1
|
Watts LM, Sparkes PC, Dewhurst HF, Guilfoyle SE, Pollard AS, Komla-Ebri D, Butterfield NC, Williams GR, Bassett JHD. The GWAS candidate far upstream element binding protein 3 (FUBP3) is required for normal skeletal growth, and adult bone mass and strength in mice. Bone 2025; 195:117472. [PMID: 40139337 DOI: 10.1016/j.bone.2025.117472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Bone mineral density (BMD) and height are highly heritable traits for which hundreds of genetic loci have been linked through genome wide association studies (GWAS). FUBP3 is a DNA and RNA binding protein best characterised as a transcriptional regulator of c-Myc, but little is known about its role in vivo. Single nucleotide polymorphisms in FUBP3 at the 9q34.11 locus have been associated with BMD, fracture and height in multiple GWAS, but FUBP3 has no previously established role in the skeleton. We analysed Fubp3-deficient mice to determine the consequence of FUBP3 deficiency in vivo. Mice lacking Fubp3 had reduced survival to adulthood and impaired skeletal growth. Bone mass was decreased, most strikingly in the vertebrae, with altered trabecular micro-architecture. Fubp3 deficient bones were also weak. These data provide the first functional demonstration that Fubp3 is required for normal skeletal growth and development and maintenance of adult bone structure and strength, indicating that FUBP3 contributes to the GWAS association of 9q34.11 with variation in height, BMD and fracture.
Collapse
Affiliation(s)
- Laura M Watts
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Penny C Sparkes
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Hannah F Dewhurst
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Siobhan E Guilfoyle
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Andrea S Pollard
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Davide Komla-Ebri
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Natalie C Butterfield
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
2
|
Zhou Y, Duan J, Zhu J, Huang Y, Zhou J, Li F, Tu T, Lin Q, Ma Y, Liu Q. Separating the effects of childhood and adult obesity on depression, subjective well-being, and suicide attempt: a Mendelian randomization study. Eur Arch Psychiatry Clin Neurosci 2025:10.1007/s00406-025-02009-9. [PMID: 40338309 DOI: 10.1007/s00406-025-02009-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/21/2025] [Indexed: 05/09/2025]
Abstract
Observational studies have linked obesity, both in childhood and adulthood, with higher risks of depression, reduced subjective well-being (SWB), and suicide attempts (SA). However, the causality remains unclear. This study aimed to investigate the causal effects of childhood and adult obesity on depression, SWB, and SA. A bidirectional two-sample Mendelian randomization (MR) was performed using genome-wide association study (GWAS) data to examine the causal effects of body mass index (BMI) on depression, SWB, and SA. The inverse variance weighted method was used for primary analysis. Univariable and multivariable MR were employed to assess the total and independent effects of early life and adult body size. Cochran's Q test and MR-Egger intercept were applied to evaluate heterogeneity and pleiotropy. Genetically predicted BMI was significantly associated with an increased risk of major depressive disorder (MDD: OR = 1.13, 95%CI = 1.06-1.22, p = 6.1 × 10⁻⁴), SA-ISGC (OR = 1.17, 95%CI = 1.08-1.27, p = 1.9 × 10⁻⁴), and SA-iPSYCH (OR = 1.31, 95%CI = 1.12-1.54, p = 6.2 × 10⁻⁴). No significant causal effects of MDD, SWB, or SA on BMI were found. Early-life body size showed no direct effect on MDD or SA. However, adult body size was directly linked to increased risks of MDD (OR = 1.32, 95%CI = 1.13-1.55, p = 4.7 × 10⁻⁴), SA-ISGC (OR = 1.24, 95%CI = 1.03-1.47, p = 0.022), and SA-iPSYCH (OR = 1.80, 95%CI = 1.29-2.50, p = 5.6 × 10⁻⁴). This study provides robust evidence supporting a causal link between obesity and an increased risk of both depression and SA, with adult body size exerting a more direct impact on these outcomes than early-life body size.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China
| | - Jiayue Duan
- Department of Endocrinology, Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Ministry of Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Shuaifuyuan No.1, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Jiayi Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China
| | - Yunying Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China
| | - Jiabao Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China
| | - Fanqi Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China
| | - Tao Tu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China
| | - Yingxu Ma
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China.
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, 410011, China.
| |
Collapse
|
3
|
Xu Y, Dong XX, Wang Y, Zhuang XY, Chen YJ, Zhang XF, Pan CW. Association Between Inflammatory Cytokines and Refractive Errors: A Bidirectional Mendelian Randomization Study. Transl Vis Sci Technol 2025; 14:1. [PMID: 40310638 PMCID: PMC12054658 DOI: 10.1167/tvst.14.5.1] [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: 12/05/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose This investigation aimed to elucidate the causal role of inflammatory cytokines in the risk of developing refractive errors. Methods Genetic variants previously associated with inflammatory cytokines served as instrumental variables in genome-wide association studies (GWASs) of European ancestry. Bidirectional two-sample Mendelian randomization (MR) analyses were conducted using summary data from GWAS meta-analyses. Rigorous sensitivity analyses were performed to validate the reliability of the MR results. Results We found that, for every unit increase in interleukin 1 receptor antagonist (IL1RA) and interleukin 2 (IL2), there was a corresponding decrease in the prevalence of myopic refractive errors by 0.235 (95% confidence interval [CI], 0.050-0.419 for fixed effects; 95% CI, 0.125-0.345 for random effects) and 0.132 (95% CI, 0.032-0.231 for fixed effects; 95% CI, 0.044-0.220 for random effects), respectively. No substantial causal associations were observed for IL1α, IL1β, IL12p70, or monocyte chemoattractant protein 1 (MCP1) with refractive errors. Conversely, reverse MR analyses failed to indicate a causal influence of refractive errors on IL1RA and IL2. Conclusions The present study offers evidence for a causal link between inflammatory cytokines and refractive errors, which could have significant implications for the early detection, surveillance, and management of refractive errors. Translational Relevance Our study underscores the importance of IL1RA and IL2 in the prevention and management of refractive errors, suggesting the feasibility of strategies for early identification, continuous surveillance, and the deployment of focused therapeutic approaches.
Collapse
Affiliation(s)
- Yue Xu
- Department of Ophthalmology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xing-Xuan Dong
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yun Wang
- Department of Ophthalmology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin-Yu Zhuang
- Department of Ophthalmology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying-Jie Chen
- Department of Ophthalmology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Feng Zhang
- Department of Ophthalmology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Chen-Wei Pan
- Department of Ophthalmology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
- School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Wang L, Hu L, Sun J, Zhao J, Zhou S, Liu L, Yu W, Hu Y, Zhou D, Meng X, Yuan Z, Zhang H, Farrington S, Timofeeva M, Ding K, Little J, Dunlop M, Theodoratou E, Li X. Trans-ancestry transcriptome-wide association and functional studies to uncover novel susceptibility genes and therapeutic targets for colorectal cancer. NPJ Precis Oncol 2025; 9:124. [PMID: 40301637 PMCID: PMC12041606 DOI: 10.1038/s41698-025-00906-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/07/2025] [Indexed: 05/01/2025] Open
Abstract
By integrating findings from large-scale omics analyses with experimental tests, this study aims to decipher susceptibility genes and the underlying biological mechanisms involved in the development of colorectal cancer (CRC). We first conducted a trans-ancestry transcriptome-wide association study (TWAS) among 57,402 CRC cases and 119,110 controls, aiming to examine how altered gene expression influences CRC risk in European and Asian populations. Then, functional experiments in (i) CRC cell lines and (ii) tumor xenografts were conducted to examine potential underlying mechanisms involved in colorectal carcinogenesis. Further, a drug sensitivity test was employed to explore possible clinical implications for CRC treatment. The TWAS identified 67 genes highly associated with CRC risk, 23 of which were novel findings. Functional annotation of variants within TWAS-identified loci revealed that the majority (93.6%) showed evidence of transcriptional regulatory mechanisms via proximal promoter or distal enhancer-promoter interactions. Among the identified susceptibility genes, splicing factor 3a subunit 3 (SF3A3) may act as an oncogene on the basis that overexpression of this gene was significantly associated with increased risk of CRC (P = 5.75 × 10-11). Further cell and animal experiments confirmed that SF3A3 plays an oncogenic role in CRC development, and the underlying biological mechanism is likely to be related to its anti-apoptosis effect. The drug sensitivity test suggested that phenethyl isothiocyanate (PEITC) targeting SF3A3 can inhibit CRC progression. This study identified novel CRC susceptibility genes and potential biological mechanisms of SF3A3 involved in CRC development, providing important insight into the etiology and potential leads to the treatment of CRC.
Collapse
Affiliation(s)
- Lijuan Wang
- School of Public Health, the Second affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Lidan Hu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jing Sun
- School of Public Health, the Second affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhui Zhao
- School of Public Health, the Second affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siyun Zhou
- School of Public Health, the Second affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lexin Liu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wei Yu
- Department of Nephrology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yeting Hu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhou
- School of Public Health, the Second affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangrui Meng
- Division of Psychiatry, University College of London, London, UK
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Honghe Zhang
- Department of Pathology and Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Susan Farrington
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Maria Timofeeva
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Danish Institute for Advanced Study (DIAS), Epidemiology, Biostatistics and Biodemography Research Unit, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for CANCER, Hangzhou, China
| | - Julian Little
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Malcolm Dunlop
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Xue Li
- School of Public Health, the Second affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Kolligundla LP, Sullivan KM, Mukhi D, Andrade-Silva M, Liu H, Guan Y, Gu X, Wu J, Doke T, Hirohama D, Guarnieri P, Hill J, Pullen SS, Kuo J, Inamoto M, Susztak K. Glutathione-specific gamma-glutamylcyclotransferase 1 ( CHAC1) increases kidney disease risk by modulating ferroptosis. Sci Transl Med 2025; 17:eadn3079. [PMID: 40267214 DOI: 10.1126/scitranslmed.adn3079] [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: 12/03/2023] [Revised: 08/20/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association studies (GWASs) have identified more than 1000 loci where genetic variants correlate with kidney function. However, the specific genes, cell types, and mechanisms influenced by these genetic variants remain largely uncharted. Here, we identified glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) on chromosome 15 as affected by GWAS variants by analyzing human kidney gene expression and methylation information. Both CHAC1 RNA and protein were expressed in the loop of Henle region in mouse and human kidneys, and CHAC1 expression was higher in patients carrying disease risk variants. Using CRISPR technology, we created mice with a single functional copy of the Chac1 gene (Chac1+/-) that displayed no baseline phenotypic alterations in kidney structure or function. These mice demonstrated resilience to kidney disease in multiple models, including folic acid-induced nephropathy, adenine-induced chronic kidney disease, and uninephrectomy-streptozotocin-induced diabetic nephropathy. We further showed that CHAC1 plays a critical role in degrading the cellular antioxidant glutathione. Tubule cells isolated from Chac1+/- mice showed increased glutathione, decreased lipid peroxidation, improved cell viability, and protection against ferroptosis. Expression of ferroptosis-associated genes was also lower in mice with only one copy of Chac1. Higher CHAC1 protein also correlated with ferroptosis-related protein abundance in kidney biopsies from patients with kidney disease. This study positions CHAC1 as an important mediator of kidney disease that influences glutathione concentrations and ferroptosis, suggesting potential avenues to explore for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Lakshmi P Kolligundla
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Katie M Sullivan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Pediatrics, Medical College of Wisconsin Pediatric Nephrology, Milwaukee, WI 53226, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Magaiver Andrade-Silva
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Yuting Guan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Xiangchen Gu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Junnan Wu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Daigoro Hirohama
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jon Hill
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jay Kuo
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | | | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| |
Collapse
|
6
|
Lorincz-Comi N, Yang Y, Ajayakumar J, Mews M, Bermudez V, Bush W, Zhu X. HORNET: tools to find genes with causal evidence and their regulatory networks using eQTLs. BIOINFORMATICS ADVANCES 2025; 5:vbaf068. [PMID: 40270926 PMCID: PMC12014422 DOI: 10.1093/bioadv/vbaf068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/17/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Motivation Nearly two decades of genome-wide association studies (GWAS) have identify thousands of disease-associated genetic variants, but very few genes with evidence of causality. Recent methodological advances demonstrate that Mendelian randomization (MR) using expression quantitative loci (eQTLs) as instrumental variables can detect potential causal genes. However, existing MR approaches are not well suited to handle the complexity of eQTL GWAS data structure and so they are subject to bias, inflation, and incorrect inference. Results We present a whole-genome regulatory network analysis tool (HORNET), which is a comprehensive set of statistical and computational tools to perform genome-wide searches for causal genes using summary level GWAS data, i.e. robust to biases from multiple sources. Applying HORNET to schizophrenia, eQTL effects in the cerebellum were spread throughout the genome, and in the cortex were more localized to select loci. Availability and implementation Freely available at https://github.com/noahlorinczcomi/HORNET or Mac, Windows, and Linux users.
Collapse
Affiliation(s)
- Noah Lorincz-Comi
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Yihe Yang
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Jayakrishnan Ajayakumar
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Makaela Mews
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Valentina Bermudez
- Case Western Reserve University Department of Neurosciences, Cleveland, OH 44106, United States
| | - William Bush
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| | - Xiaofeng Zhu
- Case Western Reserve University Department of Population and Quantitative Health Sciences, Cleveland, OH 44106, United States
| |
Collapse
|
7
|
Liu J, Guan X, Gao S, Quan L, Dou M, Yue J, Shi M, Yuan P. Novel insight of critical genes involved in breast cancer brain metastasis: evidence from a cross-tissue transcriptome association study and validation through external clinical cohorts. BMC Cancer 2025; 25:707. [PMID: 40241087 PMCID: PMC12001416 DOI: 10.1186/s12885-025-14095-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: 11/18/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Breast cancer represents the most prevalent form of tumors among females and is characterized by a significant genetic component. The brain is a frequent site of metastasis for breast cancer. Although numerous loci associated with breast cancer brain metastasis (BCBM) have been identified, the critical regulatory genes underlying BCBM remain largely unclear. METHODS The FinnGen R11 dataset was combined with Genotype-Tissue Expression Project (GTEx) for Transcriptome-wide Association Study (TWAS). The Unified Test for Molecular Signatures (UTMOST), Multimarker Analysis of Genomic Annotation (MAGMA), and Functional Summary-based Imputation (FUSION) were used to identify candidate genes. Summary-data-based mendelian randomization (SMR) and co-localization were performed further to elucidate the association between key genes and BCBM. Finally, multiple external cohorts were obtained to validate the findings. RESULT In our study, 12 new genes associated with breast cancer were identified with TWAS. Subsequently, both SMR and co-localization have shown that CAPS8 was only expressed in brain tissues including frontal cortex and cerebellar hemispheres associated with breast cancer. Potential regulation of CASP8 could occur in BCBM. Finally, the findings were ultimately validated by external clinical cohorts. CONCLUSION Our study identified key gene CASP8, which was associated with BCBM, providing new insights into the occurrence of BCBM.
Collapse
Affiliation(s)
- Jinsong Liu
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiao Guan
- State Key Lab of Molecular Oncology, Department of Pancreatic and Gastric Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Songlin Gao
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Liuliu Quan
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Min Dou
- BengBu Medical University, BengBu, 233030, China
| | - Jian Yue
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mengwu Shi
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Yuan
- Department of VIP Medical Services, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
Deng C, Xie C, Li Z, Mei J, Wang K. Multi-omics analysis identifies diagnostic circulating biomarkers and potential therapeutic targets, revealing IQGAP1 as an oncogene in gastric cancer. NPJ Precis Oncol 2025; 9:105. [PMID: 40229327 PMCID: PMC11997149 DOI: 10.1038/s41698-025-00895-9] [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: 03/30/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
This study employed a multi-omics integration approach to identify circulating biomarkers for gastric cancer (GC). We analyzed plasma and tumor tissue single-cell RNA sequencing data, along with gene and protein quantitative trait loci analyses. Leveraging data from UK Biobank and FinnGen, we investigated genetic associations with GC. Through colocalization, Mendelian Randomization, and various filtering analyses, we identified four genes (IQGAP1, KRTCAP2, PARP1, MLF2) and four proteins (EGFL9 [DLK2], ECM1, PDIA5, TIMP4) as potential GC biomarkers. These were selected based on significant genetic colocation probabilities and significant associations with GC. Seven of these biomarkers demonstrated predictive capability for GC occurrence, with AUC ranging from 0.61 to 0.99. Drug prediction analysis identified seven protein biomarkers as potential targets for immunotherapy, targeted therapies, and tumor chemotherapy. Further scRNA-seq analysis revealed significant expression differences between gastric tumor and normal tissues, particularly the upregulation of IQGAP1, which highlights its role in tumor growth.
Collapse
Affiliation(s)
- Chao Deng
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chenjun Xie
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China
| | - Zixi Li
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China
| | - Jie Mei
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, 211166, China.
| | - Kewei Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Rd, Wuxi, 214122, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
9
|
Li X, Luo S, Lin K, Soha H, Shen M, Lu F, Wang J. Causal Links Between Corneal Biomechanics and Myopia: Evidence from Bidirectional Mendelian Randomization in the UK Biobank. Bioengineering (Basel) 2025; 12:412. [PMID: 40281772 PMCID: PMC12024697 DOI: 10.3390/bioengineering12040412] [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: 03/05/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Myopia is a leading cause of visual impairment worldwide, and accumulating evidence suggests that biomechanics may be closely linked to its development. Understanding this relationship may help clarify the underlying mechanisms of myopia and guide treatment strategies. The aim of the study is to investigate the causal relationship between myopia and corneal biomechanics using the UK Biobank (UKB) database. METHODS Data from 11,064 eyes in the UKB, including refraction results and Ocular Response Analyzer (ORA) measurements, were analyzed. Eyes were categorized by spherical equivalent (SE) into emmetropia, mild myopia, moderate myopia, and high myopia. One-way ANOVA assessed differences in corneal biomechanical parameters across the varying myopia groups, while Quantile Regression (QR) explored the relationship between these parameters and myopia severity across the different quantiles. A Mendelian randomization (MR) analysis was employed to explore the causal relationships. RESULTS Significant differences in corneal biomechanical parameters and intraocular pressure (IOP) were observed across the myopia levels (p < 0.001). High myopia was associated with lower corneal hysteresis (CH), a lower corneal resistance factor (CRF), and increased IOP. The QR analysis demonstrated that lower corneal biomechanics were associated with higher degrees of myopia, with the impact of corneal biomechanics becoming more pronounced as the myopia severity increased. The MR analysis indicated that low CH (OR = 0.9943, p = 0.004) and CRF (OR = 0.9946, p = 0.002) values were risk factors for myopia, while no causal effect was found when the myopia was treated as the exposure and corneal biomechanics as the outcome. CONCLUSIONS This study establishes a causal relationship where reduced corneal biomechanics contribute to myopia, while myopia itself does not directly affect biomechanics. Corneal biomechanics could serve as a biomarker for assessing high myopia risk. These findings offer new insights into high myopia's pathological mechanisms and targeted prevention.
Collapse
Affiliation(s)
- Xuefei Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (X.L.); (S.L.); (K.L.); (H.S.); (M.S.)
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Shenglong Luo
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (X.L.); (S.L.); (K.L.); (H.S.); (M.S.)
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Kuangching Lin
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (X.L.); (S.L.); (K.L.); (H.S.); (M.S.)
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hera Soha
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (X.L.); (S.L.); (K.L.); (H.S.); (M.S.)
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Meixiao Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (X.L.); (S.L.); (K.L.); (H.S.); (M.S.)
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Fan Lu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (X.L.); (S.L.); (K.L.); (H.S.); (M.S.)
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Junjie Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; (X.L.); (S.L.); (K.L.); (H.S.); (M.S.)
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Medical Devices and Drug for Ophthalmic Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
10
|
Zeng C, Zhang C, Jia Y, Zhou H, He C, Song H. Investigating the causal impact of gut microbiota on trigeminal neuralgia: a bidirectional Mendelian randomization study. Front Microbiol 2025; 16:1420978. [PMID: 40083778 PMCID: PMC11905160 DOI: 10.3389/fmicb.2025.1420978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
Background The etiology and pathogenesis of trigeminal neuralgia remain unclear. This study examines the connection between gut microbiota and trigeminal neuralgia using Mendelian randomization analysis to provide insights into the disorder's origin and propose potential therapies based on our findings. Methods We used data from the MiBioGen consortium (13,266 participants) for gut microbiota and the IEU OpenGWAS project (800 cases, 195,047 controls) for trigeminal neuralgia. We checked for heterogeneity and horizontal pleiotropy and used the inverse variance weighting method as our main approach to study the causal link between gut bacteria and trigeminal neuralgia, MR-Egger, simple mode, weighted median, and weighted mode as supplementary methods, with a sensitivity test using leave-one-out analysis. If a bacteria-trigeminal neuralgia link was found, we conducted a reverse analysis for confirmation. Results According to the final results, these groups include Butyricimonas (Genus, id = 945, p-value = 0.007, OR = 1.742, 95% CI: 1.165-2.604), unknowngenus (Genus, id = 1000005479, p-value = 0.005, OR = 1.774, 95% CI: 1.187-2.651) and Bacteroidales (Family, p-value = 0.005, OR = 1.774, 95% CI: 1.187-2.651) were causally associated with trigeminal neuralgia. No significant results according to reverse Mendelian randomization analysis. Conclusion In our study, we identified specific gut bacteria linked to trigeminal neuralgia. To comprehensively understand their impact and mechanisms, additional randomized trials are necessary.
Collapse
Affiliation(s)
- Chuan Zeng
- The First Clinical Medical College of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chaolong Zhang
- The First Clinical Medical College of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yuxuan Jia
- The First Clinical Medical College of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Huaiyu Zhou
- The First Clinical Medical College of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chunming He
- Department of Neurosurgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Haimin Song
- Department of Neurosurgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
11
|
He Z, Chu B, Yang J, Gu J, Chen Z, Liu L, Morrison T, Belloy ME, Qi X, Hejazi N, Mathur M, Le Guen Y, Tang H, Hastie T, Ionita-laza I, Candès E, Sabatti C. Beyond guilty by association at scale: searching for causal variants on the basis of genome-wide summary statistics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.28.582621. [PMID: 38464202 PMCID: PMC10925326 DOI: 10.1101/2024.02.28.582621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Understanding the causal genetic architecture of complex phenotypes will fuel future research into disease mechanisms and potential therapies. Here, we illustrate the power of a novel framework: it detects, starting from summary statistics, and across the entire genome, sets of variants that carry non-redundant information on the phenotypes and are therefore more likely to be causal in a biological sense. The approach, implemented in open-source software, is also computationally efficient, requiring less than 15 minutes on a single CPU to perform genome-wide analysis. Through extensive genome-wide simulation studies, we show that the method can substantially outperform existing methods in false discovery rate control, statistical power and various fine-mapping criteria. In applications to a meta-analysis of ten large-scale genetic studies of Alzheimer's disease (AD), we identified 82 loci associated with AD, including 37 additional loci missed by conventional GWAS pipeline. Massively parallel reporter assays and CRISPR-Cas9 experiments have confirmed the functionality of the putative causal variants our method points to. Finally, we retrospectively analyzed summary statistics from 67 large-scale GWAS for a variety of phenotypes. Results reveal the method's capacity to robustly discover additional loci for polygenic traits and pinpoint potential causal variants underpinning each locus beyond conventional GWAS pipeline, contributing to a deeper understanding of complex genetic architectures in post-GWAS analyses.
Collapse
Affiliation(s)
- Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Chu
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - James Yang
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Jiaqi Gu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Zhaomeng Chen
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Linxi Liu
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Tim Morrison
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Michael E. Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Xinran Qi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Nima Hejazi
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Maya Mathur
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Yann Le Guen
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Hua Tang
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Trevor Hastie
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Iuliana Ionita-laza
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Emmanuel Candès
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
- Department of Mathematics, Stanford University, Stanford, CA 94305, USA
| | - Chiara Sabatti
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Zhang L, Chu Q, Jiang S, Shao B. Genetic evidence for amlodipine's protective role in gastroesophageal reflux disease: A focus on CACNB2. PLoS One 2025; 20:e0309805. [PMID: 39965006 PMCID: PMC11835245 DOI: 10.1371/journal.pone.0309805] [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: 06/07/2024] [Accepted: 08/20/2024] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE This study aims to elucidate the causal relationship between genetically predicted amlodipine use and the risk of gastroesophageal reflux disease (GERD) using a bidirectional Mendelian Randomization (MR) approach and to explore the underlying genetic and molecular mechanisms through functional enrichment analysis and the construction of a competing endogenous RNA (ceRNA) network. METHODS Publicly available GWAS datasets from the Neale Lab consortium were used, including data on amlodipine (13,693 cases, 323,466 controls) and GERD (14,316 cases, 322,843 controls). Genome-wide significant SNPs were selected as instrumental variables and clustered by linkage disequilibrium. MR analysis was conducted using R software with all five methods. Sensitivity analyses assessed pleiotropy and heterogeneity. Drug target genes were analyzed using GO and KEGG pathways. GeneMANIA was used for network visualization, and a ceRNA network was constructed with Cytoscape. Differential gene expression analysis on GERD-related datasets from GEO validated the findings. RESULTS The MR analysis indicated a significant negative association between genetically predicted amlodipine use and GERD risk (IVW OR = 0.872, 95% CI = 0.812-0.937, P = 0.0002). Sensitivity analyses confirmed the robustness of these findings, showing no evidence of pleiotropy or heterogeneity. The enrichment analysis identified key biological processes and pathways involving calcium ion transport and signaling. The ceRNA network highlighted core targets such as CACNB2, which were further validated by differential expression analysis intersecting drug target genes with GERD-related gene expression changes. CONCLUSION This study provides robust evidence of a protective effect of amlodipine against GERD, supported by genetic and molecular analyses. The findings suggest that calcium channel blockers like amlodipine could be repurposed for GERD treatment. The identification of CACNB2 and other core targets in the ceRNA network offers novel insights into the pathophysiology of GERD and potential therapeutic targets, paving the way for personalized medicine approaches to improve patient outcomes.
Collapse
Affiliation(s)
- Liuzhao Zhang
- Department of Critical Care Medicine, Anhui Jing’an Medicine Hospital, Hefei, China
| | - Quanwang Chu
- Department of Critical Care Medicine, Anhui Jing’an Medicine Hospital, Hefei, China
| | - Shuyue Jiang
- Department of Critical Care Medicine, Anhui Jing’an Medicine Hospital, Hefei, China
| | - Bo Shao
- Department of Pathology, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
13
|
Woodward DJ, Thorp JG, Middeldorp CM, Akóṣílè W, Derks EM, Gerring ZF. Leveraging pleiotropy for the improved treatment of psychiatric disorders. Mol Psychiatry 2025; 30:705-721. [PMID: 39390223 PMCID: PMC11746150 DOI: 10.1038/s41380-024-02771-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
Over 90% of drug candidates fail in clinical trials, while it takes 10-15 years and one billion US dollars to develop a single successful drug. Drug development is more challenging for psychiatric disorders, where disease comorbidity and complex symptom profiles obscure the identification of causal mechanisms for therapeutic intervention. One promising approach for determining more suitable drug candidates in clinical trials is integrating human genetic data into the selection process. Genome-wide association studies have identified thousands of replicable risk loci for psychiatric disorders, and sophisticated statistical tools are increasingly effective at using these data to pinpoint likely causal genes. These studies have also uncovered shared or pleiotropic genetic risk factors underlying comorbid psychiatric disorders. In this article, we argue that leveraging pleiotropic effects will provide opportunities to discover novel drug targets and identify more effective treatments for psychiatric disorders by targeting a common mechanism rather than treating each disease separately.
Collapse
Affiliation(s)
- Damian J Woodward
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia.
| | - Jackson G Thorp
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Christel M Middeldorp
- Department of Child and Adolescent Psychiatry and Psychology, Amsterdam UMC, Amsterdam Reproduction and Development Research Institute, Amsterdam Public Health Research Institute, 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, QLD, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, Brisbane, QLD, Australia
| | - Wọlé Akóṣílè
- Greater Brisbane Clinical School, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Eske M Derks
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Zachary F Gerring
- Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- Healthy Development and Ageing, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| |
Collapse
|
14
|
Ding M, Zou F. A Linear Mixed Model with Measurement Error Correction (LMM-MEC): A Method for Summary-data-based Multivariable Mendelian Randomization. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2023.04.25.23289099. [PMID: 37162968 PMCID: PMC10168515 DOI: 10.1101/2023.04.25.23289099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Summary-data-based multivariable Mendelian randomization (MVMR) methods, such as MVMR-Egger, MVMR-IVW, MVMR median-based, and MVMR-PRESSO, assess the causal effects of multiple risk factors on disease. However, accounting for variances in summary statistics related to risk factors remains a challenge. We propose a linear mixed model with measurement error correction (LMM-MEC) that accounts for the variance of summary statistics for both disease outcomes and risk factors. In step I, a linear mixed model is applied to account for the variance in disease summary statistics. Specifically, if heterogeneity is present in disease summary statistics, we treat it as a random effect and adopt an iteratively re-weighted least squares algorithm to estimate causal effects. In step II, we treat the variance in the summary statistics of risk factors as multiple measurement errors and apply a regression calibration method for simultaneous multiple measurement error correction. In a simulation study, when using independent genetic variants as instrumental variables (IV), our method showed comparable performance to existing MVMR methods under conditions of no pleiotropy or balanced pleiotropy with the outcome, and it exhibited higher coverage rates and power under directional pleiotropy. Similar findings were observed when using genetic variants with low to moderate linkage disequilibrium (LD) (0 < ρ 2 ≤ 0.3) as IVs, although coverage rates reduced for all methods compared to using independent genetic variants as IVs. In the application study, we examined causal associations between correlated cholesterol biomarkers and longevity. By including 739 genetic variants selected based on P values <5×10 -5 from GWAS and allowing for low LD ( ρ 2 ≤ 0.1), our method identified that large LDL-c were causally associated with lower likelihood of achieving longevity.
Collapse
|
15
|
Yazdani A, Lenz HJ, Pillonetto G, Mendez-Giraldez R, Yazdani A, Sanoff H, Hadi R, Samiei E, Venook AP, Ratain MJ, Rashid N, Vincent BG, Qu X, Wen Y, Kosorok M, Symmans WF, Shen JPYC, Lee MS, Kopetz S, Nixon AB, Bertagnolli MM, Perou CM, Innocenti F. Gene signatures derived from transcriptomic-causal networks stratify colorectal cancer patients for effective targeted therapy. COMMUNICATIONS MEDICINE 2025; 5:9. [PMID: 39779996 PMCID: PMC11711454 DOI: 10.1038/s43856-024-00728-z] [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: 07/09/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Gene signatures derived from transcriptomic-causal networks offer potential for tailoring clinical care in cancer treatment by identifying predictive and prognostic biomarkers. This study aimed to uncover such signatures in metastatic colorectal cancer (CRC) patients to aid treatment decisions. METHODS We constructed transcriptomic-causal networks and integrated gene interconnectivity into overall survival (OS) analysis to control for confounding genes. This integrative approach involved germline genotype and tumor RNA-seq data from 1165 metastatic CRC patients. The patients were enrolled in a randomized clinical trial receiving either cetuximab or bevacizumab in combination with chemotherapy. An external cohort of paired CRC normal and tumor samples, along with protein-protein interaction databases, was used for replication. RESULTS We identify promising predictive and prognostic gene signatures from pre-treatment gene expression profiles. Our study discerns sets of genes, each forming a signature that collectively contribute to define patient subgroups with different prognosis and response to the therapies. Using an external cohort, we show that the genes influencing OS within the signatures, such as FANCI and PRC1, are upregulated in CRC tumor vs. normal tissue. These signatures are highly associated with immune features, including macrophages, cytotoxicity, and wound healing. Furthermore, the corresponding proteins encoded by the genes within the signatures interact with each other and are functionally related. CONCLUSIONS This study underscores the utility of gene signatures derived from transcriptomic-causal networks in patient stratification for effective therapies. The interpretability of the findings, supported by replication, highlights the potential of these signatures to identify patients likely to benefit from cetuximab or bevacizumab.
Collapse
Affiliation(s)
- Akram Yazdani
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- University of Texas Health Science Center at Houston, Texas, TX, USA.
| | | | | | - Raul Mendez-Giraldez
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Azam Yazdani
- Center of Perioperative Genetics and Genomics, Perioperative and Pain Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hanna Sanoff
- Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Reza Hadi
- School of Mathematics, University of Science and Technology of Iran, Tehran, Iran
| | | | - Alan P Venook
- University of California at San Francisco, San Francisco, CA, USA
| | - Mark J Ratain
- Division of the Biological Sciences, University of Chicago, Chicago, IL, USA
| | - Naim Rashid
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, US
| | - Benjamin G Vincent
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Xueping Qu
- Genentech, South San Francisco, San Francisco, CA, USA
| | - Yujia Wen
- Alliance for Clinical Trials in Oncology, Chicago, IL, USA
| | - Michael Kosorok
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, US
| | - William F Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Paul Y C Shen
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael S Lee
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew B Nixon
- Duke Center for Cancer Immunotherapy, Duke University, Durham, NC, USA
| | | | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Federico Innocenti
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Elenbaas JS, Lee PC, Patel V, Stitziel NO. Decoding the Therapeutic Target SVEP1: Harnessing Molecular Trait GWASs to Unravel Mechanisms of Human Disease. Annu Rev Pharmacol Toxicol 2025; 65:131-148. [PMID: 39847464 DOI: 10.1146/annurev-pharmtox-061724-080905] [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] [Indexed: 01/25/2025]
Abstract
Although human genetics has substantial potential to illuminate novel disease pathways and facilitate drug development, identifying causal variants and deciphering their mechanisms remain challenging. We believe these challenges can be addressed, in part, by creatively repurposing the results of molecular trait genome-wide association studies (GWASs). In this review, we introduce techniques related to molecular GWASs and unconventionally apply them to understanding SVEP1, a human coronary artery disease risk locus. Our analyses highlight SVEP1's causal link to cardiometabolic disease and glaucoma, as well as the surprising discovery of SVEP1 as the first known physiologic ligand for PEAR1, a critical receptor governing platelet reactivity. We further employ these techniques to dissect the interactions between SVEP1, PEAR1, and the Ang/Tie pathway, with therapeutic implications for a constellation of diseases. This review underscores the potential of molecular GWASs to guide drug discovery and unravel the complexities of human health and disease by demonstrating an integrative approach that grounds mechanistic research in human biology.
Collapse
Affiliation(s)
- Jared S Elenbaas
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Paul C Lee
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
- Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ved Patel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
| | - Nathan O Stitziel
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA;
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
17
|
Ran H, Li C, Rizvi SMM, Zhou R, Kong L, Shuangling S, Shao Y, Wu K, Duan C, Luo J, Shi H, Wu Q, Zhang C. Integrated analyses of Mendelian randomization, eQTL, and single-cell transcriptome identify CCN3 as a potential biomarker in aortic dissection. Sci Rep 2024; 14:32062. [PMID: 39738466 PMCID: PMC11685893 DOI: 10.1038/s41598-024-83611-0] [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: 05/22/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Plasma secretory proteins are associated with various diseases, including aortic dissection (AD). However, current research on the correlation between AD and plasma protein levels is scarce or lacks specificity. This study aimed to explore plasma secretory proteins as potential biomarkers for AD. Through genome-wide association studies, expression quantitative trait locus (eQTL) analysis, and human plasma protein profiling, we identified DBNL, NPC2, SUMF2, and TFPI as high-risk genes and CCN3, PRKCSH, TEX264, and TGFBR3 as low-risk genes for AD. Further cell localization and differential expression analysis of these eight genes were conducted using single-cell data. We also examined their expression in three Gene Expression Omnibus datasets, measured their mRNA levels in AD versus normal tissues using qPCR, and assessed their protein levels in patients' blood versus healthy individuals using enzyme-linked immunosorbent assay. Our findings suggest that CCN3, consistently downregulated in both mRNA and plasma levels during AD, may have a protective role. Initial enrichment analyses of differentially expressed CCN3 cells suggested their involvement in focal adhesion, actin cytoskeleton regulation, and the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Haoyu Ran
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changying Li
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Syed M Musa Rizvi
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruiqin Zhou
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Sun Shuangling
- Department of Biochemistry, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yue Shao
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kejia Wu
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing, China
| | - Changzhu Duan
- Department of Cell Biology and Genetics, Center for Molecular Medicine and Oncology Research, Chongqing Medical University, Chongqing, China
| | - Jun Luo
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haoming Shi
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingchen Wu
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Cheng Zhang
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
18
|
Liu S, Fu Z, Liu H, Wang Y, Zhou M, Ding Z, Feng Z. Lipid Profiles, Telomere Length, and the Risk of Malignant Tumors: A Mendelian Randomization and Mediation Analysis. Biomedicines 2024; 13:13. [PMID: 39857597 PMCID: PMC11760878 DOI: 10.3390/biomedicines13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The relationship between lipid profiles, telomere length (TL), and cancer risk remains unclear. Methods: This study employed two-sample Mendelian randomization (MR) with mediation analysis to investigate their causal relationships, examining lipid profiles as exposure, TL as mediator, and nine cancer types as outcomes. We conducted our analysis using two-stage least squares (2SLS) regression integrated with inverse variance weighted (IVW) methods to address potential endogeneity and strengthen our causal inference. Results: we found that unfavorable lipid profiles were causally linked to increased TL (p < 0.05). TL showed positive causal associations with lung and hematologic cancers (OR > 1, p < 0.05). Direct associations were observed between total and low-density lipoprotein (LDL) cholesterol and gastric cancer (OR < 1, p < 0.05), and between remnant cholesterol and colorectal cancer (OR > 1, p < 0.05). Mediation analysis revealed TL as a significant mediator in the pathway from lipid profiles to cancer development (p < 0.05). No horizontal pleiotropy was detected. Conclusions: Our findings suggest that lipid metabolism disorders may influence cancer development through telomere regulation, particularly in lung and hematologic cancers. This emphasizes the importance of lipid management in cancer prevention and treatment, especially for these cancer types.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenhua Ding
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (Z.F.); (H.L.); (Y.W.); (M.Z.)
| | - Zhijun Feng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (Z.F.); (H.L.); (Y.W.); (M.Z.)
| |
Collapse
|
19
|
Zhao M, Huang X, Zheng H, Cai Y, Han W, Wang Y, Chen R. Association between hypothyroidism and obstructive sleep apnea: a bidirectional Mendelian randomization study combined with the geo database. Front Neurol 2024; 15:1420391. [PMID: 39719972 PMCID: PMC11666497 DOI: 10.3389/fneur.2024.1420391] [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: 04/20/2024] [Accepted: 11/28/2024] [Indexed: 12/26/2024] Open
Abstract
Background The causal relationship between hypothyroidism and obstructive sleep apnea (OSA) remains controversial. Therefore, our research used a bidirectional Mendelian randomization (MR) method in an attempt to determine the causal relationship between hypothyroidism and OSA. Methods From the publicly accessible genome-wide association analysis (GWAS) summary database, we obtained single nucleotide polymorphism (SNPs) data pertaining to hypothyroidism and OSA. Inverse variance weighting (IVW) was the principal method of analysis utilized, with validation also conducted via weighted median, MR-Egger, simple model, and weighted model approaches. To further evaluate the robustness of the results, heterogeneity testing, pleiotropy testing, and the "leave-one-out" sensitivity analysis were performed. Differentially expressed genes (DEGs) from the OSA dataset (GSE135917) and hypothyroidism dataset (GSE176153) derived from the Gene Expression Omnibus (GEO) database were screened using the "limma" package. The "clusterProfiler" and "GO plot" packages were used for further enrichment analysis in order to validate the findings of the MR study. The Cytoscape software was utilized to build a protein-protein interaction (PPI) network of DEGs and to screen for hub genes. Results The MR analysis showed that genetically predicted hypothyroidism was associated with an increased risk of OSA [IVW odds ratio (OR) = 1.734; 95% confidence interval (CI) = 1.073-2.801; p = 0.025]. The trend of the outcomes of the other approaches is consistent with the trend of the IVW outcome. However, the reverse MR analysis suggested no evidence for the causal effect of OSA on hypothyroidism (IVW OR = 1.002, 95% CI: 0.996-1.009, p = 0.454). The robustness of the results was confirmed by the sensitivity analysis. Bioinformatics analysis revealed that there were DEGs that hypothyroidism and OSA have in common. Conclusion Our findings suggested that hypothyroidism may increase the risk of OSA, while the effect of OSA on hypothyroidism was not found in this MR study. Thus, patients with hypothyroidism should be enhanced with screening for OSA for early diagnosis and appropriate treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuanyin Wang
- Key Laboratory of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Anhui Medical University, Hefei, China
| | - Ran Chen
- Key Laboratory of Oral Diseases Research of Anhui Province, College & Hospital of Stomatology, Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Shao M, Chen K, Zhang S, Tian M, Shen Y, Cao C, Gu N. Multiome-wide Association Studies: Novel Approaches for Understanding Diseases. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae077. [PMID: 39471467 PMCID: PMC11630051 DOI: 10.1093/gpbjnl/qzae077] [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: 08/16/2023] [Revised: 10/06/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024]
Abstract
The rapid development of multiome (transcriptome, proteome, cistrome, imaging, and regulome)-wide association study methods have opened new avenues for biologists to understand the susceptibility genes underlying complex diseases. Thorough comparisons of these methods are essential for selecting the most appropriate tool for a given research objective. This review provides a detailed categorization and summary of the statistical models, use cases, and advantages of recent multiome-wide association studies. In addition, to illustrate gene-disease association studies based on transcriptome-wide association study (TWAS), we collected 478 disease entries across 22 categories from 235 manually reviewed publications. Our analysis reveals that mental disorders are the most frequently studied diseases by TWAS, indicating its potential to deepen our understanding of the genetic architecture of complex diseases. In summary, this review underscores the importance of multiome-wide association studies in elucidating complex diseases and highlights the significance of selecting the appropriate method for each study.
Collapse
Affiliation(s)
- Mengting Shao
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Kaiyang Chen
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Shuting Zhang
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Min Tian
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Yan Shen
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Chen Cao
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Ning Gu
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210093, China
| |
Collapse
|
21
|
Soubeyrand S, Lau P, Nikpay M, Ma L, Bjorkegren JLM, McPherson R. Long Noncoding RNA TRIBAL Links the 8q24.13 Locus to Hepatic Lipid Metabolism and Coronary Artery Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004674. [PMID: 39624902 DOI: 10.1161/circgen.124.004674] [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: 03/26/2024] [Accepted: 10/11/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Genome-wide association studies identified a 20-Kb region of chromosome 8 (8q24.13) associated with plasma lipids, hepatic steatosis, and risk for coronary artery disease. The region is proximal to TRIB1, and given its well-established role in lipid regulation in animal models, TRIB1 has been proposed to mediate the contribution of the 8q24.13 locus to these traits. This region overlaps a gene encoding the primate-specific long noncoding RNA transcript TRIBAL/TRIB1AL (TRIB1-associated locus), but the contribution of TRIBAL to coronary artery disease risk remains untested. METHODS Using recently available expression quantitative trait loci data and hepatocyte models, we further investigated this locus by Mendelian randomization analysis. Following antisense oligonucleotide targeting of TRIBAL, transcription array, quantitative reverse transcription polymerase chain reaction, and enrichment analyses were performed and effects on apoB and triglyceride secretion were determined. RESULTS Mendelian randomization analysis supports a causal relationship between genetically determined hepatic TRIBAL expression and markers of hepatic steatosis and coronary artery disease risk. By contrast, expression data sets did not support expression quantitative trait loci relationships between coronary artery disease-associated variants and TRIB1. TRIBAL suppression reduced the expression of key regulators of triglyceride metabolism and bile acid synthesis. Enrichment analyses identified patterns consistent with impaired metabolic functions, including reduced triglyceride and cholesterol handling ability. Furthermore, TRIBAL suppression was associated with reduced hepatocyte secretion of triglycerides. CONCLUSIONS This work identifies TRIBAL as a gene bridging the genotype-phenotype relationship at the 8q24.13 locus with effects on genes regulating hepatocyte lipid metabolism and triglyceride secretion.
Collapse
Affiliation(s)
- Sébastien Soubeyrand
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
| | - Paulina Lau
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
| | - Majid Nikpay
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (L.M., J.L.M.B.)
| | - Johan L M Bjorkegren
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (L.M., J.L.M.B.)
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden (J.L.M.B.)
| | - Ruth McPherson
- Atherogenomics Laboratory (S.S., P.L., M.N., R.M.), University of Ottawa Heart Institute, Canada
- Division of Cardiology, Ruddy Canadian Cardiovascular Genetics Centre (R.M.), University of Ottawa Heart Institute, Canada
| |
Collapse
|
22
|
Piedade AP, Butler J, Eyre S, Orozco G. The importance of functional genomics studies in precision rheumatology. Best Pract Res Clin Rheumatol 2024; 38:101988. [PMID: 39174375 DOI: 10.1016/j.berh.2024.101988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
Rheumatic diseases, those that affect the musculoskeletal system, cause significant morbidity. Among risk factors of these diseases is a significant genetic component. Recent advances in high-throughput omics techniques now allow a comprehensive profiling of patients at a genetic level through genome-wide association studies. Without functional interpretation of variants identified through these studies, clinical insight remains limited. Strategies include statistical fine-mapping that refine the list of variants in loci associated with disease, whilst colocalization techniques attempt to attribute function to variants that overlap a genetically active chromatin annotation. Functional validation using genome editing techniques can be used to further refine genetic signals and identify key pathways in cell types relevant to rheumatic disease biology. Insight gained from the combination of genetic studies and functional validation can be used to improve precision medicine in rheumatic diseases by allowing risk prediction and drug repositioning.
Collapse
Affiliation(s)
- Ana Pires Piedade
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Jake Butler
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Stephen Eyre
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Gisela Orozco
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
23
|
Lou J, Tu M, Xu M, Cao Z, Song W. Plasma pQTL and brain eQTL integration identifies PNKP as a therapeutic target and reveals mechanistic insights into migraine pathophysiology. J Headache Pain 2024; 25:202. [PMID: 39578729 PMCID: PMC11585170 DOI: 10.1186/s10194-024-01922-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Migraine is a prevalent neurological disorder affecting 14.1% of the global population. Despite advances in genetic research, further investigation is needed to identify therapeutic targets and better understand its mechanisms. In this study, we aimed to identify drug targets and explore the relationships between gene expression, protein levels, and migraine pathophysiology. METHODS We utilized cis-pQTL data from deCODE Genetics, combined with migraine GWAS data from the GERA + UKB cohort as the discovery cohort and the FinnGen R10 cohort as the replication cohort. SMR and MR analyses identified migraine-associated protein loci. Brain eQTL data from GTEx v8 and BrainMeta v2 were used to explore causal relationships between gene expression, protein levels, and migraine risk. Mediation analysis assessed the role of metabolites, and PheWAS evaluated potential side effects. RESULTS Four loci were identified: PNKP, MRVI1, CALCB, and INPP5B. PNKP and MRVI1 showed a high level of evidence and opposing effects at the gene and protein levels. PNKP gene expression in certain brain regions was protective against migraine, while its plasma protein levels were positively associated with migraine risk. MRVI1 showed protective effects at the protein level but had the opposite effect at the gene expression level. Mediation analysis revealed that the glutamate to pyruvate ratio and 3-CMPFP mediated PNKP's effects on migraine. PheWAS indicated associations between PNKP and body composition traits, suggesting drug safety considerations. CONCLUSION PNKP and MRVI1 exhibit dual mechanisms of action at the gene and protein levels, potentially involving distinct mechanistic pathways. Among them, PNKP emerges as a promising drug target for migraine treatment, supported by multi-layered validation.
Collapse
Affiliation(s)
- Jiafei Lou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Miaoqian Tu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijian Cao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Wenwen Song
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
24
|
Auwerx C, Moix S, Kutalik Z, Reymond A. Disentangling mechanisms behind the pleiotropic effects of proximal 16p11.2 BP4-5 CNVs. Am J Hum Genet 2024; 111:2347-2361. [PMID: 39332408 PMCID: PMC11568757 DOI: 10.1016/j.ajhg.2024.08.014] [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: 03/28/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/29/2024] Open
Abstract
Whereas 16p11.2 BP4-5 copy-number variants (CNVs) represent one of the most pleiotropic etiologies of genomic syndromes in both clinical and population cohorts, the mechanisms leading to such pleiotropy remain understudied. Identifying 73 deletion and 89 duplication carrier individuals among unrelated White British UK Biobank participants, we performed a phenome-wide association study (PheWAS) between the region's copy number and 117 complex traits and diseases, mimicking four dosage models. Forty-six phenotypes (39%) were affected by 16p11.2 BP4-5 CNVs, with the deletion-only, mirror, U-shape, and duplication-only models being the best fit for 30, 10, 4, and 2 phenotypes, respectively, aligning with the stronger deleteriousness of the deletion. Upon individually adjusting CNV effects for either body mass index (BMI), height, or educational attainment (EA), we found that sixteen testable deletion-driven associations-primarily with cardiovascular and metabolic traits-were BMI dependent, with EA playing a more subtle role and no association depending on height. Bidirectional Mendelian randomization supported that 13 out of these 16 associations were secondary consequences of the CNV's impact on BMI. For the 23 traits that remained significantly associated upon individual adjustment for mediators, matched-control analyses found that 10 phenotypes, including musculoskeletal traits, liver enzymes, fluid intelligence, platelet count, and pneumonia and acute kidney injury risk, remained associated under strict Bonferroni correction, with 10 additional nominally significant associations. These results paint a complex picture of 16p11.2 BP4-5's pleiotropic pattern that involves direct effects on multiple physiological systems and indirect co-morbidities consequential to the CNV's impact on BMI and EA, acting through trait-specific dosage mechanisms.
Collapse
Affiliation(s)
- Chiara Auwerx
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland; Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Samuel Moix
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland; University Center for Primary Care and Public Health, Lausanne, Switzerland.
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
25
|
Khan M, Ludl AA, Bankier S, Björkegren JLM, Michoel T. Prediction of causal genes at GWAS loci with pleiotropic gene regulatory effects using sets of correlated instrumental variables. PLoS Genet 2024; 20:e1011473. [PMID: 39527631 PMCID: PMC11581411 DOI: 10.1371/journal.pgen.1011473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 11/21/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Multivariate Mendelian randomization (MVMR) is a statistical technique that uses sets of genetic instruments to estimate the direct causal effects of multiple exposures on an outcome of interest. At genomic loci with pleiotropic gene regulatory effects, that is, loci where the same genetic variants are associated to multiple nearby genes, MVMR can potentially be used to predict candidate causal genes. However, consensus in the field dictates that the genetic instruments in MVMR must be independent (not in linkage disequilibrium), which is usually not possible when considering a group of candidate genes from the same locus. Here we used causal inference theory to show that MVMR with correlated instruments satisfies the instrumental set condition. This is a classical result by Brito and Pearl (2002) for structural equation models that guarantees the identifiability of individual causal effects in situations where multiple exposures collectively, but not individually, separate a set of instrumental variables from an outcome variable. Extensive simulations confirmed the validity and usefulness of these theoretical results. Importantly, the causal effect estimates remained unbiased and their variance small even when instruments are highly correlated, while bias introduced by horizontal pleiotropy or LD matrix sampling error was comparable to standard MR. We applied MVMR with correlated instrumental variable sets at genome-wide significant loci for coronary artery disease (CAD) risk using expression Quantitative Trait Loci (eQTL) data from seven vascular and metabolic tissues in the STARNET study. Our method predicts causal genes at twelve loci, each associated with multiple colocated genes in multiple tissues. We confirm causal roles for PHACTR1 and ADAMTS7 in arterial tissues, among others. However, the extensive degree of regulatory pleiotropy across tissues and the limited number of causal variants in each locus still require that MVMR is run on a tissue-by-tissue basis, and testing all gene-tissue pairs with cis-eQTL associations at a given locus in a single model to predict causal gene-tissue combinations remains infeasible. Our results show that within tissues, MVMR with dependent, as opposed to independent, sets of instrumental variables significantly expands the scope for predicting causal genes in disease risk loci with pleiotropic regulatory effects. However, considering risk loci with regulatory pleiotropy that also spans across tissues remains an unsolved problem.
Collapse
Affiliation(s)
- Mariyam Khan
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Adriaan-Alexander Ludl
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Sean Bankier
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Johan L. M. Björkegren
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
- Department of Genetics & Genomic Sciences/Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Tom Michoel
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| |
Collapse
|
26
|
Liao J, Jiang L, Qin Y, Hu J, Tang Z. GENETIC PREDICTION OF CAUSAL RELATIONSHIPS BETWEEN OSTEOPOROSIS AND SEPSIS: EVIDENCE FROM MENDELIAN RANDOMIZATION WITH TWO-SAMPLE DESIGNS. Shock 2024; 62:628-632. [PMID: 38813935 DOI: 10.1097/shk.0000000000002383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
ABSTRACT Background: Recent observational studies have suggested that osteoporosis may be a risk factor for sepsis. To mitigate confounding factors and establish the causal relationship between sepsis and osteoporosis, we conducted a two-sample Mendelian randomization analysis using publicly available summary statistics. Methods: Utilizing summary data from FinnGen Biobank, we employed a two-sample Mendelian randomization (MR) analysis to predict the causal relationship between osteoporosis and sepsis. The MR analysis primarily utilized the inverse variance weighted (IVW) method, supplemented by MR-Egger, weighted median, weighted mode, and simple mode analyses, with Bayesian weighted MR (BWMR) analysis employed for result validation. Sensitivity analyses included MR-PRESSO, "leave-one-out" analysis, MR-Egger regression, and Cochran Q test. Results: In the European population, an increase of one standard deviation in osteoporosis was associated with an 11% increased risk of sepsis, with an odds ratio (OR) of 1.11 (95% CI, 1.06-1.16; P = 3.75E-06). BWMR yielded an OR of 1.11 (95% CI, 1.06-1.67; P = 1.21E-05), suggesting osteoporosis as a risk factor for sepsis. Conversely, an increase of one standard deviation in sepsis was associated with a 26% increased risk of osteoporosis, with an OR of 1.26 (95% CI, 1.11-1.16; P = 0.45E-03). BWMR yielded an OR of 1.26 (95% CI, 1.09-1.45; P = 1.45E-03), supporting sepsis as a risk factor for osteoporosis. Conclusion: There is an association between osteoporosis and sepsis, with osteoporosis serving as a risk factor for the development of sepsis, while sepsis may also promote the progression of osteoporosis.
Collapse
Affiliation(s)
- Jing Liao
- Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | | | | | | | | |
Collapse
|
27
|
Zhang W, Su CY, Yoshiji S, Lu T. MR Corge: sensitivity analysis of Mendelian randomization based on the core gene hypothesis for polygenic exposures. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 40:btae666. [PMID: 39513749 DOI: 10.1093/bioinformatics/btae666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/19/2024] [Accepted: 11/07/2024] [Indexed: 11/15/2024]
Abstract
SUMMARY Mendelian randomization is being utilized to assess causal effects of polygenic exposures, where many genetic instruments are subject to horizontal pleiotropy. Existing methods for detecting and correcting for horizontal pleiotropy have important assumptions that may not be fulfilled. Built upon the core gene hypothesis, we developed MR Corge for performing sensitivity analysis of Mendelian randomization. MR Corge identifies a small number of putative core instruments that are more likely to affect genes with a direct biological role in an exposure and obtains causal effect estimates based on these instruments, thereby reducing the risk of horizontal pleiotropy. Using positive and negative controls, we demonstrated that MR Corge estimates aligned with established biomedical knowledge and the results of randomized controlled trials. MR Corge may be widely applied to investigate polygenic exposure-outcome relationships. AVAILABILITY AND IMPLEMENTATION An open-sourced R package is available at https://github.com/zhwm/MRCorge.
Collapse
Affiliation(s)
- Wenmin Zhang
- Montreal Heart Institute, Montreal, QC, H1T 1C8, Canada
| | - Chen-Yang Su
- Quantitative Life Sciences Program, McGill University, Montreal, QC, H3A 0G4, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Satoshi Yoshiji
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, H3A 0G1, Canada
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
- Lady Davis Institute for Medical Research, Montreal, QC, H3T 1E2, Canada
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, United States
- Harvard Medical School, Boston, MA, 02115, United States
| | - Tianyuan Lu
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, 53726, United States
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53726, United States
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI, 53706, United States
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI, 53706, United States
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, 53705, United States
| |
Collapse
|
28
|
Lorincz-Comi N, Yang Y, Ajayakumar J, Mews M, Bermudez V, Bush W, Zhu X. HORNET: Tools to find genes with causal evidence and their regulatory networks using eQTLs. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.28.24316273. [PMID: 39574873 PMCID: PMC11581076 DOI: 10.1101/2024.10.28.24316273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Motivation Nearly two decades of genome-wide association studies (GWAS) have identify thousands of disease-associated genetic variants, but very few genes with evidence of causality. Recent methodological advances demonstrate that Mendelian Randomization (MR) using expression quantitative loci (eQTLs) as instrumental variables can detect potential causal genes. However, existing MR approaches are not well suited to handle the complexity of eQTL GWAS data structure and so they are subject to bias, inflation, and incorrect inference. Results We present a whole-genome regulatory network analysis tool (HORNET), which is a comprehensive set of statistical and computational tools to perform genome-wide searches for causal genes using summary level GWAS data that is robust to biases from multiple sources. Applying HORNET to schizophrenia, we identified differential magnitudes of gene expression causality. Applying HORNET to schizophrenia, we identified differential magnitudes of gene expression causality across different brain tissues. Availability and Implementation Freely available at https://github.com/noahlorinczcomi/HORNETor Mac, Windows, and Linux users. Contact njl96@case.edu .
Collapse
|
29
|
Liu J, Feng G. The causal relationship between trace element status and upper gastrointestinal ulcers: a Mendelian randomization study. Front Nutr 2024; 11:1443090. [PMID: 39539362 PMCID: PMC11557352 DOI: 10.3389/fnut.2024.1443090] [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: 06/03/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background This study aimed to investigate the bidirectional causal relationships between trace elements (such as zinc, magnesium, phosphate, and folate) and upper gastrointestinal ulcers (including gastric and duodenal ulcers). We utilized a two-sample Mendelian randomization (MR) analysis to achieve this. Methods We conducted a two-sample MR analysis using summary-level data from genome-wide association studies (GWAS) obtained from public genomics repositories. We utilized a range of MR methods, including inverse-variance weighted (IVW), MR-Egger, and weighted median methods, and conducted a meta-analysis to synthesize results across different datasets. To ensure the robustness of our findings, we performed extensive sensitivity analyses, including pleiotropy assessment, heterogeneity tests, and leave-one-out analysis. Results Our findings are significant, indicating a positive causal relationship between increased zinc levels and the risk of gastric ulcers. Moreover, magnesium and folate appear to offer potential protective effects against gastroduodenal ulcers (p < 0.05). The meta-analysis further supports the causal relationship between zinc and gastric ulcers (p < 0.05), confirming zinc's significant causal impact on this condition. Conclusion The study confirms a positive causal relationship between zinc and gastric ulcers and highlights the complexity of how trace elements regulate the progression of upper gastrointestinal ulcers. These results provide a scientific basis for dietary recommendations regarding trace element intake in clinical and public health practices. They also offer new insights into effective prevention and treatment strategies for gastric and duodenal ulcers.
Collapse
Affiliation(s)
- Jianwei Liu
- Department of Gastroenterology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Gege Feng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
30
|
Ružičková N, Hledík M, Tkačik G. Quantitative omnigenic model discovers interpretable genome-wide associations. Proc Natl Acad Sci U S A 2024; 121:e2402340121. [PMID: 39441639 PMCID: PMC11536075 DOI: 10.1073/pnas.2402340121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
As their statistical power grows, genome-wide association studies (GWAS) have identified an increasing number of loci underlying quantitative traits of interest. These loci are scattered throughout the genome and are individually responsible only for small fractions of the total heritable trait variance. The recently proposed omnigenic model provides a conceptual framework to explain these observations by postulating that numerous distant loci contribute to each complex trait via effect propagation through intracellular regulatory networks. We formalize this conceptual framework by proposing the "quantitative omnigenic model" (QOM), a statistical model that combines prior knowledge of the regulatory network topology with genomic data. By applying our model to gene expression traits in yeast, we demonstrate that QOM achieves similar gene expression prediction performance to traditional GWAS with hundreds of times less parameters, while simultaneously extracting candidate causal and quantitative chains of effect propagation through the regulatory network for every individual gene. We estimate the fraction of heritable trait variance in cis- and in trans-, break the latter down by effect propagation order, assess the trans- variance not attributable to transcriptional regulation, and show that QOM correctly accounts for the low-dimensional structure of gene expression covariance. We furthermore demonstrate the relevance of QOM for systems biology, by employing it as a statistical test for the quality of regulatory network reconstructions, and linking it to the propagation of nontranscriptional (including environmental) effects.
Collapse
Affiliation(s)
- Natália Ružičková
- Institute of Science and Technology Austria, KlosterneuburgAT-3400, Austria
| | - Michal Hledík
- Institute of Science and Technology Austria, KlosterneuburgAT-3400, Austria
| | - Gašper Tkačik
- Institute of Science and Technology Austria, KlosterneuburgAT-3400, Austria
| |
Collapse
|
31
|
Wang P, Liu S, Kong LM, Qi N. Causal Relationship Between Childhood Obesity and Sleep Apnea Syndrome: Bidirectional Two-Sample Mendelian Randomization Analysis. Nat Sci Sleep 2024; 16:1713-1723. [PMID: 39464513 PMCID: PMC11512557 DOI: 10.2147/nss.s477435] [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: 05/08/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Background Childhood obesity has become a global pandemic, leading to a range of diseases. Childhood obesity appears to be associated with an increased prevalence of sleep apnea syndrome. Sleep apnea is an inestimable risk factor for thrombosis, hypertension, cardiomyopathy and many other diseases. Therefore, exploring the relationship between childhood obesity and sleep apnea syndrome will help to understand the potential link between the two and provide research directions for future disease prevention and treatment. However, no studies have confirmed whether there is a causal relationship between childhood obesity and sleep apnea syndrome. Methods The IEU OpenGWAS project provided the GWAS-aggregated data for childhood obesity and sleep apnea syndrome. Inverse-variance weighted (IVW) was used as the main method to evaluate the causal relationship between childhood obesity and sleep apnea syndrome. Single nucleotide polymorphisms (SNPs) were regarded as instrumental variables, and the screening threshold was P <5.0×10-6. Leave-one-out method was performed to confirm the robustness of the results. Results IVW analysis confirmed a causal relationship between genetic susceptibility to childhood obesity and an increased risk of sleep apnea syndrome [odds ratio (OR)=1.12, 95% confidence interval (CI): 1.02-1.23, P=0.016]. However, two-sample MR results also showed no causal relationship between genetic susceptibility to sleep apnea syndrome and an increased risk of childhood obesity (OR=1.50, 95% CI: 0.95-2.38, P=0.083). The intercept of MR-Egger regression was close to 0, which implies that there are no confounding factors in the analysis to affect the results of two-sample MR analysis. The leave-one-out results show that the bidirectional two-sample MR analysis results were robust. Conclusion There is a causal relationship between genetic susceptibility to childhood obesity and increased risk of sleep apnea syndrome. People with a history of childhood obesity should pay more attention to physical examination to early prevention and management of sleep apnea syndrome.
Collapse
Affiliation(s)
- Ping Wang
- Department of Pediatrics, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, People’s Republic of China
| | - Shuli Liu
- Department of Pediatrics, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, People’s Republic of China
| | - Ling Min Kong
- Department of Pediatrics, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, People’s Republic of China
| | - Nannan Qi
- Department of Pediatrics, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, People’s Republic of China
| |
Collapse
|
32
|
Wen D, Li B, Guo S, Chen L, Chen B. Exploring Pathogenic Genes in Frozen Shoulder through weighted gene co-expression network analysis and Mendelian Randomization. Int J Med Sci 2024; 21:2745-2758. [PMID: 39512681 PMCID: PMC11539380 DOI: 10.7150/ijms.98505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Background: Frozen shoulder (FS) is characterized by the thickening and fibrosis of the joint capsule, leading to joint contracture and a reduction in joint volume. The precise etiology responsible for these pathological changes remains elusive. Therefore, the primary aim of this study was to explore the potential involvement of pathogenic genes in FS and analyze their underlying roles in the disease progression. Methods: Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were employed to investigate co-expressed genes potentially associated with FS. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were conducted to elucidate the potential roles of these co-expressed genes. Subsequently, Mendelian randomization (MR) analysis was performed using expression quantitative trait loci datasets for the co-expressed genes, combined with summary statistics from the genome-wide association study of FS, aiming to identify key genes causally associated with FS. The identified key genes were further validated through reverse transcription-quantitative PCR (RT-qPCR). Additionally, a nomogram model and receiver operating characteristic (ROC) curves were established to assess the diagnostic value of the hub genes. Furthermore, the infiltration of immune cells was evaluated using the CIBERSORT algorithm and the relationship between key genes and immune-infiltrating cells was analyzed. Results: 295 overlapping co-expressed genes were identified by intersecting the differentially expressed genes with the hub genes obtained from associated modules identified through WGCNA. Utilizing MR analysis, four key genes, namely ADAMTS1, NR4A2, PARD6G and SMKR1, were found to exhibit positive causal relationships with FS, which were subsequently validated through RT-qPCR analysis. Moreover, the diagnostic value of these four key genes was demonstrated through the development of a nomogram model and the construction of ROC curves. Notably, a causal relationship between ADAMTS1 and immune cell infiltration in FS was observed. Conclusion: Our study suggested genetic predisposition to higher expression levels of ADAMTS1, NR4A2, PARD6G and SMKR1, was associated with an increased risk of FS. Further investigations elucidating the functional roles of these genes will enhance our understanding of the pathogenesis of FS and may facilitate the development of targeted treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Biao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
33
|
Tambets R, Kolde A, Kolberg P, Love MI, Alasoo K. Extensive co-regulation of neighboring genes complicates the use of eQTLs in target gene prioritization. HGG ADVANCES 2024; 5:100348. [PMID: 39210598 PMCID: PMC11416642 DOI: 10.1016/j.xhgg.2024.100348] [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: 02/02/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Identifying causal genes underlying genome-wide association studies (GWASs) is a fundamental problem in human genetics. Although colocalization with gene expression quantitative trait loci (eQTLs) is often used to prioritize GWAS target genes, systematic benchmarking has been limited due to unavailability of large ground truth datasets. Here, we re-analyzed plasma protein QTL data from 3,301 individuals of the INTERVAL cohort together with 131 eQTL Catalog datasets. Focusing on variants located within or close to the affected protein identified 793 proteins with at least one cis-pQTL where we could assume that the most likely causal gene was the gene coding for the protein. We then benchmarked the ability of cis-eQTLs to recover these causal genes by comparing three Bayesian colocalization methods (coloc.susie, coloc.abf, and CLPP) and five Mendelian randomization (MR) approaches (three varieties of inverse-variance weighted MR, MR-RAPS, and MRLocus). We found that assigning fine-mapped pQTLs to their closest protein coding genes outperformed all colocalization methods regarding both precision (71.9%) and recall (76.9%). Furthermore, the colocalization method with the highest recall (coloc.susie - 46.3%) also had the lowest precision (45.1%). Combining evidence from multiple conditionally distinct colocalizing QTLs with MR increased precision to 81%, but this was accompanied by a large reduction in recall to 7.1%. Furthermore, the choice of the MR method greatly affected performance, with the standard inverse-variance-weighted MR often producing many false positives. Our results highlight that linking GWAS variants to target genes remains challenging with eQTL evidence alone, and prioritizing novel targets requires triangulation of evidence from multiple sources.
Collapse
Affiliation(s)
- Ralf Tambets
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Anastassia Kolde
- Institute of Genomics, University of Tartu, Tartu, Estonia; Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Peep Kolberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Michael I Love
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kaur Alasoo
- Institute of Computer Science, University of Tartu, Tartu, Estonia.
| |
Collapse
|
34
|
Bruner WS, Grant SFA. Translation of genome-wide association study: from genomic signals to biological insights. Front Genet 2024; 15:1375481. [PMID: 39421299 PMCID: PMC11484060 DOI: 10.3389/fgene.2024.1375481] [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: 01/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Since the turn of the 21st century, genome-wide association study (GWAS) have successfully identified genetic signals associated with a myriad of common complex traits and diseases. As we transition from establishing robust genetic associations with diverse phenotypes, the central challenge is now focused on characterizing the underlying functional mechanisms driving these signals. Previous GWAS efforts have revealed multiple variants, each conferring relatively subtle susceptibility, collectively contributing to the pathogenesis of various common diseases. Such variants can further exhibit associations with multiple other traits and differ across ancestries, plus disentangling causal variants from non-causal due to linkage disequilibrium complexities can lead to challenges in drawing direct biological conclusions. Combined with cellular context considerations, such challenges can reduce the capacity to definitively elucidate the biological significance of GWAS signals, limiting the potential to define mechanistic insights. This review will detail current and anticipated approaches for functional interpretation of GWAS signals, both in terms of characterizing the underlying causal variants and the corresponding effector genes.
Collapse
Affiliation(s)
- Winter S. Bruner
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Struan F. A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
35
|
Chen J, Shen L, Wu T, Yang Y. Unraveling the significance of AGPAT4 for the pathogenesis of endometriosis via a multi-omics approach. Hum Genet 2024; 143:1163-1174. [PMID: 38850428 PMCID: PMC11485110 DOI: 10.1007/s00439-024-02681-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Endometriosis is characterized by the ectopic proliferation of endometrial cells, posing considerable diagnostic and therapeutic challenges. Our study investigates AGPAT4's involvement in endometriosis pathogenesis, aiming to unveil new therapeutic targets. Our investigation by analyzing eQTL data from GWAS for preliminary screening. Subsequently, within the GEO dataset, we utilized four machine learning algorithms to precisely identify risk-associated genes. Gene validity was confirmed through five Mendelian Randomization methods. AGPAT4 expression was measured by Single-Cell Analysis, ELISA and immunohistochemistry. We investigated AGPAT4's effect on endometrial stromal cells using RNA interference, assessing cell proliferation, invasion, and migration with CCK8, wound-healing, and transwell assays. Protein expression was analyzed by western blot, and AGPAT4 interactions were explored using AutoDock. Our investigation identified 11 genes associated with endometriosis risk, with AGPAT4 and COMT emerging as pivotal biomarkers through machine learning analysis. AGPAT4 exhibited significant upregulation in both ectopic tissues and serum samples from patients with endometriosis. Reduced expression of AGPAT4 was observed to detrimentally impact the proliferation, invasion, and migration capabilities of endometrial stromal cells, concomitant with diminished expression of key signaling molecules such as Wnt3a, β-Catenin, MMP-9, and SNAI2. Molecular docking analyses further underscored a substantive interaction between AGPAT4 and Wnt3a.Our study highlights AGPAT4's key role in endometriosis, influencing endometrial stromal cell behavior, and identifies AGPAT4 pathways as promising therapeutic targets for this condition.
Collapse
Affiliation(s)
- Jun Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Licong Shen
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Tingting Wu
- Department of Cardiovasology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yongwen Yang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
36
|
Khan M, Ludl AA, Bankier S, Björkegren JLM, Michoel T. Prediction of causal genes at GWAS loci with pleiotropic gene regulatory effects using sets of correlated instrumental variables. ARXIV 2024:arXiv:2401.06261v3. [PMID: 38259344 PMCID: PMC10802687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Multivariate Mendelian randomization (MVMR) is a statistical technique that uses sets of genetic instruments to estimate the direct causal effects of multiple exposures on an outcome of interest. At genomic loci with pleiotropic gene regulatory effects, that is, loci where the same genetic variants are associated to multiple nearby genes, MVMR can potentially be used to predict candidate causal genes. However, consensus in the field dictates that the genetic instruments in MVMR must be independent (not in linkage disequilibrium, which is usually not possible when considering a group of candidate genes from the same locus. Here we used causal inference theory to show that MVMR with correlated instruments satisfies the instrumental set condition. This is a classical result by Brito and Pearl (2002) for structural equation models that guarantees the identifiability of individual causal effects in situations where multiple exposures collectively, but not individually, separate a set of instrumental variables from an outcome variable. Extensive simulations confirmed the validity and usefulness of these theoretical results. Importantly, the causal effect estimates remained unbiased and their variance small even when instruments are highly correlated, while bias introduced by horizontal pleiotropy or LD matrix sampling error was comparable to standard MR. We applied MVMR with correlated instrumental variable sets at genome-wide significant loci for coronary artery disease (CAD) risk using expression Quantitative Trait Loci (eQTL) data from seven vascular and metabolic tissues in the STARNET study. Our method predicts causal genes at twelve loci, each associated with multiple colocated genes in multiple tissues. We confirm causal roles for PHACTR 1 and ADAMTS 7 in arterial tissues, among others. However, the extensive degree of regulatory pleiotropy across tissues and the limited number of causal variants in each locus still require that MVMR is run on a tissue-by-tissue basis, and testing all gene-tissue pairs with cis-eQTL associations at a given locus in a single model to predict causal gene-tissue combinations remains infeasible. Our results show that within tissues, MVMR with dependent, as opposed to independent, sets of instrumental variables significantly expands the scope for predicting causal genes in disease risk loci with pleiotropic regulatory effects. However, considering risk loci with regulatory pleiotropy that also spans across tissues remains an unsolved problem.
Collapse
Affiliation(s)
- Mariyam Khan
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Adriaan-Alexander Ludl
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Sean Bankier
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Johan LM Björkegren
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
- Department of Genetics & Genomic Sciences/Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tom Michoel
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| |
Collapse
|
37
|
Timasheva Y, Lepik K, Liska O, Papp B, Kutalik Z. Widespread natural selection on metabolite levels in humans. Genome Res 2024; 34:1121-1129. [PMID: 39152035 PMCID: PMC11444169 DOI: 10.1101/gr.278756.123] [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: 11/20/2023] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Natural selection acts ubiquitously on complex human traits, predominantly constraining the occurrence of extreme phenotypes (stabilizing selection). These constraints propagate to DNA sequence variants associated with traits under selection. The genetic signatures of such evolutionary events can thus be detected via combining effect size estimates from genetic association studies and the corresponding allele frequencies. Although this approach has been successfully applied to high-level traits, the prevalence and mode of selection acting on molecular traits remain poorly understood. Here, we estimate the action of natural selection on genetic variants associated with metabolite levels, an important layer of molecular traits. By leveraging summary statistics of published genome-wide association studies with large sample sizes, we find strong evidence of stabilizing selection for 15 out of 97 plasma metabolites, with nonessential amino acids displaying especially strong selection signatures. Mendelian randomization analysis reveals that metabolites under stronger stabilizing selection display larger effects on a range of clinically relevant complex traits, suggesting that maintaining a disease-free profile may be an important source of selective constraints on the metabolome. Metabolites under strong stabilizing selection in humans are also more conserved in their concentrations among diverse mammalian species, suggesting shared selective forces across micro- and macroevolutionary timescales. Overall, this study demonstrates that variation in metabolite levels among humans is frequently shaped by natural selection and this may act through their causal impact on disease susceptibility.
Collapse
Affiliation(s)
- Yanina Timasheva
- Institute of Biochemistry and Genetics, Ufa Federal Research Center of Russian Academy of Sciences, 450054 Ufa, Russia
- Department of Medical Genetics, Bashkir State Medical University, 450008 Ufa, Russia
- Department of Computational Biology, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Kaido Lepik
- Department of Computational Biology, University of Lausanne, CH-1015 Lausanne, Switzerland
- Center for Primary Care and Public Health, University of Lausanne, CH-1010 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Orsolya Liska
- HCEMM-BRC Metabolic Systems Biology Lab, H-6726 Szeged, Hungary
- Synthetic and Systems Biology Unit, National Laboratory of Biotechnology, Institute of Biochemistry, Biological Research Centre, HUN-REN, H-6726 Szeged, Hungary
- Doctoral School of Biology, University of Szeged, H-6726 Szeged, Hungary
| | - Balázs Papp
- HCEMM-BRC Metabolic Systems Biology Lab, H-6726 Szeged, Hungary
- Synthetic and Systems Biology Unit, National Laboratory of Biotechnology, Institute of Biochemistry, Biological Research Centre, HUN-REN, H-6726 Szeged, Hungary
- National Laboratory for Health Security, Institute of Biochemistry, Biological Research Centre, HUN-REN, H-6726 Szeged, Hungary
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, CH-1015 Lausanne, Switzerland;
- Center for Primary Care and Public Health, University of Lausanne, CH-1010 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| |
Collapse
|
38
|
Kong L, Yang ZB, Chen XH, Quan XQ, Liu HT, Qiu AP. The causal relationship between triglycerides and myocardial infarction: A two-sample Mendelian randomization. Medicine (Baltimore) 2024; 103:e39595. [PMID: 39287313 PMCID: PMC11404938 DOI: 10.1097/md.0000000000039595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/27/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
The causal relationship between triglycerides and myocardial infarction (MI) was investigated using Mendelian randomization (MR) studies. Triglycerides were the exposure factor, and MI served as the outcome variable. Inverse variance weighting was used as the main analysis method, MR-Egger, and weight median as other analysis methods for MR analysis. In addition, heterogeneity test, level multivariate analysis, and sensitivity analysis were carried out. Inverse variance weighting results showed that the increase in triglyceride level affected the incidence of MI (OR = 1.287; 95% CI = 1.185-1.398; P = 1.988 × 10-9). Consistently, the results from all 3 methods indicated a statistically significant increase in the risk of MI with higher triglyceride levels (P < .05). The results showed that patients with high triglyceride levels had a higher incidence of MI, suggesting that MI should be prevented in the high triglyceride population.
Collapse
Affiliation(s)
- Lu Kong
- Department of Geriatrics, Shenzhen Longhua District Central Hospital, Shenzhen, China
- Key Laboratory of Personalized Precision Treatment for Elderly Coronary Heart Disease, Longhua District, Shenzhen, China
| | - Zhong-Bin Yang
- School of Stomatology, Hubei University of Medicine, Shiyan, China
| | - Xie-Hui Chen
- Department of Geriatrics, Shenzhen Longhua District Central Hospital, Shenzhen, China
- Key Laboratory of Personalized Precision Treatment for Elderly Coronary Heart Disease, Longhua District, Shenzhen, China
| | - Xiao-Qing Quan
- Department of Geriatrics, Shenzhen Longhua District Central Hospital, Shenzhen, China
- Key Laboratory of Personalized Precision Treatment for Elderly Coronary Heart Disease, Longhua District, Shenzhen, China
| | - Hong-Tao Liu
- Department of Cardiology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Ai-Ping Qiu
- Department of Geriatrics, Shenzhen Longhua District Central Hospital, Shenzhen, China
- Key Laboratory of Personalized Precision Treatment for Elderly Coronary Heart Disease, Longhua District, Shenzhen, China
| |
Collapse
|
39
|
Zhang W, Lan F, Zhou Q, Gu S, Li X, Wen C, Yang N, Sun C. Host genetics and gut microbiota synergistically regulate feed utilization in egg-type chickens. J Anim Sci Biotechnol 2024; 15:123. [PMID: 39245742 PMCID: PMC11382517 DOI: 10.1186/s40104-024-01076-7] [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: 04/03/2024] [Accepted: 07/14/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Feed efficiency is a crucial economic trait in poultry industry. Both host genetics and gut microbiota influence feed efficiency. However, the associations between gut microbiota and host genetics, as well as their combined contributions to feed efficiency in laying hens during the late laying period, remain largely unclear. METHODS In total, 686 laying hens were used for whole-genome resequencing and liver transcriptome sequencing. 16S rRNA gene sequencing was conducted on gut chyme (duodenum, jejunum, ileum, and cecum) and fecal samples from 705 individuals. Bioinformatic analysis was performed by integrating the genome, transcriptome, and microbiome to screen for key genetic variations, genes, and gut microbiota associated with feed efficiency. RESULTS The heritability of feed conversion ratio (FCR) and residual feed intake (RFI) was determined to be 0.28 and 0.48, respectively. The ileal and fecal microbiota accounted for 15% and 10% of the FCR variance, while the jejunal, cecal, and fecal microbiota accounted for 20%, 11%, and 10% of the RFI variance. Through SMR analysis based on summary data from liver eQTL mapping and GWAS, we further identified four protein-coding genes, SUCLA2, TNFSF13B, SERTM1, and MARVELD3, that influence feed efficiency in laying hens. The SUCLA2 and TNFSF13B genes were significantly associated with SNP 1:25664581 and SNP rs312433097, respectively. SERTM1 showed significant associations with rs730958360 and 1:33542680 and is a potential causal gene associated with the abundance of Corynebacteriaceae in feces. MARVELD3 was significantly associated with the 1:135348198 and was significantly correlated with the abundance of Enterococcus in ileum. Specifically, a lower abundance of Enterococcus in ileum and a higher abundance of Corynebacteriaceae in feces were associated with better feed efficiency. CONCLUSIONS This study confirms that both host genetics and gut microbiota can drive variations in feed efficiency. A small portion of the gut microbiota often interacts with host genes, collectively enhancing feed efficiency. Therefore, targeting both the gut microbiota and host genetic variation by supporting more efficient taxa and selective breeding could improve feed efficiency in laying hens during the late laying period.
Collapse
Affiliation(s)
- Wenxin Zhang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Fangren Lan
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Qianqian Zhou
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
40
|
Fang Q, Zhang J. Dissecting the causal relationship between moderate to vigorous physical activity levels and cognitive performance: a bidirectional two-sample Mendelian randomization study. Front Psychol 2024; 15:1368241. [PMID: 39309156 PMCID: PMC11412864 DOI: 10.3389/fpsyg.2024.1368241] [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: 01/10/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Recent studies increasingly suggest that moderate-to-vigorous physical activity (MVPA) impacts cognitive risk. However, the bidirectional nature of this relationship warrants further exploration. To address this, we employed a Mendelian randomization (MR) approach, analyzing two distinct samples. Methods These analyses utilized published genome-wide association study (GWAS) summary statistics for MVPA (n = 377,234) and cognitive performance (n = 257,841). Our primary method was the inverse variance weighted (IVW) model with random effects, aiming to deduce potential causal links. Additionally, we employed supplementary methods, including MR Egger regression, Weighted median, Weighted mode, and Simple mode. For sensitivity analysis, tools like the MR Egger test, Cochran's Q, MR PRESSO, and leave-one-out (LOO) were utilized. Results Our findings indicate a decrease in cognitive risk with increased MVPA (Odds Ratio [OR] = 0.577, 95% Confidence Interval [CI]: 0.460-0.723, p = 1.930 × 10-6). Furthermore, enhanced cognitive levels corresponded to a reduced risk of inadequate MVPA (OR = 0.866, 95% CI: 0.839-0.895, p = 1.200 × 10-18). Discussion In summary, our study demonstrates that MVPA lowers cognitive risk, while poor cognitive health may impede participation in MVPA. Overall, these findings provide valuable insights for developing personalized prevention and intervention strategies in health and sports sciences.
Collapse
Affiliation(s)
- Qi Fang
- Chengdu Sport University, Chengdu, Sichuan, China
| | - Jinmin Zhang
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
| |
Collapse
|
41
|
Kontou PI, Bagos PG. The goldmine of GWAS summary statistics: a systematic review of methods and tools. BioData Min 2024; 17:31. [PMID: 39238044 PMCID: PMC11375927 DOI: 10.1186/s13040-024-00385-x] [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: 02/09/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
Genome-wide association studies (GWAS) have revolutionized our understanding of the genetic architecture of complex traits and diseases. GWAS summary statistics have become essential tools for various genetic analyses, including meta-analysis, fine-mapping, and risk prediction. However, the increasing number of GWAS summary statistics and the diversity of software tools available for their analysis can make it challenging for researchers to select the most appropriate tools for their specific needs. This systematic review aims to provide a comprehensive overview of the currently available software tools and databases for GWAS summary statistics analysis. We conducted a comprehensive literature search to identify relevant software tools and databases. We categorized the tools and databases by their functionality, including data management, quality control, single-trait analysis, and multiple-trait analysis. We also compared the tools and databases based on their features, limitations, and user-friendliness. Our review identified a total of 305 functioning software tools and databases dedicated to GWAS summary statistics, each with unique strengths and limitations. We provide descriptions of the key features of each tool and database, including their input/output formats, data types, and computational requirements. We also discuss the overall usability and applicability of each tool for different research scenarios. This comprehensive review will serve as a valuable resource for researchers who are interested in using GWAS summary statistics to investigate the genetic basis of complex traits and diseases. By providing a detailed overview of the available tools and databases, we aim to facilitate informed tool selection and maximize the effectiveness of GWAS summary statistics analysis.
Collapse
Affiliation(s)
| | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131, Lamia, Greece.
| |
Collapse
|
42
|
Li C, Chen K, Fang Q, Shi S, Nan J, He J, Yin Y, Li X, Li J, Hou L, Hu X, Kellis M, Han X, Xiong X. Crosstalk between epitranscriptomic and epigenomic modifications and its implication in human diseases. CELL GENOMICS 2024; 4:100605. [PMID: 38981476 PMCID: PMC11406187 DOI: 10.1016/j.xgen.2024.100605] [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: 01/10/2024] [Revised: 04/17/2024] [Accepted: 06/14/2024] [Indexed: 07/11/2024]
Abstract
Crosstalk between N6-methyladenosine (m6A) and epigenomes is crucial for gene regulation, but its regulatory directionality and disease significance remain unclear. Here, we utilize quantitative trait loci (QTLs) as genetic instruments to delineate directional maps of crosstalk between m6A and two epigenomic traits, DNA methylation (DNAme) and H3K27ac. We identify 47 m6A-to-H3K27ac and 4,733 m6A-to-DNAme and, in the reverse direction, 106 H3K27ac-to-m6A and 61,775 DNAme-to-m6A regulatory loci, with differential genomic location preference observed for different regulatory directions. Integrating these maps with complex diseases, we prioritize 20 genome-wide association study (GWAS) loci for neuroticism, depression, and narcolepsy in brain; 1,767 variants for asthma and expiratory flow traits in lung; and 249 for coronary artery disease, blood pressure, and pulse rate in muscle. This study establishes disease regulatory paths, such as rs3768410-DNAme-m6A-asthma and rs56104944-m6A-DNAme-hypertension, uncovering locus-specific crosstalk between m6A and epigenomic layers and offering insights into regulatory circuits underlying human diseases.
Collapse
Affiliation(s)
- Chengyu Li
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Kexuan Chen
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Qianchen Fang
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Shaohui Shi
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Jiuhong Nan
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Jialin He
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Yafei Yin
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Xiaoyu Li
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Jingyun Li
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Lei Hou
- Department of Medicine, Biomedical Genetics Section, Boston University, Boston, MA 02118, USA
| | - Xinyang Hu
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China; The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xikun Han
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Xushen Xiong
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China.
| |
Collapse
|
43
|
Zhang CY, Jiang SJ, Cao JJ, Xu Y, Wang XY, Li R, Miao ZW. Investigating the causal relationship between gut microbiota and gastroenteropancreatic neuroendocrine neoplasms: a bidirectional Mendelian randomization study. Front Microbiol 2024; 15:1420167. [PMID: 39193433 PMCID: PMC11347282 DOI: 10.3389/fmicb.2024.1420167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Background The interaction between the intestinal flora and gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) remains poorly understood, despite the known effect of the gut microbiota on gastrointestinal adenocarcinomas. Hence, the present research aimed to determine the potential causal correlation between the intestinal flora and GEP-NENs by conducting a bidirectional Mendelian randomization (MR) analysis. Methods Two-sample MR analysis was conducted using the summary statistics of the gut microbiota from the MiBioGen consortium and those of GEP-NENs from the FinnGen research project. The inverse-variance weighted approach was utilized as the primary analytical method. To enhance the robustness of our findings, multiple sensitivity tests were performed, including Cochran's Q test for evaluating heterogeneity, the MR-Egger intercept test to detect horizontal pleiotropy, and the MR-PRESSO test to identify outliers and assess pleiotropy bias. Additionally, a leave-one-out analysis was performed to validate the consistency of our findings. The MR-Steiger test was also utilized to determine the causal direction in the correlation between the gut microbiota and GEP-NENs. Finally, a reverse MR analysis was performed to assess reverse causality between the intestinal flora and GEP-NENs. Results We identified 42 taxa of the gut microbiota that were potentially causally associated with GEP-NENs; of these taxa, 7, 8, 11, and 16 taxa were causally associated with pancreatic NENs, colorectal NENs, small intestinal NENs, and gastric NENs, respectively. After adjusting for false discovery rate (FDR) correction, we found significant causal links of Euryarchaeota with small intestinal NENs and Family XIII UCG-001 with gastric NENs. The sensitivity analyses confirmed the stability of these correlations. In the reverse MR analysis, colorectal NENs and small intestinal NENs were found to be associated with variations in 8 and 6 different taxa of the gut microbiota, respectively. After adjusting for FDR correction, no significant causal links were detected between GEP-NENs and the intestinal flora. Conclusion The present study reveals a potential causal association between certain taxa of the intestinal flora and GEP-NENs, thus providing new perspectives regarding the role of the intestinal flora in the development of these tumors. These insights could provide innovative approaches to screen and prevent these diseases.
Collapse
Affiliation(s)
- Chun-yu Zhang
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Jing-jing Cao
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Xu
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao-yu Wang
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Li
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi-wei Miao
- Zhangjiagang Hospital of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
44
|
Bledsoe X, Gamazon ER. A transcriptomic atlas of the human brain reveals genetically determined aspects of neuropsychiatric health. Am J Hum Genet 2024; 111:1559-1572. [PMID: 38925120 PMCID: PMC11339608 DOI: 10.1016/j.ajhg.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Regulation of gene expression is a vital component of neurological homeostasis. Cataloging the consequences of endogenous gene expression on the physical structure and connectivity of the brain offers a means of unifying trait-associated genetic variation with trait-associated neurological features. We perform tissue-specific transcriptome-wide association studies (TWASs) on over 3,400 neuroimaging phenotypes in the UK Biobank (N = 33,224) using our joint-tissue imputation (JTI)-TWAS method. We identify highly significant associations between predicted expression for 7,192 genes and a wide variety of measures of the brain derived from magnetic resonance imaging (MRI). Our approach generates reproducible results in internal and external replication datasets. Genetically determined expression alone is sufficient for high-fidelity reconstruction of brain structure and organization. We demonstrate complementary benefits of cross-tissue and single-tissue analyses toward an integrated neurobiology and provide evidence that gene expression outside the central nervous system provides unique insights into brain health. As an application, we provide evidence suggesting that the genetically regulated expression of schizophrenia risk genes causally affects over 73% of neurological phenotypes that are altered in individuals with schizophrenia (as identified by neuroimaging studies). Imaging features associated with neuropsychiatric traits can provide valuable insights into underlying pathophysiology. By linking neuroimaging-derived phenotypes with expression levels of specific genes, this resource represents a powerful gene prioritization schema that can improve our understanding of brain function, development, and disease. The use of multiple different cortical and subcortical atlases in the resource facilitates direct integration of these data with findings from a diverse range of clinical neuroimaging studies.
Collapse
Affiliation(s)
- Xavier Bledsoe
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Memory & Alzheimer's Center, Nashville, TN, USA.
| |
Collapse
|
45
|
Oreshkov S, Lepik K, Santoni F. pyTWMR: transcriptome-wide Mendelian randomization in python. Bioinformatics 2024; 40:btae505. [PMID: 39128017 PMCID: PMC11341121 DOI: 10.1093/bioinformatics/btae505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/08/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024] Open
Abstract
MOTIVATION Mendelian randomization (MR) is a widely used approach to estimate causal effect of variation in gene expression on complex traits. Among several MR-based algorithms, transcriptome-wide summary statistics-based Mendelian Randomization approach (TWMR) enables the uses of multiple SNPs as instruments and multiple gene expression traits as exposures to facilitate causal inference in observational studies. RESULTS Here we present a Python-based implementation of TWMR and revTWMR. Our implementation offers GPU computational support for faster computations and robust computation mode resilient to highly correlated gene expressions and genetic variants. AVAILABILITY AND IMPLEMENTATION pyTWMR is available at github.com/soreshkov/pyTWMR.
Collapse
Affiliation(s)
- Sergey Oreshkov
- Endocrine, Diabetes and Metabolism Service, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne 1005, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Kaido Lepik
- Department of Computational Biology, University of Lausanne, Lausanne 1015, Switzerland
- University Center for Primary Care and Public Health, University of Lausanne, Lausanne 1005, Switzerland
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Federico Santoni
- Endocrine, Diabetes and Metabolism Service, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne 1005, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
- Institute for Genetic and Biomedical Research (IRGB) - CNR, Monserrato 09042, Italy
| |
Collapse
|
46
|
Khan A, Unlu G, Lin P, Liu Y, Kilic E, Kenny TC, Birsoy K, Gamazon ER. Metabolic gene function discovery platform GeneMAP identifies SLC25A48 as necessary for mitochondrial choline import. Nat Genet 2024; 56:1614-1623. [PMID: 38977856 PMCID: PMC11887816 DOI: 10.1038/s41588-024-01827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 06/10/2024] [Indexed: 07/10/2024]
Abstract
Organisms maintain metabolic homeostasis through the combined functions of small-molecule transporters and enzymes. While many metabolic components have been well established, a substantial number remains without identified physiological substrates. To bridge this gap, we have leveraged large-scale plasma metabolome genome-wide association studies (GWAS) to develop a multiomic Gene-Metabolite Association Prediction (GeneMAP) discovery platform. GeneMAP can generate accurate predictions and even pinpoint genes that are distant from the variants implicated by GWAS. In particular, our analysis identified solute carrier family 25 member 48 (SLC25A48) as a genetic determinant of plasma choline levels. Mechanistically, SLC25A48 loss strongly impairs mitochondrial choline import and synthesis of its downstream metabolite betaine. Integrative rare variant and polygenic score analyses in UK Biobank provide strong evidence that the SLC25A48 causal effects on human disease may in part be mediated by the effects of choline. Altogether, our study provides a discovery platform for metabolic gene function and proposes SLC25A48 as a mitochondrial choline transporter.
Collapse
Affiliation(s)
- Artem Khan
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Gokhan Unlu
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Phillip Lin
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yuyang Liu
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Ece Kilic
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA.
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
47
|
Lee SHT, Garske KM, Arasu UT, Kar A, Miao Z, Alvarez M, Koka A, Darci-Maher N, Benhammou JN, Pan DZ, Örd T, Kaminska D, Männistö V, Heinonen S, Wabitsch M, Laakso M, Agopian VG, Pisegna JR, Pietiläinen KH, Pihlajamäki J, Kaikkonen MU, Pajukanta P. Single nucleus RNA-sequencing integrated into risk variant colocalization discovers 17 cell-type-specific abdominal obesity genes for metabolic dysfunction-associated steatotic liver disease. EBioMedicine 2024; 106:105232. [PMID: 38991381 PMCID: PMC11663762 DOI: 10.1016/j.ebiom.2024.105232] [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: 03/22/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Abdominal obesity increases the risk for non-alcoholic fatty liver disease (NAFLD), now known as metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS To elucidate the directional cell-type level biological mechanisms underlying the association between abdominal obesity and MASLD, we integrated adipose and liver single nucleus RNA-sequencing and bulk cis-expression quantitative trait locus (eQTL) data with the UK Biobank genome-wide association study (GWAS) data using colocalization. Then we used colocalized cis-eQTL variants as instrumental variables in Mendelian randomization (MR) analyses, followed by functional validation experiments on the target genes of the cis-eQTL variants. FINDINGS We identified 17 colocalized abdominal obesity GWAS variants, regulating 17 adipose cell-type marker genes. Incorporating these 17 variants into MR discovers a putative tissue-of-origin, cell-type-aware causal effect of abdominal obesity on MASLD consistently with multiple MR methods without significant evidence for pleiotropy or heterogeneity. Single cell data confirm the adipocyte-enriched mean expression of the 17 genes. Our cellular experiments across human adipogenesis identify risk variant -specific epigenetic and transcriptional mechanisms. Knocking down two of the 17 genes, PPP2R5A and SH3PXD2B, shows a marked decrease in adipocyte lipidation and significantly alters adipocyte function and adipogenesis regulator genes, including DGAT2, LPL, ADIPOQ, PPARG, and SREBF1. Furthermore, the 17 genes capture a characteristic MASLD expression signature in subcutaneous adipose tissue. INTERPRETATION Overall, we discover a significant cell-type level effect of abdominal obesity on MASLD and trace its biological effect to adipogenesis. FUNDING NIH grants R01HG010505, R01DK132775, and R01HL170604; the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (Grant No. 802825), Academy of Finland (Grants Nos. 333021), the Finnish Foundation for Cardiovascular Research the Sigrid Jusélius Foundation and the Jane and Aatos Erkko Foundation; American Association for the Study of Liver Diseases (AASLD) Advanced Transplant Hepatology award and NIH/NIDDK (P30DK41301) Pilot and Feasibility award; NIH/NIEHS F32 award (F32ES034668); Finnish Diabetes Research Foundation, Kuopio University Hospital Project grant (EVO/VTR grants 2005-2021), the Academy of Finland grant (Contract no. 138006); Academy of Finland (Grant Nos 335443, 314383, 272376 and 266286), Sigrid Jusélius Foundation, Finnish Medical Foundation, Finnish Diabetes Research Foundation, Novo Nordisk Foundation (#NNF20OC0060547, NNF17OC0027232, NNF10OC1013354) and Government Research Funds to Helsinki University Hospital; Orion Research Foundation, Maud Kuistila Foundation, Finish Medical Foundation, and University of Helsinki.
Collapse
Affiliation(s)
- Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Uma Thanigai Arasu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nicholas Darci-Maher
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jihane N Benhammou
- Vatche and Tamar Manoukian Division of Digestive Diseases and Gastroenterology, Hepatology and Parenteral Nutrition, David Geffen School of Medicine at UCLA and VA Greater Los Angeles HCS, Los Angeles, CA, USA
| | - David Z Pan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Tiit Örd
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Ville Männistö
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland; Department of Internal Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Vatche G Agopian
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joseph R Pisegna
- Department of Medicine and Human Genetics, Division of Gastroenterology, Hepatology and Parenteral Nutrition, David Geffen School of Medicine at UCLA and VA Greater Los Angeles HCS, Los Angeles, CA, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Healthy WeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Minna U Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
48
|
Qi T, Song L, Guo Y, Chen C, Yang J. From genetic associations to genes: methods, applications, and challenges. Trends Genet 2024; 40:642-667. [PMID: 38734482 DOI: 10.1016/j.tig.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024]
Abstract
Genome-wide association studies (GWASs) have identified numerous genetic loci associated with human traits and diseases. However, pinpointing the causal genes remains a challenge, which impedes the translation of GWAS findings into biological insights and medical applications. In this review, we provide an in-depth overview of the methods and technologies used for prioritizing genes from GWAS loci, including gene-based association tests, integrative analysis of GWAS and molecular quantitative trait loci (xQTL) data, linking GWAS variants to target genes through enhancer-gene connection maps, and network-based prioritization. We also outline strategies for generating context-dependent xQTL data and their applications in gene prioritization. We further highlight the potential of gene prioritization in drug repurposing. Lastly, we discuss future challenges and opportunities in this field.
Collapse
Affiliation(s)
- Ting Qi
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China.
| | - Liyang Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Yazhou Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Chang Chen
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Jian Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China; School of Life Sciences, Westlake University, Hangzhou 310024, China.
| |
Collapse
|
49
|
Çelik MH, Gagneur J, Lim RG, Wu J, Thompson LM, Xie X. Identifying dysregulated regions in amyotrophic lateral sclerosis through chromatin accessibility outliers. HGG ADVANCES 2024; 5:100318. [PMID: 38872308 PMCID: PMC11260578 DOI: 10.1016/j.xhgg.2024.100318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024] Open
Abstract
The high heritability of amyotrophic lateral sclerosis (ALS) contrasts with its low molecular diagnosis rate post-genetic testing, pointing to potential undiscovered genetic factors. To aid the exploration of these factors, we introduced EpiOut, an algorithm to identify chromatin accessibility outliers that are regions exhibiting divergent accessibility from the population baseline in a single or few samples. Annotation of accessible regions with histone chromatin immunoprecipitation sequencing and Hi-C indicates that outliers are concentrated in functional loci, especially among promoters interacting with active enhancers. Across different omics levels, outliers are robustly replicated, and chromatin accessibility outliers are reliable predictors of gene expression outliers and aberrant protein levels. When promoter accessibility does not align with gene expression, our results indicate that molecular aberrations are more likely to be linked to post-transcriptional regulation rather than transcriptional regulation. Our findings demonstrate that the outlier detection paradigm can uncover dysregulated regions in rare diseases. EpiOut is available at github.com/uci-cbcl/EpiOut.
Collapse
Affiliation(s)
- Muhammed Hasan Çelik
- Department of Computer Science, University of California Irvine, Irvine, CA, USA; Center for Complex Biological Systems, University of California Irvine, Irvine, CA, USA
| | - Julien Gagneur
- Department of Informatics, Technical University of Munich, Garching, Germany; Helmholtz Association - Munich School for Data Science (MUDS), Munich, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany; Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Ryan G Lim
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Leslie M Thompson
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA; UCI MIND, University of California Irvine, Irvine, CA, USA; Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Xiaohui Xie
- Department of Computer Science, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
50
|
Hudgins AD, Zhou S, Arey RN, Rosenfeld MG, Murphy CT, Suh Y. A systems biology-based identification and in vivo functional screening of Alzheimer's disease risk genes reveal modulators of memory function. Neuron 2024; 112:2112-2129.e4. [PMID: 38692279 PMCID: PMC11223975 DOI: 10.1016/j.neuron.2024.04.009] [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: 07/16/2022] [Revised: 10/18/2023] [Accepted: 04/08/2024] [Indexed: 05/03/2024]
Abstract
Genome-wide association studies (GWASs) have uncovered over 75 genomic loci associated with risk for late-onset Alzheimer's disease (LOAD), but identification of the underlying causal genes remains challenging. Studies of induced pluripotent stem cell (iPSC)-derived neurons from LOAD patients have demonstrated the existence of neuronal cell-intrinsic functional defects. Here, we searched for genetic contributions to neuronal dysfunction in LOAD using an integrative systems approach that incorporated multi-evidence-based gene mapping and network-analysis-based prioritization. A systematic perturbation screening of candidate risk genes in Caenorhabditis elegans (C. elegans) revealed that neuronal knockdown of the LOAD risk gene orthologs vha-10 (ATP6V1G2), cmd-1 (CALM3), amph-1 (BIN1), ephx-1 (NGEF), and pho-5 (ACP2) alters short-/intermediate-term memory function, the cognitive domain affected earliest during LOAD progression. These results highlight the impact of LOAD risk genes on evolutionarily conserved memory function, as mediated through neuronal endosomal dysfunction, and identify new targets for further mechanistic interrogation.
Collapse
Affiliation(s)
- Adam D Hudgins
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Shiyi Zhou
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Rachel N Arey
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Michael G Rosenfeld
- Department of Medicine, School of Medicine, University of California, La Jolla, CA, USA; Howard Hughes Medical Institute, University of California, La Jolla, CA, USA
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; LSI Genomics, Princeton University, Princeton, NJ, USA.
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|