods, we pinpointed 5 loci and 57 genes involved in regulating the co-occurrence of these disorders. These genetic regions were thought to be associated with the prefrontal lobes (P = 3.07E-06) and the nucleus accumbens basal ganglia (P = 2.85E-06). Lastly, utilizing Mendelian randomization (MR), we established a link indicating that individuals with ADHD were at an elevated risk of developing OSA (PIVM = 0.02, OR (95%CI):1.09 (1.01-1.17)). CONCLUSIONS This study reveals a strong genetic correlation between ADHD and OSA. It offers insights for future drug target development and sleep management in ADHD.
Collapse
Affiliation(s)
- Yao Tong
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi'an, Shaanxi Province, China
| | - Yan-Jing Chen
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Guang-Bin Cui
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi'an, Shaanxi Province, China.
| |
Collapse
|
9
|
Shou W, Zhang C, Wang Y, Wang H, Guo L, Li L, Zhang T, Huang W, Shi J. Contrastive sequence signatures between the both sides of a recombination spot reveal an adaptation at PPARD locus from standing variation for pleiotropy since out-of-Africa dispersal. BMC Genomics 2025; 26:427. [PMID: 40307732 PMCID: PMC12042533 DOI: 10.1186/s12864-025-11620-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Drug metabolism and transporter genes are a specialized class of genes involved in absorption, distribution, metabolism and excretion. They easily present distinct genetic population differentiation and are vulnerable to natural selection. RESULTS We initiated a study using a special panel of informative genetic markers in such genes and dissected the genetic structure in representative Chinese and worldwide populations. A distinctive sub-population stratification was discovered in extensive Eurasians and resulted from divergence at the PPARD locus. The contrastive sequence signatures between the both sides of a recombination spot prove a selective sweep on this locus for genetic hitchhiking effect. A genealogy-based framework demonstrates the positive selection acting from standing variation exerted a moderate pressure in Eurasians, and drove the adaptive allele up to a high frequency. The timing and tempo estimations for the genetic adaptation indicate its onset coincided with the early out-of-Africa migration of modern humans and it lasted over a prolonged evolutionary history. A phenome-wide association analysis reveals an extended cis-regulation on the local gene expression and the pleiotropy implicated in a variety of complex traits. The colocalization analyses between the genetic associations from cis-acting gene expression and complex traits signify the most likely selective pressure from physical capacity, energy metabolism, and immune-related involvement, and provide prioritization for the effective genes and casual variants. CONCLUSIONS This work has laid a foundation for following efforts to make full sense of the biological mechanisms underlying the genetic adaptation.
Collapse
Affiliation(s)
- Weihua Shou
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, 288 Qianxing Road, Kunming, Yunnan, 650228, P.R. China.
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), 2140 Xietu Road, Shanghai, 200032, P.R. China.
| | - Chenhui Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), 2140 Xietu Road, Shanghai, 200032, P.R. China
| | - Ying Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), 2140 Xietu Road, Shanghai, 200032, P.R. China
| | - Haifeng Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), 2140 Xietu Road, Shanghai, 200032, P.R. China
| | - Lei Guo
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, 288 Qianxing Road, Kunming, Yunnan, 650228, P.R. China
| | - Li Li
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, 288 Qianxing Road, Kunming, Yunnan, 650228, P.R. China
| | - Tiesong Zhang
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, 288 Qianxing Road, Kunming, Yunnan, 650228, P.R. China.
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), 2140 Xietu Road, Shanghai, 200032, P.R. China
| | - Jinxiu Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), 2140 Xietu Road, Shanghai, 200032, P.R. China.
| |
Collapse
|
10
|
Lange LM, Cerquera-Cleves C, Schipper M, Panagiotaropoulou G, Braun A, Kraft J, Awasthi S, Bell N, Posthuma D, Ripke S, Blauwendraat C, Heilbron K. Prioritizing Parkinson's disease risk genes in genome-wide association loci. NPJ Parkinsons Dis 2025; 11:77. [PMID: 40240380 PMCID: PMC12003903 DOI: 10.1038/s41531-025-00933-0] [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: 01/03/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Many drug targets in ongoing Parkinson's disease (PD) clinical trials have strong genetic links. While genome-wide association studies (GWAS) nominate regions associated with disease, pinpointing causal genes is challenging. Our aim was to prioritize additional druggable genes underlying PD GWAS signals. The polygenic priority score (PoPS) integrates genome-wide information from MAGMA gene-level associations and over 57,000 gene-level features. We applied PoPS to East Asian and European PD GWAS data and prioritized genes based on PoPS, distance to the GWAS signal, and non-synonymous credible set variants. We prioritized 46 genes, including well-established PD genes (SNCA, LRRK2, GBA1, TMEM175, VPS13C), genes with strong literature evidence supporting a mechanistic link to PD (RIT2, BAG3, SCARB2, FYN, DYRK1A, NOD2, CTSB, SV2C, ITPKB), and genes relatively unexplored in PD. Many hold potential for drug repurposing or development. We prioritized high-confidence genes with strong links to PD pathogenesis that may represent our next-best candidates for developing disease-modifying therapeutics.
Collapse
Affiliation(s)
- Lara M Lange
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Catalina Cerquera-Cleves
- Neurology Unit, Department of Neurosciences, Hospital Universitario San Ignacio, Bogotá, Colombia
- Centre de recherche du Centre Hospitalier Universitaire de Québec, Axe Neurosciences, Département de Psychiatrie et Neurosciences, Laval University, Québec, QC, Canada
| | | | - Georgia Panagiotaropoulou
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Swapnil Awasthi
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Nathaniel Bell
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Karl Heilbron
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany.
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany.
| |
Collapse
|
11
|
Huang Y, Ju C, Luo J, Li Y. Exploring the shared genetic basis of attention-deficit/hyperactivity disorder and obstructive sleep apnea: A multi-omics analysis. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111369. [PMID: 40246054 DOI: 10.1016/j.pnpbp.2025.111369] [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: 01/13/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Observational studies have suggested an association between attention-deficit/hyperactivity disorder (ADHD) and obstructive sleep apnea (OSA), but these findings are often inconsistent due to potential biases from medication use, and varying diagnostic criteria. Genetic analyses can help mitigate these confounding factors, providing additional evidence. METHODS This study evaluated the genetic correlations between ADHD and OSA using Genome-wide association study (GWAS) summary data, applying linkage disequilibrium score regression (LDSC) and SUPER GeNetic cOVariance Analyzer (SUPERGNOVA). Cross-trait association and colocalization analysis identify potential pleiotropic loci. Tissue enrichment analysis and gene-level analysis of shared genes between OSA and ADHD was conducted. Additionally, bidirectional Mendelian randomization was used to assess potential causal relationships. RESULTS We found significant genetic correlations between ADHD and OSA (rg = 0.309, p = 3.252E-27), and identified 8 novel pleiotropic loci through cross-trait association analysis. Tissue enrichment analysis showed that these shared genes were primarily concentrated in brain tissues, particularly in deep gray matter regions, and were associated with immune and inflammatory pathways. Forward Mendelian Randomization analysis showed that ADHD was significantly associated with the risk of OSA (OR 1.070, 95 % CI 1.013-1.130, p = 0.016), and reverse analysis showed that OSA was significantly associated with the risk of ADHD (OR 1.240, 95 % CI 1.106-1.390, p = 2.213E-4). CONCLUSION The findings of this study show a significant positive genetic correlation between ADHD and OSA and each is a risk factor for the other. Inflammation in specific brain regions may be the underlying mechanism for their comorbidity.
Collapse
Affiliation(s)
- Yijie Huang
- Department of Sleep Medicine, Mental Health Center of Shantou University, Shantou, Guangdong Province, China; Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong Province, China; Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong Province, China
| | - Chao Ju
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Jie Luo
- Department of Sleep Medicine, Mental Health Center of Shantou University, Shantou, Guangdong Province, China; Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong Province, China; Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong Province, China
| | - Yun Li
- Department of Sleep Medicine, Mental Health Center of Shantou University, Shantou, Guangdong Province, China; Sleep Medicine Center, Shantou University Medical College, Shantou, Guangdong Province, China; Shantou University Medical College-Faculty of Medicine of University of Manitoba Joint Laboratory of Biological Psychiatry, Shantou, Guangdong Province, China.
| |
Collapse
|
12
|
Venkatesh SS, Wittemans LBL, Palmer DS, Baya NA, Ferreira T, Hill B, Lassen FH, Parker MJ, Reibe S, Elhakeem A, Banasik K, Bruun MT, Erikstrup C, Aagard Jensen B, Juul A, Mikkelsen C, Nielsen HS, Ostrowski SR, Pedersen OB, Rohde PD, Sørensen E, Ullum H, Westergaard D, Haraldsson A, Holm H, Jonsdottir I, Olafsson I, Steingrimsdottir T, Steinthorsdottir V, Thorleifsson G, Figueredo J, Karjalainen MK, Pasanen A, Jacobs BM, Kalantzis G, Hubers N, Lippincott M, Fraser A, Lawlor DA, Timpson NJ, Nyegaard M, Stefansson K, Magi R, Laivuori H, van Heel DA, Boomsma DI, Balasubramanian R, Seminara SB, Chan YM, Laisk T, Lindgren CM. Genome-wide analyses identify 25 infertility loci and relationships with reproductive traits across the allele frequency spectrum. Nat Genet 2025:10.1038/s41588-025-02156-8. [PMID: 40229599 DOI: 10.1038/s41588-025-02156-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025]
Abstract
Genome-wide association studies (GWASs) may help inform the etiology of infertility. Here, we perform GWAS meta-analyses across seven cohorts in up to 42,629 cases and 740,619 controls and identify 25 genetic risk loci for male and female infertility. We additionally identify up to 269 genetic loci associated with follicle-stimulating hormone, luteinizing hormone, estradiol and testosterone through sex-specific GWAS meta-analyses (n = 6,095-246,862). Exome sequencing analyses reveal that women carrying testosterone-lowering rare variants in some genes are at risk of infertility. However, we find no local or genome-wide genetic correlation between female infertility and reproductive hormones. While infertility is genetically correlated with endometriosis and polycystic ovary syndrome, we find limited genetic overlap between infertility and obesity. Finally, we show that the evolutionary persistence of infertility-risk alleles may be explained by directional selection. Taken together, we provide a comprehensive view of the genetic determinants of infertility across multiple diagnostic criteria.
Collapse
Affiliation(s)
- Samvida S Venkatesh
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Laura B L Wittemans
- Novo Nordisk Research Centre Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Duncan S Palmer
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Nikolas A Baya
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Teresa Ferreira
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Barney Hill
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Frederik Heymann Lassen
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Melody J Parker
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Saskia Reibe
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Ahmed Elhakeem
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Copenhagen, Denmark
| | - Mie T Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Bitten Aagard Jensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Anders Juul
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Christina Mikkelsen
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, Copenhagen University, Copenhagen, Denmark
| | - Henriette S Nielsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, The Fertility Clinic, Hvidovre University Hospital, Copenhagen, Denmark
| | - Sisse R Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Ole B Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital-Køge, Køge, Denmark
| | - Palle Duun Rohde
- Genomic Medicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | | | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Copenhagen, Denmark
| | - Asgeir Haraldsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Children's Hospital Iceland, Landspitali University Hospital, Reykjavik, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Ingileif Jonsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali University Hospital, Reykjavik, Iceland
| | - Thora Steingrimsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Obstetrics and Gynecology, Landspitali University Hospital, Reykjavik, Iceland
| | | | | | - Jessica Figueredo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Minna K Karjalainen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Anu Pasanen
- Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Benjamin M Jacobs
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University London, London, UK
| | | | - Nikki Hubers
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Margaret Lippincott
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Abigail Fraser
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Mette Nyegaard
- Genomic Medicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Statens Serum Institut, Copenhagen, Denmark
| | - Kari Stefansson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Reedik Magi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, The Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Dorret I Boomsma
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ravikumar Balasubramanian
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Stephanie B Seminara
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yee-Ming Chan
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, UK.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
13
|
Gaudio A, Gotta F, Ponti C, Geroldi A, La Barbera A, Mandich P. GWAS by Subtraction to Disentangle RBD Genetic Background from α-Synucleinopathies. Int J Mol Sci 2025; 26:3578. [PMID: 40332088 PMCID: PMC12026788 DOI: 10.3390/ijms26083578] [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: 03/17/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Rapid eye movement (REM) sleep behavior disorder (RBD) is a parasomnia characterized by loss of muscle atonia and abnormal behaviors occurring during REM sleep. Idiopathic RBD (iRBD) is recognized as the strongest prodromal hallmark of α-synucleinopathies, with an established conversion rate to a neurodegenerative condition that reaches up to 96.6% at 15 years of follow-up. Moreover, RBD-converters display a more severe clinical trajectory compared to those that do not present with RBD. However, the extent to which iRBD represents a distinct genetic entity or an early manifestation of neurodegeneration remains unclear. To address this, we applied Genomic Structural Equation Modeling (GenomicSEM) using a GWAS-by-subtraction approach to disentangle the genetic architecture of iRBD from the shared genomic liability across α-synucleinopathies. Our findings highlight the SNCA locus as a key genetic regulator of iRBD susceptibility. While iRBD exhibits a partially distinct genetic signature, residual genomic overlap with neurodegenerative traits suggests that its genetic architecture exists along a continuum of α-synucleinopathy risk. In this scenario, the associations with neuroanatomical correlates may serve as early indicators of a trajectory toward future neurodegeneration. These findings provide a framework for identifying biomarkers that could aid in disease stratification and risk prediction, potentially improving early intervention strategies.
Collapse
Affiliation(s)
- Andrea Gaudio
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Fabio Gotta
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Clarissa Ponti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| | - Alessandro Geroldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| | - Andrea La Barbera
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Paola Mandich
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| |
Collapse
|
14
|
Xu X, Wang S, Zhou H, Tan Q, Lang Z, Zhu Y, Yuan H, Wu Z, Zhu L, Hu K, Li W, Zhou D, Wu M, Wu X. Transcriptome-wide association study of alternative polyadenylation identifies susceptibility genes in non-small cell lung cancer. Oncogene 2025:10.1038/s41388-025-03338-8. [PMID: 40205015 DOI: 10.1038/s41388-025-03338-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/09/2025] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Alternative polyadenylation (APA) plays a crucial role in cancer development and prognosis. However, the molecular characteristics of APA related to non-small cell lung cancer (NSCLC) susceptibility remain understudied, especially in East Asian populations. In this study, we constructed an atlas of APA-regulated 3' untranslated region (3'UTR) and profiled its genetic regulation in 747 lung tissue samples (including tumors and paired normal tissues) from 417 NSCLC Chinese patients. We verified a significant global shortening of 3'UTRs in tumor samples compared to normal samples and underscored the value of APA-regulation as a prognostic marker. The 3'UTR APA quantitative trait loci (3'aQTL) was identified by regressing the percentage of distal poly(A) site usage index (PDUI) value on genetic variants. We found that a significant proportion 3'aQTLs are independent of genetic regulation of expression and are specific in Chinese. We also conducted a 3'UTR APA transcriptome-wide association study (3'aTWAS) by integrating the APA regulation atlas with a genome-wide association study (GWAS) for NSCLC involving 7035 cases and 185,413 cancer-free controls. We identified NSCLC-associated genes, highlighting TUBB, TEAD3, and PPP1R10. Combining the consistent results from colocalization analysis, differential APA analysis, and survival analysis, we provide novel evidence for the role TUBB APA regulation in NSCLC and identified potential upstream regulators. Overall, our study profiled the APA regulation and highlighted the substantial role of APA in NSCLC carcinogenesis and prognosis in East Asian populations.
Collapse
Affiliation(s)
- Xiaohang Xu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, China
| | - Sicong Wang
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, China
| | - Hanyi Zhou
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Qilong Tan
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, China
| | - Zeyong Lang
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Zhu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Huadi Yuan
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zixiang Wu
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling Zhu
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kejia Hu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- National Institute for Data Science in Health and Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenyuan Li
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, China
| | - Dan Zhou
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Intelligent Preventive Medicine, Hangzhou, China
| | - Ming Wu
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xifeng Wu
- Center of Clinical Big Data and Analytics of the Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China.
- National Institute for Data Science in Health and Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- School of Medicine and Health Science, George Washington University, Washington, DC, USA.
- Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
15
|
Lin C, Xia M, Dai Y, Huang Q, Sun Z, Zhang G, Luo R, Peng Q, Li J, Wang X, Lin H, Gao X, Tang H, Shen X, Wang S, Jin L, Hao X, Zheng Y. Cross-ancestry analyses of Chinese and European populations reveal insights into the genetic architecture and disease implication of metabolites. CELL GENOMICS 2025; 5:100810. [PMID: 40118068 PMCID: PMC12008806 DOI: 10.1016/j.xgen.2025.100810] [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: 09/25/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025]
Abstract
Differential susceptibilities to various diseases and corresponding metabolite variations have been documented across diverse ethnic populations, but the genetic determinants of these disparities remain unclear. Here, we performed large-scale genome-wide association studies of 171 directly quantifiable metabolites from a nuclear magnetic resonance-based metabolomics platform in 10,792 Han Chinese individuals. We identified 15 variant-metabolite associations, eight of which were successfully replicated in an independent Chinese population (n = 4,480). By cross-ancestry meta-analysis integrating 213,397 European individuals from the UK Biobank, we identified 228 additional variant-metabolite associations and improved fine-mapping precision. Moreover, two-sample Mendelian randomization analyses revealed evidence that genetically predicted levels of triglycerides in high-density lipoprotein were associated with a higher risk of coronary artery disease and that of glycine with a lower risk of heart failure in both ancestries. These findings enhance our understanding of the shared and specific genetic architecture of metabolites as well as their roles in complex diseases across populations.
Collapse
Affiliation(s)
- Chenhao Lin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qingxia Huang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Guoqing Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; National Genomics Data Center& Bio-Med Big Data Center, University of Chinese Academy of Sciences, Chinese Academy of Science, Shanghai 200031, China
| | - Ruijin Luo
- Shanghai Southgene Technology Co., Ltd., Shanghai 201203, China
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinxi Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xiaofeng Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan University-the People's Hospital of Rugao Joint Research Institute of Longevity and Aging, Rugao, Jiangsu 226500, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xia Shen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, Guangdong 511400, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
16
|
Jiang S, Fan F, Li Q, Zuo L, Xu A, Sun C. Therapeutic Target Discovery for Multiple Myeloma: Identifying Druggable Genes via Mendelian Randomization. Biomedicines 2025; 13:885. [PMID: 40299486 PMCID: PMC12024999 DOI: 10.3390/biomedicines13040885] [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: 02/19/2025] [Revised: 03/22/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Multiple myeloma (MM) is a hematological malignancy originating from the plasma cells present in the bone marrow. Despite significant therapeutic advancements, relapse and drug resistance remain major clinical challenges, highlighting the urgent need for novel therapeutic targets. Methods: To identify potential druggable genes associated with MM, we performed Mendelian randomization (MR) analysis. Causal candidates were further validated using a single-tissue transcriptome-wide association study (TWAS), and colocalization analysis was conducted to assess shared genetic signals between gene expression and disease risk. Potential off-target effects were assessed through an MR phenome-wide association study (MR-PheWAS). Additionally, molecular docking and functional assays were used to evaluate candidate drug efficacy. Results: The MR analysis identified nine druggable genes (FDR < 0.05), among which Orosomucoid 1 (ORM1) and Oviductal Glycoprotein 1 (OVGP1) were supported by both TWAS and colocalization evidence (PPH4 > 0.75). Experimental validation demonstrated the significant downregulation of ORM1 and OVGP1 in MM cells (p < 0.05). Pregnenolone and irinotecan, identified as agonists of ORM1 and OVGP1, respectively, significantly inhibited MM cell viability, while upregulating their expression (p < 0.05). Conclusions: Our study highlights ORM1 and OVGP1 as novel therapeutic targets for MM. The efficacy of pregnenolone and irinotecan in suppressing MM cell growth suggests their potential for clinical application. These findings provide insights into MM pathogenesis and offer a promising strategy for overcoming drug resistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (S.J.); (F.F.); (Q.L.); (L.Z.); (A.X.)
| |
Collapse
|
17
|
Zhao Q, Baranova A, Liu D, Cao H, Zhang F. Bidirectional causal associations between plasma metabolites and bipolar disorder. Mol Psychiatry 2025:10.1038/s41380-025-02977-3. [PMID: 40169804 DOI: 10.1038/s41380-025-02977-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
Altered levels of human plasma metabolites have been implicated in the etiology of bipolar disorder (BD). However, the causality between metabolites and the disease was not well described. We performed a bidirectional metabolome-wide Mendelian randomization (MR) analysis to evaluate the potential causal relationships between 871 plasma metabolites and BD. We used DrugBank and ChEMBL to evaluate whether related metabolites are potential therapeutic targets. Finally, Bayesian colocalization analysis was performed to identify shared genomic loci BD and identified metabolites. Our MR results showed that six metabolites were significantly associated with a reduced risk of BD, including arachidonate (20:4n6) (OR: 0.90, 95% CI: 0.84-0.95) and sphingomyelin (d18:2/24:1, d18:1/24:2) (OR: 0.92, 95% CI: 0.87-0.96), while five metabolites were significantly associated with an increased risk of BD, including 1-palmitoyl-2-linoleoyl-GPE (16:0/18:2) (OR: 1.09, 95% CI: 1.05-1.13). However, our reverse MR analysis showed that BD was not associated with the levels of any metabolite. Additionally, the leave-one-out analysis revealed SNPs within chromosome 11 loci harboring MYRF, FADS1, and FADS2 as ones with the potential to influence partial causal effects. Druggability evaluation showed that 10 of the BD-related metabolites, such as sphingomyelin and cytidine, have been targeted by pharmacologic intervention. Colocalization analysis highlighted one colocalized region (chromosome 11q12) shared by 11 metabolites and BD and pointed to some genes as possible players, including FADS1, FADS2, FADS3, and SYT7. Our study supported a causal role of plasma metabolites in the susceptibility to BD, and the identified metabolites may provide a new avenue for the prevention and treatment of BD.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - 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, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Hongbao Cao
- School of Systems Biology, George Mason University, Fairfax, 22030, USA
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
18
|
Liefferinckx C, Stern D, Perée H, Bottieau J, Mayer A, Dubussy C, Quertinmont E, Tafciu V, Minsart C, Petrov V, Kvasz A, Coppieters W, Karim L, Rahmouni S, Georges M, Franchimont D. The identification of blood-derived response eQTLs reveals complex effects of regulatory variants on inflammatory and infectious disease risk. PLoS Genet 2025; 21:e1011599. [PMID: 40208878 PMCID: PMC12013874 DOI: 10.1371/journal.pgen.1011599] [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: 09/19/2024] [Revised: 04/22/2025] [Accepted: 01/29/2025] [Indexed: 04/12/2025] Open
Abstract
Hundreds of risk loci for immune mediated inflammatory and infectious diseases have been identified by genome-wide association studies (GWAS). Yet, what causal variants and genes in risk loci underpin the observed associations remains poorly understood for most. The identification of colocalized cis-expression Quantitative Trait Loci (cis-eQTLs) is a promising way to identify candidate causative genes. The catalogue of cis-eQTLs of the immune system is likely incomplete as many cis-eQTLs may be context-specific. We built a large cohort of 406 healthy individuals and expanded the immune cis-regulome through their whole blood transcriptome obtained after stimulation with specific toll-like receptor (TLR) agonists and T-cell receptor (TCR) antagonist. We report three mechanisms that may explain why an eQTL could only be revealed after immune stimulation. More than half of the cis-eQTLs detected in this study would have been overlooked without specific immune stimulations. We then mined this new catalogue of response (r)eQTLs, with public GWAS summary statistics of three diseases through a colocalization approach: inflammatory bowel diseases, rheumatoid arthritis and COVID-19 disease. We identified reQTL-specific colocalizations for risk loci for which no matching eQTL were reported before, revealing interesting new candidate causal genes.
Collapse
Affiliation(s)
- Claire Liefferinckx
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - David Stern
- GIGA Bioinformatics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Hélène Perée
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Jérémie Bottieau
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
| | - Alice Mayer
- GIGA Bioinformatics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Christophe Dubussy
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Eric Quertinmont
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Vjola Tafciu
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Charlotte Minsart
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Vyacheslav Petrov
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Alex Kvasz
- Software development, University of Liège, Liège, Belgium
| | - Wouter Coppieters
- GIGA Genomics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Latifa Karim
- GIGA Genomics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Souad Rahmouni
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Michel Georges
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
- WEL Research Institute & Faculty of Veterinary Medicine, Liège, Belgium
| | - Denis Franchimont
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
19
|
Harigaya Y, Matoba N, Le BD, Valone JM, Stein JL, Love MI, Valdar W. Probabilistic classification of gene-by-treatment interactions on molecular count phenotypes. PLoS Genet 2025; 21:e1011561. [PMID: 40203278 PMCID: PMC12021428 DOI: 10.1371/journal.pgen.1011561] [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: 08/08/2024] [Revised: 04/24/2025] [Accepted: 12/31/2024] [Indexed: 04/11/2025] Open
Abstract
Genetic variation can modulate response to treatment (G×T) or environmental stimuli (G×E), both of which can be highly consequential in biomedicine. An effective approach to identifying G×T signals and gaining insight into molecular mechanisms is mapping quantitative trait loci (QTL) of molecular count phenotypes, such as gene expression and chromatin accessibility, under multiple treatment conditions, which is termed response molecular QTL mapping. Although standard approaches evaluate the interaction between genetics and treatment conditions, they do not distinguish between meaningful interpretations such as whether a genetic effect is observed only in the treated condition or whether a genetic effect is observed always but accentuated in the treated condition. To address this gap, we have developed a downstream method for classifying response molecular QTLs into subclasses with meaningful genetic interpretations. Our method uses Bayesian model selection and assigns posterior probabilities to different types of G×T interactions for a given feature-SNP pair. We compare linear and nonlinear regression of log -scale counts, noting that the latter accounts for an expected biological relationship between the genotype and the molecular count phenotype. Through simulation and application to existing datasets of molecular response QTLs, we show that our method provides an intuitive and well-powered framework to report and interpret G×T interactions. We provide a software package, ClassifyGxT [1].
Collapse
Affiliation(s)
- Yuriko Harigaya
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Brandon D Le
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jordan M Valone
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Carolina Institute for Developmental Disabilities, Carrboro, North Carolina, United States of America
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - William Valdar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
20
|
Liu J, Zeng D, Wang Y, Deng F, Wu S, Deng Z. Identification of druggable targets in acute kidney injury by proteome- and transcriptome-wide Mendelian randomization and bioinformatics analysis. Biol Direct 2025; 20:38. [PMID: 40148878 PMCID: PMC11951703 DOI: 10.1186/s13062-025-00631-0] [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: 01/28/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) remains a critical condition with limited therapeutic options, predominantly managed by renal replacement therapy. The challenge of developing targeted treatments persists. METHODS We integrated genetic data related to druggable proteins and gene expression with AKI genome-wide association study (GWAS) findings. Based on multi-omics Mendelian randomization (MR), we identified the potential causal influence of 5,883 unique proteins and genes on AKI. We also performed using reverse MR and external cohort-based analysis to verify the robustness of this causal relationship. Expression patterns of these targets were examined using bulk transcriptome and single-cell transcriptome data. In addition, drug repurposing analyses were conducted to explore the potential of existing medications. We also constructed a molecular interaction network to explore the interplay between identified targets and known drugs. RESULTS Genetically predicted levels of seven proteins and twelve genes were associated with an increased risk of AKI. Of these, six targets (NCF1, TNFRSF1B, APEH, ACADSB, ADD1, and FAM3B) were prioritized based on robust evidence and validated in independent cohorts. Reverse MR showed a one-way causal relationship of targets. These targets are predominantly expressed in proximal tubular cells, endothelial cells, collecting duct-principal cells, and immune cells within both AKI-affected and normal tissues. Several promising drug repurposing opportunities were identified, such as telmisartan-NCF1, calcitriol-ACADSB, and ethinyl estradiol-ACADSB. The molecular interaction mapping and pathway integration analysis provided further insights, suggesting potential strategies for combinatorial therapies. CONCLUSIONS This extensive investigation identified several promising therapeutic targets for AKI and highlighted opportunities for drug repurposing. These findings offer valuable insights that could shape future research and the development of targeted treatments.
Collapse
Affiliation(s)
- Jiachen Liu
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dianjie Zeng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
| | - Fei Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuiqing Wu
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China.
| | - Zebin Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China.
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Feng H, Chen Z, Li J, Feng J, Yang F, Meng F, Yin H, Guo Y, Xu H, Liu Y, Liu R, Lou W, Liu L, Han X, Su H, Zhang L. Unveiling circulating targets in pancreatic cancer: Insights from proteogenomic evidence and clinical cohorts. iScience 2025; 28:111693. [PMID: 40060891 PMCID: PMC11889678 DOI: 10.1016/j.isci.2024.111693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 09/23/2024] [Accepted: 12/23/2024] [Indexed: 03/04/2025] Open
Abstract
Pancreatic cancer (PC), characterized by the absence of effective biomarkers and therapies, remains highly fatal. Data regarding the correlations between PC risk and individual plasma proteome known for minimally invasive biomarkers are scarce. Here, we analyzed 1,345 human plasma proteins using proteome-wide association studies, identifying 78 proteins significantly associated with PC risk. Of these, four proteins (ROR1, FN1, APOA5, and ABO) showed the most substantial causal link to PC, confirmed through Mendelian randomization and colocalization analyses. Data from two clinical cohorts further demonstrated that FN1 and ABO were notably overexpressed in both blood and tumor samples from PC patients, compared to healthy controls or para-tumor tissues. Additionally, elevated FN1 and ABO levels correlated with shorter median survival in patients. Multiple drugs targeting FN1 or ROR1 are available or in clinical trials. These findings suggest that plasma protein FN1 associated with PC holds potential as both prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Haokang Feng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhixue Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jianang Li
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiale Feng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Fei Yang
- Innovative Institute of Tumor Immunity and Medicine (ITIM), Hefei, Anhui, China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Fansheng Meng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hanlin Yin
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuquan Guo
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxin Liu
- Department of Basic Medicine and Institute of Liver Diseases, Shan Xi Medical University, Taiyuan 030000, China
| | - Runjie Liu
- Department of Basic Medicine and Institute of Liver Diseases, Shan Xi Medical University, Taiyuan 030000, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- The Shanghai Geriatrics Medical Center, Zhongshan Hospital MinHang MeiLong Branch, Fudan University, Shanghai 201104, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xu Han
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hua Su
- Institutes of Biomedical Sciences, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
22
|
Mori Y, van Dijk EHC, Miyake M, Hosoda Y, den Hollander AI, Yzer S, Miki A, Chen LJ, Ahn J, Takahashi A, Morino K, Nakao SY, Hoyng CB, Ng DSC, Cen LP, Chen H, Ng TK, Pang CP, Joo K, Sato T, Sakata Y, Tajima A, Tabara Y, Park KH, Matsuda F, Yamashiro K, Honda S, Nagasaki M, Boon CJF, Tsujikawa A. Genome-wide association and multi-omics analyses provide insights into the disease mechanisms of central serous chorioretinopathy. Sci Rep 2025; 15:9158. [PMID: 40097481 PMCID: PMC11914043 DOI: 10.1038/s41598-025-92210-6] [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: 01/23/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Central serous chorioretinopathy (CSC) is a major cause of vision loss, especially in middle-aged men, and its chronic subtype can lead to legal blindness. Despite its clinical importance, the underlying mechanisms of CSC need further clarification. In this study, we conducted a meta-analysis of three genome-wide association studies (GWASs) for CSC consisting of 8811 Asians and Caucasians, followed by replication in an additional 4338 Asians. We identified four genome-wide hits, including a novel hit (rs12960630 at LINC01924-CDH7, Pmeta = 2.97 × 10-9). A phenome-wide association study for rs12960630 showed a positive correlation between its CSC risk allele with plasma cortisol concentration. Expression/splicing quantitative trait loci (QTL) analyses showed an association of all these hits with the expression and/or splicing of genes in genital organs, which may explain the sex differences in CSC. Protein QTL also suggested the protein-level contribution of the complement factor H pathway to CSC pathogenesis.
Collapse
Affiliation(s)
- Yuki Mori
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Elon H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Masahiro Miyake
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan.
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | | | | | - Suzanne Yzer
- Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Akiko Miki
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeeyun Ahn
- Seoul National University, College of Medicine, Seoul, Korea
- SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Ayako Takahashi
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
| | - Kazuya Morino
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shin-Ya Nakao
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Danny S C Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling-Ping Cen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Haoyu Chen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Kwangsic Joo
- Seoul National University, College of Medicine, Seoul, Korea
- Seoul National University Bundang Hospital, Seongnam, Korea
| | - Takehiro Sato
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasuhiko Sakata
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasuharu Tabara
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Kyu Hyung Park
- Seoul National University, College of Medicine, Seoul, Korea
- Seoul National University Hospital, Seoul, Korea
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Science, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Shigeru Honda
- Department of Ophthalmology and Visual Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masao Nagasaki
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Ophthalmology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Akitaka Tsujikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara, Sakyo, Kyoto, 606-8507, Japan
| |
Collapse
|
23
|
Chen Y, Deng J, Zhang Z, Wang C, Yu X. Genetic overlap between multi-site chronic pain and cognition: a large-scale genome-wide cross-trait analysis. Front Neurosci 2025; 19:1466278. [PMID: 40177376 PMCID: PMC11962220 DOI: 10.3389/fnins.2025.1466278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025] Open
Abstract
Background Different studies have consistently demonstrated a positive correlation between chronic pain and cognitive changes. This study aimed to explore the genetic factors underlying the relationship between chronic pain and cognitive traits, and to investigate whether an inherent causal connection exists between them. Method The genetic contributions of chronic multi-site pain and eight cognitive traits were investigated based on Genome-wide association studies (GWAS) data. Linkage disequilibrium score regression (LDSC) was employed to assess the genetic correlations between each pair of traits. The shared genetic components of these traits were investigated by identifying single nucleotide polymorphisms (SNPs) with pleiotropic effects using the Cross Phenotype Association (CPASSOC) method. Furthermore, enrichment analysis and transcriptome-wide association studies (TWAS) were performed to characterize the significant associations between genetic traits. The latent causal variable model (LCV) was employed to explore the potential causal relationship between both traits. Results A significant negative genetic correlation was found between chronic pain and several cognitive functions, particularly intelligence (rg = -0. 11, p = 7.77 × 10-64). CPASSOC identified 150 pleiotropic loci. A co-localization analysis was conducted, which identified 20 loci exhibiting pleiotropic effects at the same genomic position. The LCV analysis indicated no causal relationship between both traits. Conclusion The present work contributed to an enhanced understanding of the complex genetic interplay between cognitive function and chronic pain.
Collapse
Affiliation(s)
- Yanjing Chen
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiankai Deng
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiyi Zhang
- Department of Massage, Quanzhou Orthopedic-Traumatological Hospital, Quanzhou, Fujian, China
| | - Chenlin Wang
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Xuegao Yu
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Zhou X, Ye X, Yao J, Lin X, Weng Y, Huang Y, Lu Y, Shang J, Nong L. Identification and validation of transcriptome-wide association study-derived genes as potential druggable targets for osteoarthritis. Bone Joint Res 2025; 14:224-235. [PMID: 40079200 PMCID: PMC11904851 DOI: 10.1302/2046-3758.143.bjr-2024-0251.r1] [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] [Indexed: 03/14/2025] Open
Abstract
Aims Osteoarthritis (OA) is a widespread chronic degenerative joint disease with an increasing global impact. The pathogenesis of OA involves complex interactions between genetic and environmental factors. Despite this, the specific genetic mechanisms underlying OA remain only partially understood, hindering the development of targeted therapeutic strategies. Methods A transcriptome-wide association study (TWAS) was conducted for site-specific OA phenotypes using functional summary-based imputation (FUSION). High-confidence candidate genes were identified through rigorous quality control measures, including joint/conditional analysis, permutation tests, best model evaluation, and colocalization analysis. Co-expression network analysis was performed to elucidate the functional biology of these candidate genes. Druggable gene targets and their structural models were retrieved from the DrugBank and SWISS-MODEL databases. Finally, the enrichment of mitogen-activated protein kinase 3 (MAPK3) and SMAD3 in OA was validated biochemically using in vitro and in vivo OA models, as well as human histological sections. Results Utilizing the FUSION algorithm, TWAS identified 794 candidate genes for OA. After quality control, 14 genes were classified as high-confidence genes, with seven recognized as potential drug targets including GCAT, MAPK3, MST1R, PFKM, RAD9A, SMAD3, and USAP8. Co-expression analysis revealed a strong biological association between SMAD3 and MAPK3. Both in vitro and in vivo experiments demonstrated high activity and enriched expression of these two genes in OA. Conclusion The present study identified tissue-specific candidate genes and validated high-confidence druggable targets for OA, providing new insights into the genetic landscape and biological processes involved in OA. Further functional studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai, China
| | - Xinjian Ye
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Jiapei Yao
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Xiaolong Lin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yiping Weng
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Yaojun Lu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - JingJing Shang
- Department of Pharmacy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Luming Nong
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
25
|
Zhan H, Cammann D, Cummings JL, Dong X, Chen J. Biomarker identification for Alzheimer's disease through integration of comprehensive Mendelian randomization and proteomics data. J Transl Med 2025; 23:278. [PMID: 40050982 PMCID: PMC11884171 DOI: 10.1186/s12967-025-06317-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/23/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the main cause of dementia with few effective therapies. We aimed to identify potential plasma biomarkers or drug targets for AD by investigating the causal association between plasma proteins and AD by integrating comprehensive Mendelian randomization (MR) and multi-omics data. METHODS Using two-sample MR, cis protein quantitative trait loci (cis-pQTLs) for 1,916 plasma proteins were used as an exposure to infer their causal effect on AD liability in individuals of European ancestry, with two large-scale AD genome-wide association study (GWAS) datasets as the outcome for discovery and replication. Significant causal relationships were validated by sensitivity analyses, reverse MR analysis, and Bayesian colocalization analysis. Additionally, we investigated the causal associations at the transcriptional level with cis gene expression quantitative trait loci (cis-eQTLs) data across brain tissues and blood in European ancestry populations, as well as causal plasma proteins in African ancestry populations. RESULTS In those of European ancestry, the genetically predicted levels of five plasma proteins (BLNK, CD2AP, GRN, PILRA, and PILRB) were causally associated with AD. Among these five proteins, GRN was protective against AD, while the rest were risk factors. Consistent causal effects were found in the brain for cis-eQTLs of GRN, BLNK, and CD2AP, while the same was true for PILRA in the blood. None of the plasma proteins were significantly associated with AD in persons of African ancestry. CONCLUSIONS Comprehensive MR analyses with multi-omics data identified five plasma proteins that had causal effects on AD, highlighting potential biomarkers or drug targets for better diagnosis and treatment for AD.
Collapse
Affiliation(s)
- Hui Zhan
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
- School of Life Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Xianjun Dong
- Stephen and Denise Adams Center for Parkinson's Disease Research, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurology and Section of Biomedical Informatics and Data Science (BIDS), Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jingchun Chen
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
- School of Life Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
| |
Collapse
|
26
|
Qi Y, Jiang H, Lun Y, Gang Q, Shen S, Zhang H, Liu M, Wang Y, Zhang J. Protein Drug Targets for Abdominal Aortic Aneurysm and Proteomic Associations Between Modifiable Risk Factors and Abdominal Aortic Aneurysm. J Am Heart Assoc 2025; 14:e037802. [PMID: 40008516 DOI: 10.1161/jaha.124.037802] [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: 08/05/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a severe aortic disease for which no pharmacological interventions have yet been developed. This investigation focused on identifying protein-based therapeutic targets and assessing how proteins mediate the interplay between modifiable risk factors and AAA development. METHODS Causal inferences between plasma proteins and AAA were drawn using 2-sample Mendelian randomization, followed by comprehensive sensitivity testing, colocalization, and replication efforts. Further analyses included database interrogation, single-cell RNA data analysis, enrichment analysis, protein-protein interaction networks, and immunohistochemistry to map the tissue-specific expression of these proteins, their expression within AAA tissues, and their biological roles. Mediation Mendelian randomization was employed to evaluate the mediating effects of AAA-related proteins on the associations between AAA and 3 risk factors: hypertension, smoking, and obesity. RESULTS A total of 43 proteins were identified as having causal links to AAA. Colocalization analysis pinpointed 13 proteins with strong evidence of colocalization with AAA. Of these, the causal involvement of 10 proteins was substantiated by external validation data. Consistent evidence for PCSK9 (proprotein convertase subtilisin/kexin type 9), IL6R (interleukin-6R), ECM1 (extracellular matrix protein 1), and ANGPTL4 (angiopoietin-related protein 4) was further validated through tissue immunohistochemistry and blood data. Moreover, Mendelian randomization analysis identified 10 proteins as mediators of the influence of hypertension, smoking, and obesity on AAA development. CONCLUSIONS This analysis identifies 4 proteins (PCSK9, IL6R, ECM1, and ANGPTL4) as high-priority therapeutic targets for AAA and emphasizes the intermediary role of plasma proteins in linking hypertension, smoking, obesity, and AAA. Further investigations are needed to clarify the specific roles of these proteins in AAA pathology.
Collapse
Affiliation(s)
- Yao Qi
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Jiang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yu Lun
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Qingwei Gang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Shikai Shen
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Han Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Mingyu Liu
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Yixian Wang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| | - Jian Zhang
- Department of Vascular and Thyroid Surgery The First Hospital of China Medical University Shenyang Liaoning China
| |
Collapse
|
27
|
Yang W, Xiao W, Liu X, Li H, Huang T, Fan D. Testosterone Supplementation: A Potential Therapeutic Strategy for Amyotrophic Lateral Sclerosis. Biomedicines 2025; 13:622. [PMID: 40149599 PMCID: PMC11940241 DOI: 10.3390/biomedicines13030622] [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: 01/08/2025] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives: Amyotrophic lateral sclerosis (ALS) is a progressive and fatal disease characterized by the degeneration of spinal cord and brain neurons. Proteomics combined with Mendelian randomization (MR) is an effective method for finding disease treatment targets. Methods: We aimed to seek new therapeutic targets for ALS. A large-scale GWAS on proteomics (4907 circulatory protein) with 35,559 individuals was included as the exposure data; a GWAS with 138,086 ALS patients was used as the outcome data; we found that a high level of sex hormone-binding globulin (SHBG) is a risk factor by MR analysis. Colocalization analyses were used to validate the causality between SHBG and ALS further. Functional enrichment found a high level of SHBG was associated with a low level of bioavailable testosterone. Two-sample MR confirmed the association of SHBG (400,210 samples), bioavailable testosterone (367,289 samples), and ALS. Results: A high level of SHBG, and a low level of bioavailable testosterone are risk factors for ALS. Conclusions: A low level of bioavailable testosterone is a risk factor for ALS. Although our study is relatively limited and cannot fully confirm that testosterone supplementation has a therapeutic effect on ALS, it offers a promising direction for ALS therapy.
Collapse
Affiliation(s)
- Wenzhi Yang
- Department of Neurology, Peking University Third Hospital, Beijing 100080, China; (W.Y.); (X.L.)
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100080, China
| | - Wendi Xiao
- School of Public Health, Peking University, Beijing 100080, China
| | - Xiangyi Liu
- Department of Neurology, Peking University Third Hospital, Beijing 100080, China; (W.Y.); (X.L.)
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100080, China
| | - Hui Li
- Key Laboratory of Carcinogenesis and Translational Research, Peking University, Beijing 100080, China
| | - Tao Huang
- School of Public Health, Peking University, Beijing 100080, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing 100080, China; (W.Y.); (X.L.)
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100080, China
| |
Collapse
|
28
|
Ray NR, Bradley J, Yilmaz E, Kizil C, Kurup JT, Martin ER, Klein HU, Kunkle BW, Bennett DA, De Jager PL, Beecham GW, Cruchaga C, Reitz C. Local genetic covariance analysis with lipid traits identifies novel loci for early-onset Alzheimer's Disease. PLoS Genet 2025; 21:e1011631. [PMID: 40096060 PMCID: PMC11984970 DOI: 10.1371/journal.pgen.1011631] [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: 09/12/2024] [Revised: 04/10/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
The genetic component of early-onset Alzheimer disease (EOAD), accounting for ~10% of all Alzheimer's disease (AD) cases, is largely unexplained. Recent studies suggest that EOAD may be enriched for variants acting in the lipid pathway. The current study examines the shared genetic heritability between EOAD and the lipid pathway using genome-wide multi-trait genetic covariance analyses. Summary statistics were obtained from the GWAS meta-analyses of EOAD by the Alzheimer's Disease Genetics Consortium (n=19,668) and five blood lipid traits by the Global Lipids Genetics Consortium (n=1,320,016). The significant results were compared between the EOAD and lipids GWAS and genetic covariance analyses were performed via SUPERGNOVA. Genes in linkage disequilibrium (LD) with top EOAD hits in identified regions of covariance with lipid traits were scored and ranked for causality by combining evidence from gene-based analysis, AD-risk scores incorporating transcriptomic and proteomic evidence, eQTL data, eQTL colocalization analyses, DNA methylation data, and single-cell RNA sequencing analyses. Direct comparison of GWAS results showed 5 loci overlapping between EOAD and at least one lipid trait harboring APOE, TREM2, MS4A4E, LILRA5, and LRRC25. Local genetic covariance analyses identified 3 regions of covariance between EOAD and at least one lipid trait. Gene prioritization nominated 3 likely causative genes at these loci: ANKDD1B, CUZD1, and MS4A64.The current study identified genetic covariance between EOAD and lipids, providing further evidence of shared genetic architecture and mechanistic pathways between the two traits.
Collapse
Affiliation(s)
- Nicholas R. Ray
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Joseph Bradley
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Elanur Yilmaz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Jiji T. Kurup
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Eden R. Martin
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Hans-Ulrich Klein
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Brian W. Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - David A. Bennett
- RUSH Alzheimer’s Disease Center, RUSH University, Chicago, Illinois, United States of America
| | - Philip L. De Jager
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | | | - Gary W. Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Christiane Reitz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| |
Collapse
|
29
|
Michaëlsson K, Zheng R, Baron JA, Fall T, Wolk A, Lind L, Höijer J, Brunius C, Warensjö Lemming E, Titova OE, Svennblad B, Larsson SC, Yuan S, Melhus H, Byberg L, Brooke HL. Cardio-metabolic-related plasma proteins reveal biological links between cardiovascular diseases and fragility fractures: a cohort and Mendelian randomisation investigation. EBioMedicine 2025; 113:105580. [PMID: 39919333 PMCID: PMC11848109 DOI: 10.1016/j.ebiom.2025.105580] [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: 03/05/2024] [Revised: 12/17/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND How cardiovascular diseases (CVD) predispose to a higher risk of fragility fractures is not well understood. Both contribute to significant components of disease burden and health expenditure. Poor bone quality, central obesity, sarcopenia, falls, and low grip strength are independent risk factors for hip and other fragility fractures and also for CVD and early death. METHODS We used proteomics and a cohort design combined with Mendelian randomisation analysis to understand shared mechanisms for developing CVD and fragility fractures, two significant sources of disease burden and health expenditure. We primarily aimed to discover and replicate the association of 274 cardio-metabolic-related proteins with future rates of hip and any fracture in two separate population-based cohorts, with a total of 12,314 women and men. FINDINGS The average age at baseline was 68 years in the discovery cohort of women and 74 years in the mixed-sex replication cohort. During 100,619 person-years of follow-up, 2168 had any fracture, and 538 had a hip fracture. Our analysis resulted in 24 cardiometabolic proteins associated with fracture risk: 20 with hip fracture, 9 with any fracture, and 5 with both. The associations remained even if protein concentrations were measured from specimens taken during preclinical stages of cardio-metabolic diseases, and 19 associations remained after adjustment for bone mineral density. Twenty-two of the proteins were associated with total body fat mass or lean body mass. Mendelian randomisation (MR) analysis supported causality since genetically predicted levels of SOST (Sclerostin), CCDC80 (Coiled-coil domain-containing protein 80), NT-proBNP (N-terminal prohormone brain natriuretic peptide), and BNP (Brain natriuretic peptide) were associated with risk of hip fracture. MR analysis also revealed a possible negative impact on bone mineral density (BMD) by genetically predicted higher levels of SOST, CCDC80, and TIMP4 (Metalloproteinase inhibitor 4). The MR association with BMD was positive for PTX3 (Pentraxin-related protein) and SPP1 (Osteopontin). Genetically predicted higher concentrations of SOST and lower concentrations of SPP1 also conferred a higher risk of falls and lowered grip strength. The genetically determined concentration of nine proteins influenced fat mass, and one influenced lean body mass. INTERPRETATION These data reveal biological links between cardiovascular diseases and fragility fractures. The proteins should be further evaluated as shared targets for developing pharmacological interventions to prevent fractures and cardiovascular disease. FUNDING The study was supported by funding from the Swedish Research Council (https://www.vr.se; grants No. 2015-03257, 2017-00644, 2017-06100, and 2019-01291 to Karl Michaëlsson) and funding from Olle Engkvist Byggmästares stiftelse (SOEB).
Collapse
Affiliation(s)
- Karl Michaëlsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Rui Zheng
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - John A Baron
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Tove Fall
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Alicja Wolk
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonas Höijer
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Carl Brunius
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Eva Warensjö Lemming
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Olga E Titova
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Bodil Svennblad
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susanna C Larsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Melhus
- Clinical Pharmacology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Liisa Byberg
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hannah L Brooke
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
30
|
Hecker D, Song X, Baumgarten N, Diagel A, Katsaouni N, Li L, Li S, Maji RK, Behjati Ardakani F, Ma L, Tews D, Wabitsch M, Björkegren JL, Schunkert H, Chen Z, Schulz MH. Cell type-specific epigenetic regulatory circuitry of coronary artery disease loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639228. [PMID: 40027824 PMCID: PMC11870499 DOI: 10.1101/2025.02.20.639228] [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: 03/05/2025]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Recently, hundreds of genomic loci have been shown to increase CAD risk, however, the molecular mechanisms underlying signals from CAD risk loci remain largely unclear. We sought to pinpoint the candidate causal coding and non-coding genes of CAD risk loci in a cell type-specific fashion. We integrated the latest statistics of CAD genetics from over one million individuals with epigenetic data from 45 relevant cell types to identify genes whose regulation is affected by CAD-associated single nucleotide variants (SNVs) via epigenetic mechanisms. Applying two statistical approaches, we identified 1,580 genes likely involved in CAD, about half of which have not been associated with the disease so far. Enrichment analysis and phenome-wide association studies linked the novel candidate genes to disease-specific pathways and CAD risk factors, corroborating their disease relevance. We showed that CAD-SNVs are enriched to regulate gene expression by affecting the binding of transcription factors (TFs) with cellular specificity. Of all the candidate genes, 23.5% represented non-coding RNAs (ncRNA), which likewise showed strong cell type specificity. We conducted a proof-of-concept biological validation for the novel CAD ncRNA gene IQCH-AS1 . CRISPR/Cas9-based gene knockout of IQCH-AS1 , in a human preadipocyte strain, resulted in reduced preadipocyte proliferation, less adipocyte lipid accumulation, and atherogenic cytokine profile. The cellular data are in line with the reduction of IQCH-AS1 in adipose tissues of CAD patients and the negative impact of risk alleles on its expression, suggesting IQCH-AS1 to be protective for CAD. Our study not only pinpoints CAD candidate genes in a cell type-specific fashion but also spotlights the roles of the understudied ncRNA genes in CAD genetics.
Collapse
Affiliation(s)
- Dennis Hecker
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Xiaoning Song
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nina Baumgarten
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Anastasiia Diagel
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nikoletta Katsaouni
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Ling Li
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Shuangyue Li
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ranjan Kumar Maji
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Fatemeh Behjati Ardakani
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| | - Lijiang Ma
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York 10029, USA
| | - Daniel Tews
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075 Ulm, Germany
| | - Martin Wabitsch
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075 Ulm, Germany
| | - Johan L.M. Björkegren
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York 10029, USA
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, 14157 Huddinge, Sweden
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Zhifen Chen
- Department of Cardiology, German Heart Centre Munich, School of Medicine and Health, Technical University of Munich, 80636 Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany
| | - Marcel H. Schulz
- Department of Medicine, Institute for Computational Genomic Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Rhein-Main, Germany
| |
Collapse
|
31
|
Wang ZZ, Yang JL, Zhang ZY, Wang PB. Genetic insights into the shared molecular mechanisms of Crohn's disease and breast cancer: a Mendelian randomization and deep learning approach. Discov Oncol 2025; 16:198. [PMID: 39964572 PMCID: PMC11836263 DOI: 10.1007/s12672-025-01978-6] [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: 09/24/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
The objective of this study was to explore the potential genetic link between Crohn's disease and breast cancer, with a focus on identifying druggable genes that may have therapeutic relevance. We assessed the causal relationship between these diseases through Mendelian randomization and investigated gene-drug interactions using computational predictions. This study sought to identify common genetic pathways possibly involved in immune responses and cancer progression, providing a foundation for future targeted treatment research. The dataset comprises single nucleotide polymorphisms used as instrumental variables for Crohn's disease, analyzed to explore their possible impact on breast cancer risk. Gene ontology and pathway enrichment analyses were conducted to identify genes shared between the two conditions, supported by protein-protein interaction networks, colocalization analyses, and deep learning-based predictions of gene-drug interactions. The identified hub genes and predicted gene-drug interactions offer preliminary insights into possible therapeutic targets for breast cancer and immune-related conditions. This dataset may be valuable for researchers studying genetic links between autoimmune diseases and cancer and for those interested in the early identification of potential drug targets.
Collapse
Affiliation(s)
- Zhuang Zhuang Wang
- Graduate School of Bengbu Medical University, No. 2600 Donghai Avenue, Bengbu, 233030, China
| | - Ju Lin Yang
- Graduate School of Bengbu Medical University, No. 2600 Donghai Avenue, Bengbu, 233030, China
| | - Zong Yao Zhang
- Department of General Surgery, The First Hospital of Anhui University of Science and Technology, No.203 Huai Bin Road, Tian Jia' an District, Huainan, 232007, China.
| | - Pei Bin Wang
- Department of General Surgery, The First Hospital of Anhui University of Science and Technology, No.203 Huai Bin Road, Tian Jia' an District, Huainan, 232007, China.
| |
Collapse
|
32
|
Fu L, Liu Q, Cheng H, Zhao X, Xiong J, Mi J. Insights Into Causal Effects of Genetically Proxied Lipids and Lipid-Modifying Drug Targets on Cardiometabolic Diseases. J Am Heart Assoc 2025; 14:e038857. [PMID: 39868518 PMCID: PMC12074789 DOI: 10.1161/jaha.124.038857] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The differential impact of serum lipids and their targets for lipid modification on cardiometabolic disease risk is debated. This study used Mendelian randomization to investigate the causal relationships and underlying mechanisms. METHODS Genetic variants related to lipid profiles and targets for lipid modification were sourced from the Global Lipids Genetics Consortium. Summary data for 10 cardiometabolic diseases were compiled from both discovery and replication data sets. Expression quantitative trait loci data from relevant tissues were employed to evaluate significant lipid-modifying drug targets. Comprehensive analyses including colocalization, mediation, and bioinformatics were conducted to validate the results and investigate potential mediators and mechanisms. RESULTS Significant causal associations were identified between lipids, lipid-modifying drug targets, and various cardiometabolic diseases. Notably, genetic enhancement of LPL (lipoprotein lipase) was linked to reduced risks of myocardial infarction (odds ratio [OR]1, 0.65 [95% CI, 0.57-0.75], P1=2.60×10-9; OR2, 0.59 [95% CI, 0.49-0.72], P2=1.52×10-7), ischemic heart disease (OR1, 0.968 [95% CI, 0.962-0.975], P1=5.50×10-23; OR2, 0.64 [95% CI, 0.55-0.73], P2=1.72×10-10), and coronary heart disease (OR1, 0.980 [95% CI, 0.975-0.985], P1=3.63×10-14; OR2, 0.64 [95% CI, 0.54-0.75], P2=6.62×10-8) across 2 data sets. Moreover, significant Mendelian randomization and strong colocalization associations for the expression of LPL in blood and subcutaneous adipose tissue were linked with myocardial infarction (OR, 0.918 [95% CI, 0.872-0.967], P=1.24×10-3; PP.H4, 0.99) and coronary heart disease (OR, 0.991 [95% CI, 0.983-0.999], P=0.041; PP.H4=0.92). Glucose levels and blood pressure were identified as mediators in the total effect of LPL on cardiometabolic outcomes. CONCLUSIONS The study substantiates the causal role of lipids in specific cardiometabolic diseases, highlighting LPL as a potent drug target. The effects of LPL are suggested to be influenced by changes in glucose and blood pressure, providing insights into its mechanism of action.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
| | - Qin Liu
- Department of UltrasoundChildren’s Hospital of the Capital Institute of PediatricsBeijingChina
| | - Hong Cheng
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Xiaoyuan Zhao
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control DepartmentShenzhen Center for Chronic Disease ControlShenzhenChina
| | - Jie Mi
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
- Key Laboratory of Major Diseases in Children, Ministry of EducationChina
| |
Collapse
|
33
|
Kouhsar M, Weymouth L, Smith AR, Imm J, Bredemeyer C, Wedatilake Y, Torkamani A, Bergh S, Selbæk G, Mill J, Ballard C, Sweet RA, Kofler J, Creese B, Pishva E, Lunnon K. A brain DNA co-methylation network analysis of psychosis in Alzheimer's disease. Alzheimers Dement 2025; 21:e14501. [PMID: 39936280 PMCID: PMC11815327 DOI: 10.1002/alz.14501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The presence of psychosis in Alzheimer's disease (AD) is suggested to be associated with distinct molecular and neuropathological profiles in the brain. METHODS We assessed brain DNA methylation in AD donors with psychosis (AD+P) and without psychosis (AD-P) using the EPIC array. Weighted gene correlation network analysis identified modules of co-methylated genes in a discovery cohort (PITT-ADRC: N = 113 AD+P, N = 40 AD-P), with validation in an independent cohort (BDR: N = 79 AD+P, N = 117 AD-P), with Gene Ontology and cell-type enrichment analysis. Genetic data were integrated to identify methylation quantitative trait loci (mQTLs), which were co-localized with GWAS for related traits. RESULTS We replicated one AD+P associated module, which was enriched for synaptic pathways and in excitatory and inhibitory neurons. mQTLs in this module co-localized with variants associated with schizophrenia and educational attainment. DISCUSSION This represents the largest epigenetic study of AD+P to date, identifying pleiotropic relationships between AD+P and related traits. HIGHLIGHTS DNA methylation was assessed in the prefrontal cortex in subjects with AD+P and AD-P. WGCNA identified six modules of co-methylated loci associated with AD+P in a discovery cohort. One of the modules was replicated in an independent cohort. This module was enriched for synaptic genes and in excitatory and inhibitory neurons. mQTLs mapping to genes in the module co-localized with GWAS loci for schizophrenia and educational attainment.
Collapse
Affiliation(s)
- Morteza Kouhsar
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Luke Weymouth
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Adam R. Smith
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Jennifer Imm
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Claudia Bredemeyer
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Yehani Wedatilake
- Norwegian National Centre for Aging and HealthVestfold Hospital TrustTønsbergNorway
- Research Centre for Age‐related Functional Decline and DiseaseInnlandet Hospital TrustOttestadNorway
| | | | - Sverre Bergh
- Norwegian National Centre for Aging and HealthVestfold Hospital TrustTønsbergNorway
- Research Centre for Age‐related Functional Decline and DiseaseInnlandet Hospital TrustOttestadNorway
| | - Geir Selbæk
- Norwegian National Centre for Aging and HealthVestfold Hospital TrustTønsbergNorway
- Department of Geriatric MedicineOslo University HospitalNydalenOsloNorway
| | - Jonathan Mill
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Clive Ballard
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| | - Robert A. Sweet
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Julia Kofler
- Department of PathologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Byron Creese
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
- Division of PsychologyDepartment of Life SciencesBrunel University LondonUxbridgeUK
| | - Ehsan Pishva
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs)Faculty of HealthMedicine and Life Sciences (FHML)Maastricht UniversityMaastrichtThe Netherlands
| | - Katie Lunnon
- Department of Clinical and Biomedical SciencesFaculty of Health and Life SciencesUniversity of ExeterExeterDevonUK
| |
Collapse
|
34
|
Choudhury M, Yamamoto R, Xiao X. Genetic architecture of RNA editing, splicing and gene expression in schizophrenia. Hum Mol Genet 2025; 34:277-290. [PMID: 39656777 PMCID: PMC11792240 DOI: 10.1093/hmg/ddae172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Genome wide association studies (GWAS) have been conducted over the past decades to investigate the underlying genetic origin of neuropsychiatric diseases, such as schizophrenia (SCZ). While these studies demonstrated the significance of disease-phenotype associations, there is a pressing need to fully characterize the functional relevance of disease-associated genetic variants. Functional genetic loci can affect transcriptional and post-transcriptional phenotypes that may contribute to disease pathology. Here, we investigate the associations between genetic variation and RNA editing, splicing, and overall gene expression through identification of quantitative trait loci (QTL) in the CommonMind Consortium SCZ cohort. We find that editing QTL (edQTL), splicing QTL (sQTL) and expression QTL (eQTL) possess both unique and common gene targets, which are involved in many disease-relevant pathways, including brain function and immune response. We identified two QTL that fall into all three QTL categories (seedQTL), one of which, rs146498205, targets the lincRNA gene, RP11-156P1.3. In addition, we observe that the RNA binding protein AKAP1, with known roles in neuronal regulation and mitochondrial function, had enriched binding sites among edQTL, including the seedQTL, rs146498205. We conduct colocalization with various brain disorders and find that all QTL have top colocalizations with SCZ and related neuropsychiatric diseases. Furthermore, we identify QTL within biologically relevant GWAS loci, such as in ELA2, an important tRNA processing gene associated with SCZ risk. This work presents the investigation of multiple QTL types in parallel and demonstrates how they target both distinct and overlapping SCZ-relevant genes and pathways.
Collapse
Affiliation(s)
- Mudra Choudhury
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, United States
| | - Ryo Yamamoto
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, United States
| | - Xinshu Xiao
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, United States
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 612 Charles E. Young Drive East, Box 957246, Los Angeles, CA 90095-7246, United States
- Molecular Biology Institute, University of California, Los Angeles, 611 Charles E. Young Drive East, Los Angeles, CA 90095-1570, United States
| |
Collapse
|
35
|
Yang W, Liu X, Fan D. Low CD3 level is a risk factor for amyotrophic lateral sclerosis: a Mendelian randomization study. Amyotroph Lateral Scler Frontotemporal Degener 2025; 26:64-72. [PMID: 39316061 DOI: 10.1080/21678421.2024.2407408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal disease characterized by neuronal degeneration of the spinal cord and brain and believed to be related to the immune system. In this study, our aim is to use Mendelian randomization (MR) to search for immune markers related to ALS. A total of 731 immune cell traits were included in this study. MR analysis was used to identify the causality between 731 immune cell traits (with 3,757 Europeans) and ALS (with 138,086 Europeans). Colocalization analysis was used to verify the found causality, protein-protein interaction prediction was used to look for the interacting proteins that are known to be involved in ALS. We found low expression levels of CD3 on central memory CD8+ T cell is risk factor for ALS (OR = 0.90, 95% CI: 0.86-0.95, P = 0.0000303). CD3 can interact with three ALS-related proteins: VCP, HLA-DRA and HLA-DRB5, which are associated with adaptive immune response. Our study reported for the first time that low-level CD3 is a risk factor for ALS and the possible mechanism, which could provide a potential strategy for ALS diagnosis and therapy.
Collapse
Affiliation(s)
- Wenzhi Yang
- Department of Neurology, Peking University Third Hospital Beijing, China and
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Xiangyi Liu
- Department of Neurology, Peking University Third Hospital Beijing, China and
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital Beijing, China and
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| |
Collapse
|
36
|
Burgess S, Cronjé HT, deGoma E, Chyung Y, Gill D. Human Genetic Evidence to Inform Clinical Development of IL-6 Signaling Inhibition for Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2025; 45:323-331. [PMID: 39633572 PMCID: PMC7617413 DOI: 10.1161/atvbaha.124.321988] [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: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) represents a significant cause of mortality, yet no medical therapies have proven efficacious. The aim of the current study was to leverage human genetic evidence to inform clinical development of IL-6 (interleukin-6) signaling inhibition for the treatment of AAA. METHODS Associations of rs2228145, a missense variant in the IL6R gene region, are expressed per additional copy of the C allele, corresponding to the genetically predicted effect of IL-6 signaling inhibition. We consider genetic associations with AAA risk in the AAAgen consortium (39 221 cases and 1 086 107 controls) and UK Biobank (1963 cases and 365 680 controls). To validate against known effects of IL-6 signaling inhibition, we present associations with rheumatoid arthritis, polymyalgia rheumatica, and severe COVID-19. To explore mechanism specificity, we present associations with thoracic aortic aneurysm, intracranial aneurysm, and coronary artery disease. We further explored genetic associations in clinically relevant subgroups of the population. RESULTS We observed strong genetic associations with AAA risk in the AAAgen consortium, UK Biobank, and FinnGen (odds ratios: 0.91 [95% CI, 0.90-0.92], P=4×10-30; 0.90 [95% CI, 0.84-0.96], P=0.001; and 0.86 [95% CI, 0.82-0.91], P=7×10-9, respectively). The association was similar for fatal AAA but with greater uncertainty due to the lower number of events. The association with AAA was of greater magnitude than associations with coronary artery disease and even rheumatological disorders for which IL-6 inhibitors have been approved. No strong associations were observed with thoracic aortic aneurysm or intracranial aneurysm. Associations attenuated toward the null in populations with concomitant rheumatological or connective tissue disease. CONCLUSIONS Inhibition of IL-6 signaling is a promising strategy for treating AAA but not other types of aneurysmal disease. These findings serve to help inform clinical development of IL-6 signaling inhibition for AAA treatment.
Collapse
Affiliation(s)
- Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
- Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
- Sequoia Genetics, London, United Kingdom
| | - Héléne T. Cronjé
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
- Sequoia Genetics, London, United Kingdom
| | | | | | | |
Collapse
|
37
|
Huang Y, Stinson SE, Thodberg M, Holm LA, Thielemann R, Sulek K, Lund MAV, Fonvig CE, Kim M, Trost K, Juel HB, Nielsen T, Rossing P, Thiele M, Krag A, Legido-Quigley C, Holm JC, Hansen T. Genetic factors shaping the plasma lipidome and the relations to cardiometabolic risk in children and adolescents. EBioMedicine 2025; 112:105537. [PMID: 39753034 PMCID: PMC11754075 DOI: 10.1016/j.ebiom.2024.105537] [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: 07/24/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Lipid species are emerging as biomarkers for cardiometabolic risk in both adults and children. The genetic regulation of lipid species and their impact on cardiometabolic risk during early life remain unexplored. METHODS Using mass spectrometry-based lipidomics, we measured 227 plasma lipid species in 1149 children and adolescents (44.8% boys) with a median age of 11.2 years. We performed genome-wide association analyses to identify genetic variants influencing lipid species. Colocalisation and Mendelian randomisation (MR) analyses were performed to infer causality between lipid species and cardiometabolic outcomes. FINDINGS We identified 37 genome-wide significant loci for 52 lipid species, nine of which are previously unreported. Colocalisation analyses revealed that seven lipid loci shared genetic variants associated with adult cardiometabolic outcomes. One-sample MR analysis identified positive causal associations between ceramides and liver enzymes, sphingomyelins and hemoglobin A1c (HbA1c), and phosphatidylethanolamines and high-sensitivity C-reactive protein in children and adolescents. Two-sample MR using adult-based summary statistics showed consistent direction of associations and indicated additional causal links, specifically between ceramides and elevated HbA1c levels, and phosphatidylinositols with elevated liver enzymes. INTERPRETATION These findings highlight the potential long-term implications of plasma lipid genetic determinants on cardiometabolic risk. FUNDING Novo Nordisk Foundation, The Innovation Fund Denmark, The Danish Heart Foundation, EU Horizon, and LundbeckFonden.
Collapse
Affiliation(s)
- Yun Huang
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Sara Elizabeth Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Malte Thodberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Denmark
| | - Roman Thielemann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | - Morten Asp Vonsild Lund
- The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Cilius Esmann Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Min Kim
- Steno Diabetes Center Copenhagen, Denmark
| | - Kajetan Trost
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Denmark
| | - Helene Bæk Juel
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Trine Nielsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Medical Department, Zeeland University Hospital, Køge, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Maja Thiele
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Denmark; Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Aleksander Krag
- Centre for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Denmark; Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Cristina Legido-Quigley
- Steno Diabetes Center Copenhagen, Denmark; Institute of Pharmaceutical Science, King's College London, United Kingdom
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
38
|
Lee MA, Burley KL, Hazelwood EL, Moore S, Lewis SJ, Goudswaard LJ. Exploring the role of circulating proteins in multiple myeloma risk: a Mendelian randomization study. Sci Rep 2025; 15:3752. [PMID: 39885253 PMCID: PMC11782597 DOI: 10.1038/s41598-025-86222-5] [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: 07/25/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025] Open
Abstract
Multiple myeloma (MM) is an incurable blood cancer with unclear aetiology. Proteomics is a valuable tool in exploring mechanisms of disease. We investigated the causal relationship between circulating proteins and MM risk, using two of the largest cohorts with proteomics data to-date. We performed bidirectional two-sample Mendelian randomization (MR; forward MR = causal effect estimation of proteins and MM risk; reverse MR = causal effect estimation of MM risk and proteins). Summary statistics for plasma proteins were obtained from genome-wide association studies performed using SomaLogic (N = 35,559; deCODE) and Olink (N = 34,557; UK Biobank; UKB) proteomic platforms and for MM risk from a meta-analysis of UKB and FinnGen (case = 1649; control = 727,247) or FinnGen only (case = 1085; control = 271,463). Cis-SNPs associated with protein levels were used to instrument circulating proteins. We evaluated proteins for the consistency of directions of effect across MR analyses (with 95% confidence intervals not overlapping the null) and corroborating evidence from genetic colocalization. In the forward MR, 994 (SomaLogic) and 1570 (Olink) proteins were instrumentable. 440 proteins were analysed in both deCODE and UKB; 302 (69%) of these showed consistent directions of effect in the forward MR. Seven proteins had 95% confidence intervals (CIs) that did not overlap the null in both forward MR analyses and did not have evidence for an effect in the reverse direction: higher levels of dermatopontin (DPT), beta-crystallin B1 (CRYBB1), interleukin-18-binding protein (IL18BP) and vascular endothelial growth factor receptor 2 (KDR) and lower levels of odorant-binding protein 2b (OBP2B), glutamate-cysteine ligase regulatory subunit (GCLM) and gamma-crystallin D (CRYGD) were implicated in increasing MM risk. Evidence from genetic colocalization did not meet our threshold for a shared causal signal between any of these proteins and MM risk (h4 < 0.8). Our results highlight seven circulating proteins which may be involved in MM risk. Although evidence from genetic colocalization suggests these associations may not be robust to the effects of horizontal pleiotropy, these proteins may be useful markers of MM risk. Future work should explore the utility of these proteins in disease prediction or prevention using proteomic data from patients with MM or precursor conditions.
Collapse
Affiliation(s)
- Matthew A Lee
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organisation, Lyon, France
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Kate L Burley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Emma L Hazelwood
- Population Health Sciences, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Sally Moore
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Sarah J Lewis
- Population Health Sciences, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Lucy J Goudswaard
- Population Health Sciences, University of Bristol, Bristol, UK.
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| |
Collapse
|
39
|
Guo H, Gao J, Gong L, Wang Y. Multi-omics analysis reveals novel causal pathways in psoriasis pathogenesis. J Transl Med 2025; 23:100. [PMID: 39844246 PMCID: PMC11752815 DOI: 10.1186/s12967-025-06099-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND To elucidate the genetic and molecular mechanisms underlying psoriasis by employing an integrative multi-omics approach, using summary-data-based Mendelian randomization (SMR) to infer causal relationships among DNA methylation, gene expression, and protein levels in relation to psoriasis risk. METHODS We conducted SMR analyses integrating genome-wide association study (GWAS) summary statistics with methylation quantitative trait loci (mQTL), expression quantitative trait loci (eQTL), and protein quantitative trait loci (pQTL) data. Publicly available datasets were utilized, including psoriasis GWAS data from the European Molecular Biology Laboratory-European Bioinformatics Institute and the UK Biobank. Heterogeneity in dependent instruments (HEIDI) test and colocalization analyses were performed to identify shared causal variants, and multi-omics integration was employed to construct potential regulatory pathways. RESULTS Our analyses identified significant causal associations between DNA methylation, gene expression, protein abundance, and psoriasis risk. We discovered two pathways involving the long non-coding RNA RP11-977G19.11 and apolipoprotein F (APOF). Methylation at sites cg26804944 and cg02705573 was negatively associated with RP11-977G19.11 expression. Reduced expression of RP11-977G19.11 was linked to increased APOF levels, which were positively associated with a higher risk of psoriasis. Methylation at sites cg00172967, cg00294382, and cg24773560 was positively associated with RP11-977G19.11 expression. Elevated expression of RP11-977G19.11 was associated with decreased APOF levels, reducing the risk of psoriasis. Colocalization analysis highlighted APOF as a key protein in psoriasis pathogenesis. Validation using skin tissue, EBV-transformed lymphocytes data and inflammation-related protein panels confirmed the associations of RP11-977G19.11 and APOF with psoriasis. CONCLUSIONS Our multi-omics analysis provides preliminary evidence for potential molecular mechanisms in psoriasis pathogenesis. Through the integration of GWAS and molecular QTL data, we identify candidate pathways that may be relevant to disease biology. While these findings require extensive experimental validation, they offer a framework for future investigations into the molecular basis of psoriasis.
Collapse
Affiliation(s)
- Hua Guo
- Department of Academic Research, The Second Hospital of Shandong University, Jinan, Shandong, China
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jinyang Gao
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Liping Gong
- Department of Academic Research, The Second Hospital of Shandong University, Jinan, Shandong, China.
| | - Yanqing Wang
- Department of Academic Research, The Second Hospital of Shandong University, Jinan, Shandong, China.
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
40
|
Göteson A, Holmén-Larsson J, Celik H, Pelanis A, Sellgren CM, Sparding T, Pålsson E, Zetterberg H, Blennow K, Jonsson L, Gobom J, Landén M. Mapping the Cerebrospinal Fluid Proteome in Bipolar Disorder. Biol Psychiatry 2025:S0006-3223(25)00029-0. [PMID: 39827936 DOI: 10.1016/j.biopsych.2025.01.007] [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: 07/18/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Bipolar disorder (BD) is a severe psychiatric condition with unclear etiology and no established biomarkers. Here, we aimed to characterize the cerebrospinal fluid (CSF) proteome in euthymic individuals with BD to identify potential protein biomarkers. METHODS We used nano-flow liquid chromatography coupled to high-resolution mass spectrometry to quantify over 2000 CSF proteins in 374 individuals from two independent clinical cohorts (n = 164 cases + 89 controls and 66 cases + 55 controls, respectively). A subset of the cases was followed longitudinally and reexamined after a median of 6.5 years. RESULTS Differential abundance analysis revealed 41 proteins with robust case-control association in both cohorts. These included lower levels of synaptic proteins (e.g., APP, CLSTN1, NPTX2, NRXN1) and axon guidance and cell adhesion molecules (e.g., NEO1, NCAM1, SEMA7A) and higher levels of blood-brain barrier integrity proteins (e.g., VTN, SERPIN3) and complement components (e.g., C1RL, C3, C5). The findings were consistently driven by the BD type 1 subtype. Comparing BD type 1 participants with control participants increased discoverability, revealing 86 replicated associations despite a loss of statistical power. Moreover, longitudinal analyses of coexpression modules revealed dynamic changes in the CSF proteome composition that correlated with clinical outcomes, including disease severity, future manic episodes, and symptom improvement. Finally, we conducted association analyses of CSF proteins with genetic risk loci for BD and schizophrenia. CONCLUSIONS This study represents the first large-scale untargeted profiling of the CSF proteome in BD, unveiling potential biomarkers and providing in vivo support for altered synaptic and brain connectivity processes, impaired neurovascular integrity, and complement activation in the pathology of BD.
Collapse
Affiliation(s)
- Andreas Göteson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Jessica Holmén-Larsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Hatice Celik
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Aurimantas Pelanis
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Sweden
| | - Timea Sparding
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Erik Pålsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Dementia Research Centre, University College London Institute of Neurology, London, United Kingdom; UK Dementia Research Institute, University College London, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute of Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of University of Science and Technology, Hefei, China
| | - Lina Jonsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Mikael Landén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Chen Y, Zhang Z, Chen Y, Liu P, Yi S, Fan C, Zhao W, Liu J. Investigating the shared genetic links between hypothyroidism and psychiatric disorders: a large-scale genomewide cross-trait analysis. J Affect Disord 2025; 369:312-320. [PMID: 39353512 DOI: 10.1016/j.jad.2024.08.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 07/17/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Associations between thyroid diseases and psychiatric disorders have been mainly described before. However, the genetic mechanism behind hypothyroidism and psychiatric disorders remains unexplained. METHODS We examined the genetic architecture of hypothyroidism and 8 psychiatric disorders. Firstly, the global and local genetic relationship between the paired traits was explored. Secondly, cross-trait analysis was performed to investigate the genomic loci and genes between psychiatric disorders and hypothyroidism. Thirdly, the significant expression of these genes and the causal relationships were investigated. Lastly, enrichment analysis was conducted on these genes to explore their biological mechanisms. RESULTS We observed significant positive genetic correlations between psychiatric disorders and hypothyroidism. The cross-trait meta-analysis identified 62 shared genetic loci between hypothyroidism and psychiatric disorders. The colocalization analysis additionally revealed 15 potential pleiotropic loci with a posterior probabilities.H4 (PP·H4) value >0.7. We also found 2308 genes shared between both traits, which were highly enriched in biological pathways such as immune cell differentiation and autoimmune diseases, as well as in tissue structures like the frontal cortex and cerebral cortex. Especially, many pleiotropic genes were significantly expressed for multiple pairwise traits, such as BCL11B, RERE, and SUOX. Lastly, the Latent causal variable model (LCV) analysis did not find any causal components in the genetic structure between them. LIMITATIONS The limitations of this study include that the conclusions were drawn from a European population. CONCLUSIONS These findings not only deepens our understanding of their biological mechanisms but also has significant implications for the intervention and treatment of these diseases.
Collapse
Affiliation(s)
- Yanjing Chen
- Department of Radiology, Second Xiangya Hospital, Central South University, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China.
| | - Zhiyi Zhang
- Fujian University of Traditional Chinese Medicine, 1#, Qiuyang Road, Fuzhou, Fujian Province 350122, People's Republic of China.
| | - Yongyi Chen
- Clinical Research Center for Medical Imaging in Hunan Province, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China.
| | - Ping Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China.
| | - Sijie Yi
- Department of Radiology, Second Xiangya Hospital, Central South University, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China.
| | - Chunhua Fan
- Department of Radiology, Second Xiangya Hospital, Central South University, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China.
| | - Wei Zhao
- Department of Radiology, Second Xiangya Hospital, Central South University, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China; Clinical Research Center for Medical Imaging in Hunan Province, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China.
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China; Clinical Research Center for Medical Imaging in Hunan Province, 139#, Central Renmin Road, Changsha, Hunan Province 410011, People's Republic of China.
| |
Collapse
|
42
|
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
|
43
|
Downie CG, Highland HM, Alotaibi M, Welch BM, Howard AG, Cheng S, Miller N, Jain M, Kaplan RC, Lilly AG, Long T, Sofer T, Thyagarajan B, Yu B, North KE, Avery CL. Genome-wide association study reveals shared and distinct genetic architecture of fatty acids and oxylipins in the Hispanic Community Health Study/Study of Latinos. HGG ADVANCES 2025; 6:100390. [PMID: 39644095 PMCID: PMC11751521 DOI: 10.1016/j.xhgg.2024.100390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
Bioactive fatty acid-derived oxylipin molecules play key roles mediating inflammation and oxidative stress. Circulating levels of fatty acids and oxylipins are influenced by environmental and genetic factors; characterizing the genetic architecture of bioactive lipids could yield new insights into underlying biology. We performed a genome-wide association study (GWAS) of 81 fatty acids and oxylipins in 11,584 Hispanic Community Health Study/Study of Latinos (HCHS/SOL) participants with genetic and lipidomic data measured at study baseline (58.6% female, mean age = 46.1 years (standard deviation 13.8)). Additionally, given the effects of central obesity on inflammation, we examined interactions with waist circumference using two-degree-of-freedom joint tests. Thirty-three of the 81 oxylipins and fatty acids were significantly heritable (heritability range: 0-32.7%). Forty (49.4%) oxylipins and fatty acids had at least one genome-wide significant (p < 6.94E-11) variant resulting in 19 independent genetic loci. Six loci (lead variant minor allele frequency [MAF] range: 0.08-0.50), including desaturase-encoding FADS and OATP1B1 transporter protein-encoding SLCO1B1, exhibited associations with two or more fatty acids and oxylipins. At several of these loci, there was evidence of colocalization of the top variant across fatty acids and oxylipins. The remaining loci were only associated with one oxylipin or fatty acid and included several CYP loci. We also identified an additional rare variant (MAF = 0.002) near CARS2 in two-degree-of-freedom tests. Our analyses revealed shared and distinct genetic architecture underlying fatty acids and oxylipins, providing insights into genetic factors and motivating work to characterize these compounds and elucidate their roles in disease.
Collapse
Affiliation(s)
- Carolina G Downie
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Heather M Highland
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mona Alotaibi
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, San Diego, CA, USA
| | - Barrett M Welch
- School of Public Health, University of Nevada, Reno, Reno, NV, USA
| | - Annie Green Howard
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Mohit Jain
- Sapient Bioanalytics, San Diego, CA, USA; Departments of Medicine and Pharmacology, University of California, San Diego, San Diego, CA, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA; Public Health Sciences Division, Fred Hutchison Cancer Center, Seattle, WA, USA
| | - Adam G Lilly
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tao Long
- Sapient Bioanalytics, San Diego, CA, USA
| | - Tamar Sofer
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas Health Science Center at Houston, School of Public Health, Houston, TX, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christy L Avery
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
44
|
Liu P, Xiao J, Xiao J, Zhou J. Metformin and the risk of malignant tumors of digestive system: a mendelian randomization study. Diabetol Metab Syndr 2025; 17:6. [PMID: 39773528 PMCID: PMC11705776 DOI: 10.1186/s13098-024-01573-9] [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: 09/26/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Observational studies suggest that metformin may reduce the risk of malignant tumors of the digestive system (MTDS), but these findings are often confounded by bias and unmeasured variables. Recent meta-analyses have questioned these associations, emphasizing the need for robust causal inference. METHODS Mendelian randomization (MR) was used to evaluate the causal relationship between metformin and MTDS. Genetic variants associated with metformin's molecular targets were selected from GTEx, eQTLGen, and UK Biobank and validated using genetic colocalization to ensure instrument validity. GWAS summary statistics for MTDS, encompassing up to 314,193 controls and 6,847 colorectal cancer cases, were obtained from FinnGen and EBI. The primary analysis employed the inverse-variance weighted (IVW) method, supplemented by MR-Egger, weighted median, and weighted mode analyses. Bonferroni correction was applied to adjust for multiple testing across 14 cancer types. RESULTS Genetically proxied metformin use was associated with an increased risk of colorectal cancer (OR = 2.38, 95%CI = 1.38-4.09, P = 0.0018) and related subtypes. No causal relationship was found for hepatocellular carcinoma, gastric cancer, pancreatic cancer, or other digestive system cancers. The robustness of these findings was supported by sensitivity analyses, which indicated no significant pleiotropy, and leave-one-out tests. CONCLUSION This study provides robust genetic evidence that metformin use increases the risk of colorectal cancer, challenging its role as a preventive agent for digestive cancers. These findings emphasize the need for clinicians to carefully evaluate the risks and benefits of metformin, particularly in populations at higher risk for colorectal cancer. Future research should focus on delineating the mechanisms underlying this association to optimize the use of metformin in clinical practice.
Collapse
Affiliation(s)
- Ping Liu
- Department of Radiation Oncology and Hunan Key Laboratory of Translational Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, HunanCancer Hospital, Changsha, China
| | - Junqi Xiao
- Cancer center, The Second Affiliated Hospital, Hengyang Medical School, University of South China, No.35 of Jiefang Avenue, Hengyang City, P. R. China
| | - Jinghuang Xiao
- Department of Radiation Oncology and Hunan Key Laboratory of Translational Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, HunanCancer Hospital, Changsha, China
| | - Jumei Zhou
- Department of Radiation Oncology and Hunan Key Laboratory of Translational Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, HunanCancer Hospital, Changsha, China.
| |
Collapse
|
45
|
Xing X, Xu S, Wang Y, Shen Z, Wen S, Zhang Y, Ruan G, Cai G. Evaluating the Causal Effect of Circulating Proteome on Glycemic Traits: Evidence From Mendelian Randomization. Diabetes 2025; 74:108-119. [PMID: 39418314 DOI: 10.2337/db24-0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Exploring the mechanisms underlying abnormal glycemic traits is important for deciphering type 2 diabetes and characterizing novel drug targets. This study aimed to decipher the causal associations of circulating proteins with fasting glucose (FG), 2-h glucose after an oral glucose challenge (2hGlu), fasting insulin (FI), and glycated hemoglobin (HbA1c) using large-scale proteome-wide Mendelian randomization (MR) analyses. Genetic data on plasma proteomes were obtained from 10 proteomic genome-wide association studies. Both cis-protein quantitative trait loci (pQTLs) and cis + trans-pQTLs MR analyses were conducted. Bayesian colocalization, Steiger filtering analysis, assessment of protein-altering variants, and mapping expression QTLs to pQTLs were performed to investigate the reliability of the MR findings. Protein-protein interaction, pathway enrichment analysis, and evaluation of drug targets were performed. Thirty-three proteins were identified with causal effects on FG, FI, or HbA1c but not 2hGlu in the cis-pQTL analysis, and 93 proteins had causal effects on glycemic traits in the cis + trans-pQTLs analysis. Most proteins were either considered druggable or drug targets. In conclusion, many novel circulating protein biomarkers were identified to be causally associated with glycemic traits. These biomarkers enhance the understanding of molecular etiology and provide insights into the screening, monitoring, and treatment of diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Xing Xing
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Siqi Xu
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yining Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Ziyuan Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Simin Wen
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangfeng Ruan
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Guoqi Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
46
|
Brotman SM, El-Sayed Moustafa JS, Guan L, Broadaway KA, Wang D, Jackson AU, Welch R, Currin KW, Tomlinson M, Vadlamudi S, Stringham HM, Roberts AL, Lakka TA, Oravilahti A, Fernandes Silva L, Narisu N, Erdos MR, Yan T, Bonnycastle LL, Raulerson CK, Raza Y, Yan X, Parker SCJ, Kuusisto J, Pajukanta P, Tuomilehto J, Collins FS, Boehnke M, Love MI, Koistinen HA, Laakso M, Mohlke KL, Small KS, Scott LJ. Adipose tissue eQTL meta-analysis highlights the contribution of allelic heterogeneity to gene expression regulation and cardiometabolic traits. Nat Genet 2025; 57:180-192. [PMID: 39747594 DOI: 10.1038/s41588-024-01982-6] [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: 09/21/2023] [Accepted: 10/11/2024] [Indexed: 01/04/2025]
Abstract
Complete characterization of the genetic effects on gene expression is needed to elucidate tissue biology and the etiology of complex traits. In the present study, we analyzed 2,344 subcutaneous adipose tissue samples and identified 34,774 conditionally distinct expression quantitative trait locus (eQTL) signals at 18,476 genes. Over half of eQTL genes exhibited at least two eQTL signals. Compared with primary eQTL signals, nonprimary eQTL signals had lower effect sizes, lower minor allele frequencies and less promoter enrichment; they corresponded to genes with higher heritability and higher tolerance for loss of function. Colocalization of eQTLs with genome-wide association study (GWAS) signals for 28 cardiometabolic traits identified 1,835 genes. Inclusion of nonprimary eQTL signals increased discovery of colocalized GWAS-eQTL signals by 46%. Furthermore, 21 genes with ≥2 colocalized GWAS-eQTL signals showed a mediating gene dosage effect on the GWAS trait. Thus, expanded eQTL identification reveals more mechanisms underlying complex traits and improves understanding of the complexity of gene expression regulation.
Collapse
Affiliation(s)
- Sarah M Brotman
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | - Li Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dongmeng Wang
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Kevin W Currin
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Max Tomlinson
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | | | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Amy L Roberts
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lori L Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Yasrab Raza
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Xinyu Yan
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Stephen C J Parker
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Kuusisto
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics and Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Heikki A Koistinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki and Department of Medicine, Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Ding K, Qin X, Wang H, Wang K, Kang X, Yu Y, Liu Y, Gong H, Wu T, Chen D, Hu Y, Wang T, Wu Y. Identification of shared genetic etiology of cardiovascular and cerebrovascular diseases through common cardiometabolic risk factors. Commun Biol 2024; 7:1703. [PMID: 39730871 DOI: 10.1038/s42003-024-07417-6] [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: 06/14/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) and cerebrovascular diseases (CeVDs) are closely related vascular diseases, sharing common cardiometabolic risk factors (RFs). Although pleiotropic genetic variants of these two diseases have been reported, their underlying pathological mechanisms are still unclear. Leveraging GWAS summary data and using genetic correlation, pleiotropic variants identification, and colocalization analyses, we identified 11 colocalized loci for CVDs-CeVDs-BP (blood pressure), CVDs-CeVDs-LIP (lipid traits), and CVDs-CeVDs-cIMT (carotid intima-media thickness) triplets. No shared causal loci were found for CVDs-CeVDs-T2D (type 2 diabetes) or CVDs-CeVDs-BMI (body mass index) triplets. The 11 loci were mapped to 12 genes, namely CASZ1, CDKN1A, TWIST1, CDKN2B, ABO, SWAP70, SH2B3, LRCH1, FES, GOSR2, RPRML, and LDLR, where both GOSR2 and RPRML were mapped to one locus. They were enriched in pathways related to cellular response to external stimulus and regulation of the phosphate metabolic process and were highly expressed in endothelial cells, epithelial cells, and smooth muscle cells. Multi-omics analysis revealed methylation of two genes (CASZ1 and LRCH1) may play a causal role in the genetic pleiotropy. Notably, these pleiotropic loci are highly enriched in the targets of antihypertensive drugs, which further emphasizes the role of the blood pressure regulation pathway in the shared etiology of CVDs and CeVDs.
Collapse
Affiliation(s)
- Kexin Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| | - Huairong Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Kun Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xiaoying Kang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Yao Yu
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Haiying Gong
- Fangshan District Center for Disease Control and Prevention, Beijing, China
| | - Tao Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dafang Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Yonghua Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Tao Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yiqun Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| |
Collapse
|
48
|
Bian R, Li D, Xu X, Zhang L. The impact of immunity on the risk of coronary artery disease: insights from a multiomics study. Postgrad Med J 2024; 101:50-59. [PMID: 39180487 DOI: 10.1093/postmj/qgae105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/16/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Immune inflammation is intricately associated with coronary artery disease (CAD) progression, necessitating the pursuit of more efficacious therapeutic strategies. This study aimed to uncover potential therapeutic targets for CAD and myocardial infarction (MI) by elucidating the causal connection between regulatory immune-related genes (RIRGs) and these disorders. METHODOLOGY We performed summary data-based Mendelian randomization analysis to assess the therapeutic targets linked to expression quantitative trait loci and methylation quantitative trait loci of RIRGs in relation to CAD and MI. Independent validation cohorts and datasets from coronary artery and left ventricular heart tissue were analyzed. To strengthen causal inference, colocalization analysis and PhenoScanner phenotype scans were employed. RESULTS Utilizing multiomics integration, we pinpointed EIF2B2, FCHO1, and DDT as CAD risk genes. Notably, EIF2B2 and FCHO1 displayed significant associations with MI. High EIF2B2 expression, regulated by cg16144293, heightened CAD and MI risk at rs175438. In contrast, enhanced FCHO1 expression, modulated by cg18329931, reduced CAD and MI risk at rs13382133. DDT upregulation influenced by cg11060661 and cg09664220 was associated with decreased CAD risk at rs5760120. Colocalization analysis firmly established these relationships. CONCLUSION EIF2B2, FCHO1, and DDT represent risk loci for CAD progression within RIRGs. Our identification of these genes enhances understanding of CAD pathogenesis and directs future drug development efforts.
Collapse
Affiliation(s)
- Rutao Bian
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| | - Dongyu Li
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| | - Xuegong Xu
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| | - Li Zhang
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450000, China
- The Affiliated Zhengzhou Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450000, China
| |
Collapse
|
49
|
Khullar S, Huang X, Ramesh R, Svaren J, Wang D. NetREm: Network Regression Embeddings reveal cell-type transcription factor coordination for gene regulation. BIOINFORMATICS ADVANCES 2024; 5:vbae206. [PMID: 40260118 PMCID: PMC12011367 DOI: 10.1093/bioadv/vbae206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/22/2024] [Accepted: 12/18/2024] [Indexed: 04/23/2025]
Abstract
Motivation Transcription factor (TF) coordination plays a key role in gene regulation via direct and/or indirect protein-protein interactions (PPIs) and co-binding to regulatory elements on DNA. Single-cell technologies facilitate gene expression measurement for individual cells and cell-type identification, yet the connection between TF-TF coordination and target gene (TG) regulation of various cell types remains unclear. Results To address this, we introduce our innovative computational approach, Network Regression Embeddings (NetREm), to reveal cell-type TF-TF coordination activities for TG regulation. NetREm leverages network-constrained regularization, using prior knowledge of PPIs among TFs, to analyze single-cell gene expression data, uncovering cell-type coordinating TFs and identifying revolutionary TF-TG candidate regulatory network links. NetREm's performance is validated using simulation studies and benchmarked across several datasets in humans, mice, yeast. Further, we showcase NetREm's ability to prioritize valid novel human TF-TF coordination links in 9 peripheral blood mononuclear and 42 immune cell sub-types. We apply NetREm to examine cell-type networks in central and peripheral nerve systems (e.g. neuronal, glial, Schwann cells) and in Alzheimer's disease versus Controls. Top predictions are validated with experimental data from rat, mouse, and human models. Additional functional genomics data helps link genetic variants to our TF-TG regulatory and TF-TF coordination networks. Availability and implementation https://github.com/SaniyaKhullar/NetREm.
Collapse
Affiliation(s)
- Saniya Khullar
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53076, United States
| | - Xiang Huang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Raghu Ramesh
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Comparative Biomedical Sciences Training Program, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53076, United States
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI 53706, United States
| |
Collapse
|
50
|
Salenius K, Väljä N, Thusberg S, Iris F, Ladd-Acosta C, Roos C, Nykter M, Fasano A, Autio R, Lin J. Exploring autism spectrum disorder and co-occurring trait associations to elucidate multivariate genetic mechanisms and insights. BMC Psychiatry 2024; 24:934. [PMID: 39696186 DOI: 10.1186/s12888-024-06392-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a partially heritable neurodevelopmental trait, and people with ASD may also have other co-occurring trait such as ADHD, anxiety disorders, depression, mental health issues, learning difficulty, physical health traits and communication challenges. The concomitant development of ASD and other neurological traits is assumed to result from a complex interplay between genetics and the environment. However, only a limited number of studies have performed multivariate genome-wide association studies (GWAS) for ASD. METHODS We conducted to-date the largest multivariate GWAS on ASD and 8 ASD co-occurring traits (ADHD, ADHD childhood, anxiety stress (ASDR), bipolar (BIP), disruptive behaviour (DBD), educational attainment (EA), major depression, and schizophrenia (SCZ)) using summary statistics from leading studies. Multivariate associations and central traits were further identified. Subsequently, colocalization and Mendelian randomization (MR) analysis were performed on the associations identified with the central traits containing ASD. To further validate our findings, pathway and quantified trait loci (QTL) resources as well as independent datasets consisting of 112 (45 probands) whole genome sequence data from the GEMMA project were utilized. RESULTS Multivariate GWAS resulted in 637 significant associations (p < 5e-8), among which 322 are reported for the first time for any trait. 37 SNPs were identified to contain ASD and one or more traits in their central trait set, including variants mapped to known SFARI ASD genes MAPT, CADPS and NEGR1 as well as novel ASD genes KANSL1, NSF and NTM, associated with immune response, synaptic transmission, and neurite growth respectively. Mendelian randomization analyses found that genetic liability for ADHD childhood, ASRD and DBT has causal effects on the risk of ASD while genetic liability for ASD has causal effects on the risk of ADHD, ADHD childhood, BIP, WA, MDD and SCZ. Frequency differences of SNPs found in NTM and CADPS genes, respectively associated with neurite growth and neural/endocrine calcium regulation, were found between GEMMA ASD probands and controls. Pathway, QTL and cell type enrichment implicated microbiome, enteric inflammation, and central nervous system enrichments. CONCLUSIONS Our study, combining multivariate GWAS with systematic decomposition, identified novel genetic associations related to ASD and ASD co-occurring driver traits. Statistical tests were applied to discern evidence for shared and interpretable liability between ASD and co-occurring traits. These findings expand upon the current understanding of the complex genetics regulating ASD and reveal insights of neuronal brain disruptions potentially driving development and manifestation.
Collapse
Affiliation(s)
- Karoliina Salenius
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Niina Väljä
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Sini Thusberg
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | | | - Christine Ladd-Acosta
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | | | - Matti Nykter
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Alessio Fasano
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
- Harvard Medical School, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Reija Autio
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Jake Lin
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|