1
|
Mews MA, Naj AC, Griswold AJ, Below JE, Bush WS. Brain and blood transcriptome-wide association studies identify five novel genes associated with Alzheimer's disease. J Alzheimers Dis 2025; 105:228-244. [PMID: 40111921 DOI: 10.1177/13872877251326288] [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: 03/22/2025]
Abstract
BackgroundGenome-wide association studies (GWAS) have identified numerous genetic variants associated with Alzheimer's disease (AD), but their functional implications remain unclear. Transcriptome-wide association studies (TWAS) offer enhanced statistical power by analyzing genetic associations at the gene level rather than at the variant level, enabling assessment of how genetically-regulated gene expression influences AD risk. However, previous AD-TWAS have been limited by small expression quantitative trait loci (eQTL) reference datasets or reliance on AD-by-proxy phenotypes.ObjectiveTo perform the most powerful AD-TWAS to date using summary statistics from the largest available brain and blood cis-eQTL meta-analyses applied to the largest clinically-adjudicated AD GWAS.MethodsWe implemented the OTTERS TWAS pipeline to predict gene expression using the largest available cis-eQTL data from cortical brain tissue (MetaBrain; N = 2683) and blood (eQTLGen; N = 31,684), and then applied these models to AD-GWAS data (Cases = 21,982; Controls = 44,944).ResultsWe identified and validated five novel gene associations in cortical brain tissue (PRKAG1, C3orf62, LYSMD4, ZNF439, SLC11A2) and six genes proximal to known AD-related GWAS loci (Blood: MYBPC3; Brain: MTCH2, CYB561, MADD, PSMA5, ANXA11). Further, using causal eQTL fine-mapping, we generated sparse models that retained the strength of the AD-TWAS association for MTCH2, MADD, ZNF439, CYB561, and MYBPC3.ConclusionsOur comprehensive AD-TWAS discovered new gene associations and provided insights into the functional relevance of previously associated variants, which enables us to further understand the genetic architecture underlying AD risk.
Collapse
Affiliation(s)
- Makaela A Mews
- System Biology and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Adam C Naj
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute and Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
2
|
Xu Z, Lin Q, Cai X, Zhong Z, Teng J, Li B, Zeng H, Gao Y, Cai Z, Wang X, Shi L, Wang X, Wang Y, Zhang Z, Lin Y, Liu S, Yin H, Bai Z, Wei C, Zhou J, Zhang W, Zhang X, Shi S, Wu J, Diao S, Liu Y, Pan X, Feng X, Liu R, Su Z, Chang C, Zhu Q, Wu Y, Zhou Z, Bai L, Li K, Wang Q, Pan Y, Xu Z, Peng X, Mei S, Mo D, Liu X, Zhang H, Yuan X, Liu Y, Liu GE, Su G, Sahana G, Lund MS, Ma L, Xiang R, Shen X, Li P, Huang R, Ballester M, Crespo-Piazuelo D, Amills M, Clop A, Karlskov-Mortensen P, Fredholm M, Tang G, Li M, Li X, Ding X, Li J, Chen Y, Zhang Q, Zhao Y, Zhao F, Fang L, Zhang Z. Integrating large-scale meta-GWAS and PigGTEx resources to decipher the genetic basis of 232 complex traits in pigs. Natl Sci Rev 2025; 12:nwaf048. [PMID: 40330097 PMCID: PMC12051865 DOI: 10.1093/nsr/nwaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/13/2025] [Accepted: 01/26/2025] [Indexed: 05/08/2025] Open
Abstract
Understanding the molecular and cellular mechanisms underlying complex traits in pigs is crucial for enhancing genetic gain via artificial selection and utilizing pigs as models for human disease and biology. Here, we conducted comprehensive genome-wide association studies (GWAS) followed by a cross-breed meta-analysis for 232 complex traits and a within-breed meta-analysis for 12 traits, using 28.3 million imputed sequence variants in 70 328 animals across 14 pig breeds. We identified 6878 quantitative trait loci (QTL) for 139 complex traits. Leveraging the Pig Genotype-Tissue Expression resource, we systematically investigated the biological context and regulatory mechanisms behind these trait-QTLs, ultimately prioritizing 14 829 variant-gene-tissue-trait regulatory circuits. For instance, rs344053754 regulates UGT2B31 expression in the liver and intestines, potentially by modulating enhancer activity, ultimately influencing litter weight at weaning in pigs. Furthermore, we observed conservation of certain genetic and regulatory mechanisms underlying complex traits between humans and pigs. Overall, our cross-breed meta-GWAS in pigs provides invaluable resources and novel insights into the genetic regulatory and evolutionary mechanisms of complex traits in mammals.
Collapse
Affiliation(s)
- Zhiting Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qing Lin
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaodian Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhanming Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jinyan Teng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Bingjie Li
- Department of Animal and Veterinary Sciences, The Roslin Institute Building, Scotland's Rural College (SRUC), Easter Bush, Midlothian EH25 9RG, UK
| | - Haonan Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yahui Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service (ARS), U.S.Department of Agriculture (USDA), Beltsville, Maryland 20705, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland 20742, USA
| | - Zexi Cai
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus 8000, Denmark
| | - Xiaoqing Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Liangyu Shi
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xue Wang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yi Wang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zipeng Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yu Lin
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Shuli Liu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Hongwei Yin
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Zhonghao Bai
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus 8000, Denmark
| | - Chen Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wenjing Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoke Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shaolei Shi
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shuqi Diao
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yuqiang Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiangchun Pan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xueyan Feng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Ruiqi Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhanqin Su
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Chengjie Chang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianghui Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yuwei Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | | | - Zhongyin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Lijing Bai
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Kui Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Qishan Wang
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuchun Pan
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhong Xu
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Provincial Academy of Agricultural Sciences, Wuhan 430064, China
| | - Xianwen Peng
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Provincial Academy of Agricultural Sciences, Wuhan 430064, China
| | - Shuqi Mei
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Provincial Academy of Agricultural Sciences, Wuhan 430064, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Hao Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaolong Yuan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yang Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service (ARS), U.S.Department of Agriculture (USDA), Beltsville, Maryland 20705, USA
| | - Guosheng Su
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus 8000, Denmark
| | - Goutam Sahana
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus 8000, Denmark
| | - Mogens Sandø Lund
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus 8000, Denmark
| | - Li Ma
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland 20742, USA
| | - Ruidong Xiang
- Faculty of Veterinary & Agricultural Science, University of Melbourne, Parkville, VIC 3010, Australia
- Agriculture Victoria Research, AgriBio Centre for AgriBiosciences, Bundoora, VIC 3083, Australia
| | - Xia Shen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou 510000, China
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh EH16 4UX, UK
| | - Pinghua Li
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory in Nanjing for Evaluation and Utilization of Livestock and Poultry (Pigs) Resources, Ministry of Agriculture and Rural Areas, Nanjing 210095, China
| | - Ruihua Huang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory in Nanjing for Evaluation and Utilization of Livestock and Poultry (Pigs) Resources, Ministry of Agriculture and Rural Areas, Nanjing 210095, China
| | - Maria Ballester
- Animal Breeding and Genetics Programme, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Torre Marimon, Caldes de Montbui 08140, Spain
| | - Daniel Crespo-Piazuelo
- Animal Breeding and Genetics Programme, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Torre Marimon, Caldes de Montbui 08140, Spain
| | - Marcel Amills
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus de la Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Alex Clop
- Department of Animal Genetics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus de la Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Peter Karlskov-Mortensen
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C 1870, Denmark
| | - Merete Fredholm
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C 1870, Denmark
| | - Guoqing Tang
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingzhou Li
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuewei Li
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiangdong Ding
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiaqi Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Qin Zhang
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an 271018, China
| | - Yunxiang Zhao
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Fuping Zhao
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus 8000, Denmark
| | - Zhe Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
3
|
Bai YS, Wang DL, Lee MC, Wang CC, Fang WH, Chuang SW, Chen YH, Su H, Chen CJ, Su SL. Dissect Gender-Dependent Susceptibility SNPs in Progressive Osteoarthritis Using Regulator Motif Candidate of Genetic Association Strategy (RMCGA). Int J Mol Sci 2025; 26:4117. [PMID: 40362356 PMCID: PMC12071535 DOI: 10.3390/ijms26094117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
The role of gender in osteoarthritis (OA) has been reported. However, knowledge on whether gender-specific regulatory SNPs are determining factors in OA is limited. We aimed to identify susceptible gender-specific SNPs of transcription factor binding sites in OA. We used a modified NF-κB binding motif from an RNA sequencing data-inferred OA-associated upstream regulator to define genome-wide potential NF-κB binding sites, which were aligned to the Taiwan BioBank SNP database to identify susceptible SNPs. A case-control study was conducted to verify SNPs with OA determined by a logistic model. The functional assessment was validated using the Genotype-Tissue Expression Portal database. We collected 533 OA patients and 614 healthy controls. Two of nine novel OA-associated SNPs were identified to be significant. For males, the variant of rs73164856 in the aldose reductase gene enhancer was identified to be a protective factor of severe OA patients [odds ratio (OR): 0.17, 95% confidence interval (CI): 0.04-0.73]. For females, the variant of the rs545654 in the neuronal NOS (nNOS) gene was identified to be a detrimental factor of severe OA patients (OR: 2.07, 95% CI: 1.15-3.73). The gene expression analysis demonstrated a lower expression of the AKR1B15 gene (p = 0.00019) upon the rs73164856 T allele; meanwhile, it showed a higher expression of the nNOS gene (p = 1.2 × 10-17) upon the rs545654 T allele. This study identifies susceptible gender-specific SNPs of NF-κB binding sites in severe OA and validates the RMCGA, which sheds light on genetic determinants by gender in advanced OA.
Collapse
Affiliation(s)
- Yin-Shiuan Bai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (Y.-S.B.); (D.-L.W.)
| | - Ding-Lian Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (Y.-S.B.); (D.-L.W.)
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Meng-Chang Lee
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Chih-Chien Wang
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan;
- School of Medicine, National Defense Medical Center, Taipei 114201, Taiwan;
| | - Wen-Hui Fang
- School of Medicine, National Defense Medical Center, Taipei 114201, Taiwan;
- Department of Family and Community Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Su-Wen Chuang
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Yu-Hsuan Chen
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| | - Hao Su
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan
| | - Cheng-Jung Chen
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan;
- Taichung Veterans General Hospital Chiayi Branch, Chiayi City 60090, Taiwan
| | - Sui-Lung Su
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (Y.-S.B.); (D.-L.W.)
- School of Public Health, National Defense Medical Center, Taipei 114201, Taiwan; (M.-C.L.); (S.-W.C.); (Y.-H.C.); (H.S.)
| |
Collapse
|
4
|
Zhong X, Mitchell R, Billstrand C, Thompson EE, Sakabe NJ, Aneas I, Salamone IM, Gu J, Sperling AI, Schoettler N, Nóbrega MA, He X, Ober C. Integration of functional genomics and statistical fine-mapping systematically characterizes adult-onset and childhood-onset asthma genetic associations. Genome Med 2025; 17:35. [PMID: 40205616 PMCID: PMC11983851 DOI: 10.1186/s13073-025-01459-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: 08/13/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified hundreds of loci underlying adult-onset asthma (AOA) and childhood-onset asthma (COA). However, the causal variants, regulatory elements, and effector genes at these loci are largely unknown. METHODS We performed heritability enrichment analysis to determine relevant cell types for AOA and COA, respectively. Next, we fine-mapped putative causal variants at AOA and COA loci. To improve the resolution of fine-mapping, we integrated ATAC-seq data in blood and lung cell types to annotate variants in candidate cis-regulatory elements (CREs). We then computationally prioritized candidate CREs underlying asthma risk, experimentally assessed their enhancer activity by massively parallel reporter assay (MPRA) in bronchial epithelial cells (BECs) and further validated a subset by luciferase assays. Combining chromatin interaction data and expression quantitative trait loci, we nominated genes targeted by candidate CREs and prioritized effector genes for AOA and COA. RESULTS Heritability enrichment analysis suggested a shared role of immune cells in the development of both AOA and COA while highlighting the distinct contribution of lung structural cells in COA. Functional fine-mapping uncovered 21 and 67 credible sets for AOA and COA, respectively, with only 16% shared between the two. Notably, one-third of the loci contained multiple credible sets. Our CRE prioritization strategy nominated 62 and 169 candidate CREs for AOA and COA, respectively. Over 60% of these candidate CREs showed open chromatin in multiple cell lineages, suggesting their potential pleiotropic effects in different cell types. Furthermore, COA candidate CREs were enriched for enhancers experimentally validated by MPRA in BECs. The prioritized effector genes included many genes involved in immune and inflammatory responses. Notably, multiple genes, including TNFSF4, a drug target undergoing clinical trials, were supported by two independent GWAS signals, indicating widespread allelic heterogeneity. Four out of six selected candidate CREs demonstrated allele-specific regulatory properties in luciferase assays in BECs. CONCLUSIONS We present a comprehensive characterization of causal variants, regulatory elements, and effector genes underlying AOA and COA genetics. Our results supported a distinct genetic basis between AOA and COA and highlighted regulatory complexity at many GWAS loci marked by both extensive pleiotropy and allelic heterogeneity.
Collapse
Affiliation(s)
- Xiaoyuan Zhong
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Robert Mitchell
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Emma E Thompson
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Noboru J Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Isabella M Salamone
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Jing Gu
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Anne I Sperling
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nathan Schoettler
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Marcelo A Nóbrega
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
5
|
Wu Y, Liu J, Zou J, Zhang M, Hu Z, Zeng Y, Dai J, Wei L, Liu S, Liu G, Huang G. Time-series analysis reveals metabolic and transcriptional dynamics during mulberry fruit development and ripening. Int J Biol Macromol 2025; 301:140288. [PMID: 39863218 DOI: 10.1016/j.ijbiomac.2025.140288] [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: 11/21/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Understanding the global transcriptomic and metabolic changes during mulberry growth and development is essential for the enhancing fruit quality and optimizing breeding strategies. By integrating phenotypic, metabolomic, and transcriptomic data across 18 developmental and ripening stages of Da10 mulberry fruit, a global map of gene expression and metabolic changes was generated. Analysis revealed a gradual progression of morphological, metabolic, and transcriptional changes throughout the development and ripening phases. In this study, a new transcriptome transition, which was highly related to stress resistance, was observed after the full ripening stage. Moreover, a novel method was devised by integrating metabolome and phenotypic data to assess fruit quality and determine optimal harvest times early in the supply chain. Phase-specific co-expression networks involved in photosynthesis, quality regulation, and plant immunity were also constructed. Notably, eight flavonoids and six hub genes emerged as potential natural edible coatings or gene-editing targets for mulberry fruit to enhance resistance against biotic and abiotic stress. These findings should facilitate further research on stress resistance, post-harvest management, and sustainable agricultural development.
Collapse
Affiliation(s)
- Yilei Wu
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| | - Jiang Liu
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| | - Jian Zou
- College of Life Science, China West Normal University, Nanchong, Sichuan, China.
| | - Minhui Zhang
- College of Life Science, China West Normal University, Nanchong, Sichuan, China.
| | - Zhou Hu
- College of Life Science, China West Normal University, Nanchong, Sichuan, China.
| | - Yichun Zeng
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| | - Jie Dai
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| | - Ling Wei
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| | - Sanmei Liu
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| | - Gang Liu
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| | - Gaiqun Huang
- Sericultural Research Institute, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China; Institute of Special Economic Animal and Plant, Sichuan Academy of Agricultural Sciences, Nanchong, Sichuan, China.
| |
Collapse
|
6
|
Xu Y, Yang Y, Song H, Li M, Shi W, Yu T, Lin J, Yu Y. The Role of Exerkines in the Treatment of Knee Osteoarthritis: From Mechanisms to Exercise Strategies. Orthop Surg 2025; 17:1021-1035. [PMID: 39854050 PMCID: PMC11962297 DOI: 10.1111/os.14365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
With the increasing prevalence of knee osteoarthritis (KOA), the limitations of traditional treatments, such as their limited efficacy in halting disease progression and their potential side effects, are becoming more evident. This situation has prompted scientists to seek more effective strategies. In recent years, exercise therapy has gained prominence in KOA treatment due to its safety, efficacy, and cost-effectiveness, which are underpinned by the molecular actions of exerkines. Unlike conventional therapies, exerkines offer specific advantages by targeting inflammatory responses, enhancing chondrocyte proliferation, and slowing cartilage degradation at the molecular level. This review explores the potential mechanisms involved in and application prospects of exerkines in KOA treatment and provides a comprehensive analysis of their role. Studies show that appropriate exercise not only promotes overall health, but also positively impacts KOA by stimulating exerkine production. The effectiveness of exerkines, however, is influenced by exercise modality, intensity, and duration of exercise, making the development of personalized exercise plans crucial for KOA patients. Based on these insights, this paper proposes targeted exercise strategies designed to maximize exerkine benefits, aiming to provide novel perspectives for KOA prevention and treatment.
Collapse
Affiliation(s)
- Yuxiong Xu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Yizhuo Yang
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Hanan Song
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Ming Li
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Weihao Shi
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Tongwu Yu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Jianhao Lin
- Arthritis Clinic & Research CenterPeking University People's HospitalBeijingChina
| | - Yanli Yu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| |
Collapse
|
7
|
Wenz BM, He Y, Chen NC, Pickrell JK, Li JH, Dudek MF, Li T, Keener R, Voight BF, Brown CD, Battle A. Genotype inference from aggregated chromatin accessibility data reveals genetic regulatory mechanisms. Genome Biol 2025; 26:81. [PMID: 40159496 PMCID: PMC11956263 DOI: 10.1186/s13059-025-03538-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Understanding the genetic causes underlying variability in chromatin accessibility can shed light on the molecular mechanisms through which genetic variants may affect complex traits. Thousands of ATAC-seq samples have been collected that hold information about chromatin accessibility across diverse cell types and contexts, but most of these are not paired with genetic information and come from distinct projects and laboratories. RESULTS We report here joint genotyping, chromatin accessibility peak calling, and discovery of quantitative trait loci which influence chromatin accessibility (caQTLs), demonstrating the capability of performing caQTL analysis on a large scale in a diverse sample set without pre-existing genotype information. Using 10,293 profiling samples representing 1454 unique donor individuals across 653 studies from public databases, we catalog 24,159 caQTLs in total. After joint discovery analysis, we cluster samples based on accessible chromatin profiles to identify context-specific caQTLs. We find that caQTLs are strongly enriched for annotations of gene regulatory elements across diverse cell types and tissues and are often linked with genetic variation associated with changes in expression (eQTLs), indicating that caQTLs can mediate genetic effects on gene expression. We demonstrate sharing of causal variants for chromatin accessibility across human traits, enabling a more complete picture of the genetic mechanisms underlying complex human phenotypes. CONCLUSIONS Our work provides a proof of principle for caQTL calling from previously ungenotyped samples and represents one of the largest, most diverse caQTL resources currently available, informing mechanisms of genetic regulation of gene expression and contribution to disease.
Collapse
Affiliation(s)
- Brandon M Wenz
- Genetics and Epigenetics Program, Cell and Molecular Biology Graduate Group, Biomedical Graduate Studies, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Yuan He
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Nae-Chyun Chen
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, 21218, USA
| | | | | | - Max F Dudek
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Taibo Li
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Rebecca Keener
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Christopher D Brown
- Department of Genetics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Data Science and AI Institute, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
8
|
Ye Z, Hong D, Yuan J, Xu P, Liu W. Assessing the influence of plasma metabolites on chronic skin ulcer risk: a two-sample Mendelian randomization study. Sci Rep 2025; 15:10001. [PMID: 40121277 PMCID: PMC11929753 DOI: 10.1038/s41598-025-94311-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Chronic skin ulcers, although rare, pose severe and debilitating challenges. The identification of causal metabolite biomarkers presents an opportunity to refine effective risk assessment strategies for this condition. In this study, we conducted a comprehensive Two-Sample Mendelian Randomization (TSMR) investigation to delineate the potential causal effects of plasma metabolites on chronic skin ulcer risk. Exposure data comprised 14,296 participants with 913 metabolites from INTERVAL/EPIC-Norfolk, and 8,299 participants with 1,091 metabolites and 309 ratios from the Canadian Longitudinal Study on Aging (CLSA). Outcome data came from the finngen_R9_L12_CHRONICULCEROFSKIN (1,840 cases, 353,088 controls) and UK Biobank Chronic ulcer of skin (495 cases, 455,853 controls) cohorts. Leveraging the inverse-variance weighted (IVW) method, alongside MR-Egger and MR-PRESSO sensitivity analyses, we evaluated metabolite associations with chronic skin ulcer risk. Further assessment involved a phenome-wide MR (Phe-MR) analysis to explore potential repercussions of targeting identified metabolites for intervention. Our study identified 12 distinct metabolites significantly associated with chronic skin ulcers, demonstrating consistent and replicable results. Notably, X-19,141 exhibited the highest reproducibility. These findings highlight novel plasma metabolites relevant to chronic skin ulcers, offering theoretical underpinnings for mechanistic research and clinical strategies in prevention and treatment.
Collapse
Affiliation(s)
- Zheng Ye
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, 558000, Guizhou, China
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Jiaqi Yuan
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China.
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, 558000, Guizhou, China.
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China.
| | - Wenbin Liu
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China.
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China.
| |
Collapse
|
9
|
Ahmad S, Khan HM, Nawaz A, Samad MA, Cho H, Sarfraz H, Aziz Y, Rouached H, Shahzad Z. Genome-wide association studies and transcriptomics reveal mechanisms explaining the diversity of wheat root responses to nutrient availability. JOURNAL OF EXPERIMENTAL BOTANY 2025; 76:1458-1472. [PMID: 38551389 DOI: 10.1093/jxb/erae141] [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: 02/12/2024] [Accepted: 03/28/2024] [Indexed: 03/15/2025]
Abstract
Nutrient availability profoundly influences plant root system architecture, which critically determines crop productivity. While Arabidopsis has provided important insights into the genetic responses to nutrient deficiency, translating this knowledge to crops, particularly wheat, remains a subject of inquiry. Here, examining a diverse wheat population under varying nitrogen (N), phosphorus (P), potassium (K), and iron (Fe) levels, we uncover a spectrum of root responses, spanning from growth inhibition to stimulation, highlighting genotype-specific strategies. Furthermore, we reveal a nuanced interplay between macronutrient deficiency (N, P, and K) and Fe availability, emphasizing the central role of Fe in modulating root architecture. Through genome-wide association mapping, we identify 11 quantitative trait loci underlying root traits under varying nutrient availabilities, including homologous genes previously validated in Arabidopsis, supporting our findings. In addition, utilizing transcriptomics, reactive oxygen species (ROS) imaging, and antioxidant treatment, we uncover that wheat root growth inhibition by nutrient deficiency is attributed to ROS accumulation, akin to the role of ROS in governing Arabidopsis root responses to nutrient deficiency. Therefore, our study reveals the conservation of molecular and physiological mechanisms between Arabidopsis and wheat to adjust root growth to nutrient availability, paving the way for targeted crop improvement strategies aimed at increasing nutrient use efficiency.
Collapse
Affiliation(s)
- Suhaib Ahmad
- Department of Life Sciences, SBASSE, Lahore University of Management Sciences, Pakistan
| | - Hafiza Madeeha Khan
- Department of Life Sciences, SBASSE, Lahore University of Management Sciences, Pakistan
| | - Amjad Nawaz
- Department of Life Sciences, SBASSE, Lahore University of Management Sciences, Pakistan
| | - Muhammad Abdul Samad
- Department of Life Sciences, SBASSE, Lahore University of Management Sciences, Pakistan
| | - Huikyong Cho
- The Plant Resilience Institute, Michigan State University, East Lansing, MI 48824, USA
- Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Hira Sarfraz
- Department of Life Sciences, SBASSE, Lahore University of Management Sciences, Pakistan
| | - Yasir Aziz
- Department of Life Sciences, SBASSE, Lahore University of Management Sciences, Pakistan
| | - Hatem Rouached
- The Plant Resilience Institute, Michigan State University, East Lansing, MI 48824, USA
- Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Zaigham Shahzad
- Department of Life Sciences, SBASSE, Lahore University of Management Sciences, Pakistan
| |
Collapse
|
10
|
Lee D, Gunamalai L, Kannan J, Vickery K, Yaacov O, Onuchic-Whitford AC, Chakravarti A, Kapoor A. Massively parallel reporter assays identify functional enhancer variants at QT interval GWAS loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642686. [PMID: 40161821 PMCID: PMC11952420 DOI: 10.1101/2025.03.11.642686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Genome-wide association studies (GWAS) have identified >30 loci with multiple common noncoding variants explaining interindividual electrocardiographic QT interval (QTi) variation. Of the many types of noncoding functional elements, here we sought to identify transcriptional enhancers with sequence variation and their cognate transcription factors (TFs) that alter the expression of proximal cardiac genes to affect QTi variation. We used massively parallel reporter assays (MPRA) in mouse cardiomyocyte HL-1 cells to screen for functional enhancer variants among 1,018 QTi-associated GWAS variants that overlap candidate cardiac enhancers across 31 loci. We identified 445 GWAS variant-containing enhancers of which 79 showed significant allelic difference in enhancer activity across 21 GWAS loci, with multiple enhancer variants per locus. Of these, we predicted differential binding by cardiac TFs, including AP-1, ATF-1, GATA2, MEF2, NKX2.5, SRF and TBX5 which are known to play key roles in development and homeostasis, at 49 enhancer variants. Finally, we used expression quantitative trait locus mapping and predicted promoter-enhancer contacts to identify 14 candidate target genes through analyses of 36 enhancer variants at 16 loci. This study provides strong evidence for 14 cardiac genes, 10 of them novel, impacting on QTi variation, beyond explaining observed genetic associations.
Collapse
Affiliation(s)
- Dongwon Lee
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Lavanya Gunamalai
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jeerthi Kannan
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Kyla Vickery
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Or Yaacov
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY, USA
| | - Ana C. Onuchic-Whitford
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Renal division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Aravinda Chakravarti
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY, USA
| | - Ashish Kapoor
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
11
|
Jan SM, Fahira A, Hassan ESG, Abdelhameed AS, Wei D, Wadood A. Integrative approaches to m6A and m5C RNA modifications in autism spectrum disorder revealing potential causal variants. Mamm Genome 2025; 36:280-292. [PMID: 39738578 DOI: 10.1007/s00335-024-10095-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder that currently affects approximately 1-2% of the global population. Genome-wide studies have identified several loci associated with ASD; however, pinpointing causal variants remains elusive. Therefore, functional studies are essential to discover potential therapeutics for ASD. RNA modification plays a crucial role in the post-transcriptional regulation of mRNA, with m6A and m5C being the most prevalent internal modifications. Recent research indicates their involvement in the regulation of key genes associated with ASD. In this study, we conducted an integrative genomic analysis of ASD, incorporating m6A and m5C variants, followed by cis-eQTL, gene differential expression, and gene enrichment analyses to identify causal variants from a genome-wide study of ASD. We identified 20,708 common m6A-SNPs and 2,407 common m5C-SNPs. Among these, 647 m6A-SNPs exhibited cis-eQTL signals with a p-value < 0.05, while only 81 m5C-SNPs with a p-value < 0.05 showed cis-eQTL signals. Most of these were functional loss variants, with 38 variants representing the most significant common m6A/m5C-SNPs associated with key ASD-related genes. In the gene differential expression analysis, seven proximal genes corresponding to significant m6A/m5C-SNPs were differentially expressed in at least one of the three microarray gene expression profiles of ASD. Key differentially expressed genes corresponding to m6A/m5C cis-variants included KIAA1671 (rs5752063, rs12627825), INTS1 (rs67049052, rs10237910), VSIG10 (rs7965350), TJP2 (rs3812536), FAM167A (rs9693108), TMEM8A (rs1802752), and NUP43 (rs3924871, rs7818, rs9383844, rs9767113). Cell-specific cis-eQTL analysis for proximal gene identification, combined with gene expression datasets from single-cell RNA-seq analysis, would validate the causal relationship of gene regulation in brain-specific regions, and experimental validation in cell lines would achieve the goal of precision medicine.
Collapse
Affiliation(s)
- Syed Mansoor Jan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Aamir Fahira
- Key Laboratory of Big Data Mining and Precision Drug, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Design of Guangdong Medical University, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Eman S G Hassan
- Pharmacology Department, Egyptian Drug Authority (EDA), Formerly National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Ali Saber Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Dongqing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
| |
Collapse
|
12
|
Dang X, Teng Z, Yang Y, Li W, Liu J, Hui L, Zhou D, Gong D, Dai SS, Li Y, Li X, Lv L, Zeng Y, Yuan Y, Ma X, Liu Z, Li T, Luo XJ. Gene-level analysis reveals the genetic aetiology and therapeutic targets of schizophrenia. Nat Hum Behav 2025; 9:609-624. [PMID: 39753749 DOI: 10.1038/s41562-024-02091-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/18/2024] [Indexed: 03/27/2025]
Abstract
Genome-wide association studies (GWASs) have reported multiple risk loci for schizophrenia (SCZ). However, the majority of the associations were from populations of European ancestry. Here we conducted a large-scale GWAS in Eastern Asian populations (29,519 cases and 44,392 controls) and identified ten Eastern Asian-specific risk loci, two of which have not been previously reported. A further cross-ancestry GWAS meta-analysis (96,806 cases and 492,818 controls) including populations from diverse ancestries identified 61 previously unreported risk loci. Systematic variant-level analysis, including fine mapping, functional genomics and expression quantitative trait loci, prioritized potential causal variants. Gene-level analyses, including transcriptome-wide association study, proteome-wide association study and Mendelian randomization, nominated the potential causal genes. By integrating evidence from layers of different analyses, we prioritized the most plausible causal genes for SCZ, such as ACE, CNNM2, SNAP91, ABCB9 and GATAD2A. Finally, drug repurposing showed that ACE, CA14, MAPK3 and MAPT are potential therapeutic targets for SCZ. Our study not only showed the power of cross-ancestry GWAS in deciphering the genetic aetiology of SCZ, but also uncovered new genetic risk loci, potential causal variants and genes and therapeutic targets for SCZ.
Collapse
Affiliation(s)
- Xinglun Dang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Zhaowei Teng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Kunming, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Jiewei Liu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, China
| | - Li Hui
- Research Center of Biological Psychiatry, Suzhou Guangji Hospital, Suzhou Medical College of Soochow University, Suzhou, China
| | - Dongsheng Zhou
- Department of Psychiatry, Affiliated Kangning Hospital of Ningbo University (Ningbo Kangning Hospital), Ningbo, China
| | - Daohua Gong
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Shan-Shan Dai
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Yifan Li
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Xingxing Li
- Department of Psychiatry, Affiliated Kangning Hospital of Ningbo University (Ningbo Kangning Hospital), Ningbo, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorders, Xinxiang Medical University, Xinxiang, China
| | - Yong Zeng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Kunming, China
| | - Yonggui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Xiancang Ma
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Tao Li
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xiong-Jian Luo
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China.
| |
Collapse
|
13
|
Kellman LN, Neela PH, Srinivasan S, Siprashvili Z, Shanderson RL, Hong AW, Rao D, Porter DF, Reynolds DL, Meyers RM, Guo MG, Yang X, Zhao Y, Wozniak GG, Donohue LKH, Shenoy R, Ko LA, Nguyen DT, Mondal S, Garcia OS, Elcavage LE, Elfaki I, Abell NS, Tao S, Lopez CM, Montgomery SB, Khavari PA. Functional analysis of cancer-associated germline risk variants. Nat Genet 2025; 57:718-728. [PMID: 39962238 DOI: 10.1038/s41588-024-02070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/20/2024] [Indexed: 03/15/2025]
Abstract
Single-nucleotide variants (SNVs) in regulatory DNA are linked to inherited cancer risk. Massively parallel reporter assays of 4,041 SNVs linked to 13 neoplasms comprising >90% of human malignancies were performed in pertinent primary human cell types and then integrated with matching chromatin accessibility, DNA looping and expression quantitative trait loci data to nominate 380 potentially regulatory SNVs and their putative target genes. The latter highlighted specific protein networks in lifetime cancer risk, including mitochondrial translation, DNA damage repair and Rho GTPase activity. A CRISPR knockout screen demonstrated that a subset of germline putative risk genes also enables the growth of established cancers. Editing one SNV, rs10411210 , showed that its risk allele increases rhophilin RHPN2 expression and stimulus-responsive RhoA activation, indicating that individual SNVs may upregulate cancer-linked pathways. These functional data are a resource for variant prioritization efforts and further interrogation of the mechanisms underlying inherited risk for cancer.
Collapse
Affiliation(s)
- Laura N Kellman
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Poornima H Neela
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Suhas Srinivasan
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald L Shanderson
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Audrey W Hong
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deepti Rao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Douglas F Porter
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - David L Reynolds
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Robin M Meyers
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Margaret G Guo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xue Yang
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yang Zhao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Glenn G Wozniak
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura K H Donohue
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rajani Shenoy
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa A Ko
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Duy T Nguyen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Smarajit Mondal
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Omar S Garcia
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lara E Elcavage
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ibtihal Elfaki
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathan S Abell
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Shiying Tao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher M Lopez
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephen B Montgomery
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA.
| |
Collapse
|
14
|
Webster AK, Willis JH, Johnson E, Sarkies P, Phillips PC. Gene expression variation across genetically identical individuals predicts reproductive traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.13.562270. [PMID: 37873136 PMCID: PMC10592811 DOI: 10.1101/2023.10.13.562270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
In recent decades, genome-wide association studies (GWAS) have been the major approach to understand the biological basis of individual differences in traits and diseases. However, GWAS approaches have limited predictive power to explain individual differences, particularly for complex traits and diseases in which environmental factors play a substantial role in their etiology. Indeed, individual differences persist even in genetically identical individuals, although fully separating genetic and environmental causation is difficult in most organisms. To understand the basis of individual differences in the absence of genetic differences, we measured two quantitative reproductive traits in 180 genetically identical young adult Caenorhabditis elegans roundworms in a shared environment and performed single-individual transcriptomics on each worm. We identified hundreds of genes for which expression variation was strongly associated with reproductive traits, some of which depended on individuals' historical environments and some of which was random. Multiple small sets of genes together were highly predictive of reproductive traits, explaining on average over half and over a quarter of variation in the two traits. We manipulated mRNA levels of predictive genes to identify a set of causal genes, demonstrating the utility of this approach for both prediction and understanding underlying biology. Finally, we found that the chromatin environment of predictive genes was enriched for H3K27 trimethylation, suggesting that gene expression variation may be driven in part by chromatin structure. Together, this work shows that individual, non-genetic differences in gene expression are both highly predictive and causal in shaping reproductive traits.
Collapse
|
15
|
Zhong X, Mitchell R, Billstrand C, Thompson E, Sakabe NJ, Aneas I, Salamone IM, Gu J, Sperling AI, Schoettler N, Nóbrega MA, He X, Ober C. Integration of functional genomics and statistical fine-mapping systematically characterizes adult-onset and childhood-onset asthma genetic associations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322088. [PMID: 40034789 PMCID: PMC11875274 DOI: 10.1101/2025.02.11.25322088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Genome-wide association studies (GWAS) have identified hundreds of loci underlying adult-onset asthma (AOA) and childhood-onset asthma (COA). However, the causal variants, regulatory elements, and effector genes at these loci are largely unknown. Methods We performed heritability enrichment analysis to determine relevant cell types for AOA and COA, respectively. Next, we fine-mapped putative causal variants at AOA and COA loci. To improve the resolution of fine-mapping, we integrated ATAC-seq data in blood and lung cell types to annotate variants in candidate cis-regulatory elements (CREs). We then computationally prioritized candidate CREs underlying asthma risk, experimentally assessed their enhancer activity by massively parallel reporter assay (MPRA) in bronchial epithelial cells (BECs) and further validated a subset by luciferase assays. Combining chromatin interaction data and expression quantitative trait loci, we nominated genes targeted by candidate CREs and prioritized effector genes for AOA and COA. Results Heritability enrichment analysis suggested a shared role of immune cells in the development of both AOA and COA while highlighting the distinct contribution of lung structural cells in COA. Functional fine-mapping uncovered 21 and 67 credible sets for AOA and COA, respectively, with only 16% shared between the two. Notably, one-third of the loci contained multiple credible sets. Our CRE prioritization strategy nominated 62 and 169 candidate CREs for AOA and COA, respectively. Over 60% of these candidate CREs showed open chromatin in multiple cell lineages, suggesting their potential pleiotropic effects in different cell types. Furthermore, COA candidate CREs were enriched for enhancers experimentally validated by MPRA in BECs. The prioritized effector genes included many genes involved in immune and inflammatory responses. Notably, multiple genes, including TNFSF4, a drug target undergoing clinical trials, were supported by two independent GWAS signals, indicating widespread allelic heterogeneity. Four out of six selected candidate CREs demonstrated allele-specific regulatory properties in luciferase assays in BECs. Conclusions We present a comprehensive characterization of causal variants, regulatory elements, and effector genes underlying AOA and COA genetics. Our results supported a distinct genetic basis between AOA and COA and highlighted regulatory complexity at many GWAS loci marked by both extensive pleiotropy and allelic heterogeneity.
Collapse
Affiliation(s)
- Xiaoyuan Zhong
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Robert Mitchell
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Emma Thompson
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Noboru J. Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Jing Gu
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Anne I. Sperling
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nathan Schoettler
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Marcelo A. Nóbrega
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
16
|
Zhou Y, Zhang Y, Xu Q, Sun X, Chen Y. The evaluation of targeted exome sequencing of candidate genes in a Han Chinese population with primary open-angle glaucoma. Hum Mol Genet 2025; 34:435-443. [PMID: 39776193 DOI: 10.1093/hmg/ddae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Primary open-angle glaucoma (POAG), known as a common ocular disease with genetic heterogeneity, is characterized by progressive optic disc atrophy and visual field defects. This study aimed to assess the contribution of previously reported POAG-associated genes and investigate potential functional variations and genotype-phenotype correlations in a Han Chinese population. DNA from 500 cases and 500 controls was pooled and sequenced using a customized panel of 398 candidate genes. After prioritization, 21 SNPs from 16 genes were genotyped in the first replication cohort (500 cases and 500 controls), and 9 SNPs were genotyped in the second replication cohort (500 cases and 500 controls). Allelic associations and odds ratios were adjusted for age and sex, while linear regression assessed SNP correlations with POAG endophenotypes. Haplotype analysis and linkage disequilibrium were performed using Haploview. In silico prediction tools were used to predict pathogenicity and function. SNPs from MFN2, DGKG, PKHD1, PTPRJ, and LTBP2 were associated with POAG in at least one cohort, and SNPs from EXOC2, PTPRJ, and LTBP2 showed significant correlations with intraocular pressure. Additionally, haplotype analysis revealed a significant association between the EXOC2 TGC haplotype and POAG risk. We validated several candidate genes and identified novel SNPs, providing further insight into the genetic architecture of POAG in the Han Chinese population.
Collapse
Affiliation(s)
- Yiwen Zhou
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Youjia Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Qingdan Xu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai 200031, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, 83 Fenyang Road, Xuhui District, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yuhong Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai 200031, China
- Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, 83 Fenyang Road, Xuhui District, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| |
Collapse
|
17
|
Zhang X, Wang L, Zhao J, Zhao H. Knockoff procedure improves causal gene identifications in conditional transcriptome-wide association studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636660. [PMID: 39974930 PMCID: PMC11838583 DOI: 10.1101/2025.02.05.636660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Transcriptome-wide association studies (TWASs) have been developed to nominate candidate genes associated with complex traits by integrating genome-wide association studies (GWASs) with expression quantitative trait loci (eQTL) data. However, most existing TWAS methods evaluate the marginal association between a single gene and the trait of interest without accounting for other genes within the same genomic region or the same gene from different tissues. Additionally, false-positive gene-trait pairs can arise due to correlations with the direct effects of genetic variants. In this study, we introduce TWASKnockoff, a new knockoff-based framework for detecting causal gene-tissue pairs using GWAS summary statistics and eQTL data. Unlike marginal testing in traditional TWAS methods, TWASKnockoff examines the conditional independence for each gene-trait pair, considering both correlations in cis-predicted expression across genes and correlations between gene expression levels and genetic variants. TWASKnockoff estimates the theoretical correlation matrix for all genetic elements (cis-predicted expression across genes and genotypes for genetic variants) by averaging estimations from parametric boot-strap samples and then performs knockoff-based inference to detect causal gene-trait pairs while controlling the false discovery rate (FDR). Through empirical simulations and an application to type 2 diabetes (T2D) data, we demonstrate that TWASKnockoff achieves superior FDR control and improves the average power in detecting causal gene-trait pairs at a fixed FDR level.
Collapse
|
18
|
Xi X, Li J, Jia J, Meng Q, Li C, Wang X, Wei L, Zhang X. A mechanism-informed deep neural network enables prioritization of regulators that drive cell state transitions. Nat Commun 2025; 16:1284. [PMID: 39900922 PMCID: PMC11790924 DOI: 10.1038/s41467-025-56475-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
Cells are regulated at multiple levels, from regulations of individual genes to interactions across multiple genes. Some recent neural network models can connect molecular changes to cellular phenotypes, but their design lacks modeling of regulatory mechanisms, limiting the decoding of regulations behind key cellular events, such as cell state transitions. Here, we present regX, a deep neural network incorporating both gene-level regulation and gene-gene interaction mechanisms, which enables prioritizing potential driver regulators of cell state transitions and providing mechanistic interpretations. Applied to single-cell multi-omics data on type 2 diabetes and hair follicle development, regX reliably prioritizes key transcription factors and candidate cis-regulatory elements that drive cell state transitions. Some regulators reveal potential new therapeutic targets, drug repurposing possibilities, and putative causal single nucleotide polymorphisms. This method to analyze single-cell multi-omics data demonstrates how the interpretable design of neural networks can better decode biological systems.
Collapse
Affiliation(s)
- Xi Xi
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Jiaqi Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Jinmeng Jia
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Qiuchen Meng
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Chen Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Xiaowo Wang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Lei Wei
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST / Department of Automation, Tsinghua University, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
19
|
Lord KA, Chen FL, Karlsson EK. An Evolutionary Perspective on Dog Behavioral Genetics. Annu Rev Anim Biosci 2025; 13:167-188. [PMID: 39413150 DOI: 10.1146/annurev-animal-111523-101954] [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: 10/18/2024]
Abstract
Dogs have played an outsized role in the field of behavioral genetics since its earliest days. Their unique evolutionary history and ubiquity in the modern world make them a potentially powerful model system for discovering how genetic changes lead to changes in behavior. Genomic technology has supercharged this potential by enabling scientists to sequence the DNA of thousands of dogs and test for correlations with behavioral traits. However, fractures in the early history of animal behavior between biological and psychological subfields may be impeding progress. In addition, canine behavioral genetics has included almost exclusively dogs from modern breeds, who represent just a small fraction of all dog diversity. By expanding the scope of dog behavior studies, and incorporating an evolutionary perspective on canine behavioral genetics, we can move beyond associations to understanding the complex interactions between genes and environment that lead to dog behavior.
Collapse
Affiliation(s)
- Kathryn A Lord
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; , ,
- Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Frances L Chen
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; , ,
- Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Elinor K Karlsson
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; , ,
- Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
20
|
Nguyen PT, Coetzee SG, Silacheva I, Hazelett DJ. Genome-wide association studies are enriched for interacting genes. BioData Min 2025; 18:3. [PMID: 39815328 PMCID: PMC11734473 DOI: 10.1186/s13040-024-00421-w] [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: 10/03/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND With recent advances in single cell technology, high-throughput methods provide unique insight into disease mechanisms and more importantly, cell type origin. Here, we used multi-omics data to understand how genetic variants from genome-wide association studies influence development of disease. We show in principle how to use genetic algorithms with normal, matching pairs of single-nucleus RNA- and ATAC-seq, genome annotations, and protein-protein interaction data to describe the genes and cell types collectively and their contribution to increased risk. RESULTS We used genetic algorithms to measure fitness of gene-cell set proposals against a series of objective functions that capture data and annotations. The highest information objective function captured protein-protein interactions. We observed significantly greater fitness scores and subgraph sizes in foreground vs. matching sets of control variants. Furthermore, our model reliably identified known targets and ligand-receptor pairs, consistent with prior studies. CONCLUSIONS Our findings suggested that application of genetic algorithms to association studies can generate a coherent cellular model of risk from a set of susceptibility variants. Further, we showed, using breast cancer as an example, that such variants have a greater number of physical interactions than expected due to chance.
Collapse
Affiliation(s)
- Peter T Nguyen
- The Department of Biomedical and Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Simon G Coetzee
- The Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90069, USA
| | - Irina Silacheva
- The Department of Biomedical and Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Dennis J Hazelett
- The Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90069, USA.
| |
Collapse
|
21
|
Cao C, Tian M, Li Z, Zhu W, Huang P, Yang S. GWAShug: a comprehensive platform for decoding the shared genetic basis between complex traits based on summary statistics. Nucleic Acids Res 2025; 53:D1006-D1015. [PMID: 39380491 PMCID: PMC11701566 DOI: 10.1093/nar/gkae873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
The shared genetic basis offers very valuable insights into the etiology, diagnosis and therapy of complex traits. However, a comprehensive resource providing shared genetic basis using the accessible summary statistics is currently lacking. It is challenging to analyze the shared genetic basis due to the difficulty in selecting parameters and the complexity of pipeline implementation. To address these issues, we introduce GWAShug, a platform featuring a standardized best-practice pipeline with four trait level methods and three molecular level methods. Based on stringent quality control, the GWAShug resource module includes 539 high-quality GWAS summary statistics for European and East Asian populations, covering 54 945 pairs between a measurement-based and a disease-based trait and 43 902 pairs between two disease-based traits. Users can easily search for shared genetic basis information by trait name, MeSH term and category, and access detailed gene information across different trait pairs. The platform facilitates interactive visualization and analysis of shared genetic basic results, allowing users to explore data dynamically. Results can be conveniently downloaded via FTP links. Additionally, we offer an online analysis module that allows users to analyze their own summary statistics, providing comprehensive tables, figures and interactive visualization and analysis. GWAShug is freely accessible at http://www.gwashug.com.
Collapse
Affiliation(s)
- Chen Cao
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Min Tian
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhenghui Li
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wenyan Zhu
- Department of Biostatistics, Centre for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Peng Huang
- Department of Epidemiology, Centre for Global Health, School of Public Health, National Vaccine Innovation Platform, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Sheng Yang
- Department of Biostatistics, Centre for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
22
|
Zhou H, Quach A, Nair M, Abasht B, Kong B, Bowker B. Omics based technology application in poultry meat research. Poult Sci 2025; 104:104643. [PMID: 39662255 PMCID: PMC11697050 DOI: 10.1016/j.psj.2024.104643] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Omics techniques, including genomics, transcriptomics, proteomics, metabolomics, and lipidomics, analyze entire sets of biological molecules to seek comprehensive knowledge on a particular phenotype. These approaches have been extensively utilized to identify both biomarkers and biological mechanisms for various physiological conditions in livestock and poultry. The purpose of this symposium was not only to focus on how recent omics technologies can be used to gather, integrate, and interpret data produced by various methodologies in poultry research, but also to highlight how omics and bioinformatics have increased our understanding of poultry meat quality problems and other complex traits. This Poultry Science Association symposium paper includes 5 sections that cover: 1) functional annotation of cis-regulatory elements in the genome informs genetic control of complex traits in poultry, 2) mass spectrometry for proteomics, metabolomics, and lipidomics, 3) proteomic approaches to investigate meat quality, 4) spatial transcriptomics and metabolomics studies of wooden breast disease, and 5) multiomics analyses on chicken meat quality and spaghetti meat. These topics provide insights into the molecular components that contribute to the structure, function, and dynamics of the underlying mechanisms influencing meat quality traits, including chicken breast myopathies. This information will ultimately contribute to improving the quality and composition of poultry products.
Collapse
Affiliation(s)
- Huaijun Zhou
- Department of Animal Science, University of California, Davis, Davis, CA, USA
| | | | - Mahesh Nair
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, USA
| | - Behnam Abasht
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, USA
| | - Byungwhi Kong
- USDA, Agricultural Research Service, U.S. National Poultry Research Center, Quality & Safety Assessment Research Unit, Athens, GA, USA.
| | - Brian Bowker
- USDA, Agricultural Research Service, U.S. National Poultry Research Center, Quality & Safety Assessment Research Unit, Athens, GA, USA
| |
Collapse
|
23
|
Brotman SM, El-Sayed Moustafa JS, Guan L, Broadaway KA, Wang D, Jackson AU, Welch R, Currin KW, Tomlinson M, Vadlamudi S, Stringham HM, Roberts AL, Lakka TA, Oravilahti A, Fernandes Silva L, Narisu N, Erdos MR, Yan T, Bonnycastle LL, Raulerson CK, Raza Y, Yan X, Parker SCJ, Kuusisto J, Pajukanta P, Tuomilehto J, Collins FS, Boehnke M, Love MI, Koistinen HA, Laakso M, Mohlke KL, Small KS, Scott LJ. Adipose tissue eQTL meta-analysis highlights the contribution of allelic heterogeneity to gene expression regulation and cardiometabolic traits. Nat Genet 2025; 57:180-192. [PMID: 39747594 DOI: 10.1038/s41588-024-01982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/11/2024] [Indexed: 01/04/2025]
Abstract
Complete characterization of the genetic effects on gene expression is needed to elucidate tissue biology and the etiology of complex traits. In the present study, we analyzed 2,344 subcutaneous adipose tissue samples and identified 34,774 conditionally distinct expression quantitative trait locus (eQTL) signals at 18,476 genes. Over half of eQTL genes exhibited at least two eQTL signals. Compared with primary eQTL signals, nonprimary eQTL signals had lower effect sizes, lower minor allele frequencies and less promoter enrichment; they corresponded to genes with higher heritability and higher tolerance for loss of function. Colocalization of eQTLs with genome-wide association study (GWAS) signals for 28 cardiometabolic traits identified 1,835 genes. Inclusion of nonprimary eQTL signals increased discovery of colocalized GWAS-eQTL signals by 46%. Furthermore, 21 genes with ≥2 colocalized GWAS-eQTL signals showed a mediating gene dosage effect on the GWAS trait. Thus, expanded eQTL identification reveals more mechanisms underlying complex traits and improves understanding of the complexity of gene expression regulation.
Collapse
Affiliation(s)
- Sarah M Brotman
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | - Li Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dongmeng Wang
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Kevin W Currin
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Max Tomlinson
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | | | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Amy L Roberts
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lori L Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Yasrab Raza
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Xinyu Yan
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Stephen C J Parker
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Kuusisto
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics and Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Heikki A Koistinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki and Department of Medicine, Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
Liu X, Guo H, Kang M, Fu W, Li H, Ji H, Zhao J, Fang Y, Du M, Xue Y. Multi-step gene set analysis identified HTR3 family genes involving childhood acute lymphoblastic leukemia susceptibility. Arch Toxicol 2025; 99:299-307. [PMID: 39322821 DOI: 10.1007/s00204-024-03881-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
In our previous conventional genome-wide association study (GWAS), WWOX was a susceptibility gene associated with acute lymphoblastic leukemia (ALL) development. Nowadays, advancements in genetic association analyses promote an in-depth exploration of ALL genomics. We conducted a two-step enrichment analysis at both gene and pathway levels based on ALL GWAS data including 269 cases and 1039 controls of Chinese descent. The following functional prediction and experiments were used to evaluate the genetic biology of candidate variants and genes. The serotonin-activated cation-selective channel complex gene-set was a potential biological pathway involved in ALL occurrence. Of which, individuals carrying the T allele of rs33940208 exhibited a prominent reduced risk of ALL [odds ratio (OR) = 0.71, 95% confidence interval (CI) = 0.53 to 0.96, P = 2.81 × 10-2], whereas those with the A allele of rs6779545 demonstrated an increased risk (OR = 1.23, 95% CI = 1.01 to 1.51, P = 4.11 × 10-2). Notably, the two variants displayed a better prediction capability when combined, that the risk of developing childhood ALL increased by 131% in subjects with 2-4 risk alleles compared to those with 0-1 risk alleles (Ptrend = 2.05 × 10-3). In addition, the T allele of rs33940208 could reduce HTR3A mRNA level, while the A allele of rs6779545 increased HTR3D mRNA expression. In this study, we identified HTR3A rs33940208 and HTR3D rs6779545 as potential susceptibility loci for ALL in Chinese children. Future validation and functional research will elucidate the underlying molecular processes, refining preventive strategies for this disease.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, 211166, China
| | - Honghao Guo
- Department of Hematology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Meiyun Kang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Wenfeng Fu
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Huiqin Li
- Department of Genetic Toxicology and Environmental Genomics, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongsheng Ji
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, 211166, China
| | - Jiou Zhao
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Yongjun Fang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China.
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Key Laboratory of Hematology, Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, China.
| | - Mulong Du
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing, 211166, China.
- Department of Genetic Toxicology and Environmental Genomics, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Yao Xue
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.
- Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China.
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Key Laboratory of Hematology, Nanjing Medical University, No. 72 Guangzhou Road, Nanjing, 210008, China.
| |
Collapse
|
25
|
Song Rong S, Larson A, Wiggs JL. ATXN2 loss of function results in glaucoma-related features supporting a role for Ataxin-2 in primary open-angle glaucoma (POAG) pathogenesis. Vision Res 2025; 226:108508. [PMID: 39488861 DOI: 10.1016/j.visres.2024.108508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide. The most common form, primary open-angle glaucoma (POAG), is a genetically complex trait with high heritability. Genome-wide association studies have identified significant POAG and IOP association of a genomic region on chromosome 12 that includes ATXN2 as well as 7 other genes. Association of protein disrupting ATXN2 variants in the NEIGHBORHOOD case-control cohort and the UK Biobank suggests that ATXN2 is a key gene in this locus. To investigate functional effects, we utilized a zebrafish (Danio rerio) CRISPR/Cas9 edited atxn2-knockdown line to show that loss of atxn2 results in reduced eye size, diminished retinal ganglion cells (RGC), increased intraocular pressure (IOP), and impaired visual function in zebrafish. Complementation assays supported functional effects for 14 POAG-associated human ATXN2 missense variants. These results suggest a loss-of-function mechanism underlying a potential role for ATXN2 in POAG pathogenesis.
Collapse
Affiliation(s)
- Shi Song Rong
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, MA, USA
| | - Anna Larson
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, MA, USA
| | - Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, MA, USA.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Kaushal JB, Raut P, Muniyan S, Siddiqui JA, Alsafwani ZW, Seshacharyulu P, Nair SS, Tewari AK, Batra SK. Racial disparity in prostate cancer: an outlook in genetic and molecular landscape. Cancer Metastasis Rev 2024; 43:1233-1255. [PMID: 38902476 PMCID: PMC11560487 DOI: 10.1007/s10555-024-10193-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/04/2024] [Indexed: 06/22/2024]
Abstract
Prostate cancer (PCa) incidence, morbidity, and mortality rates are significantly impacted by racial disparities. Despite innovative therapeutic approaches and advancements in prevention, men of African American (AA) ancestry are at a higher risk of developing PCa and have a more aggressive and metastatic form of the disease at the time of initial PCa diagnosis than other races. Research on PCa has underlined the biological and molecular basis of racial disparity and emphasized the genetic aspect as the fundamental component of racial inequality. Furthermore, the lower enrollment rate, limited access to national-level cancer facilities, and deferred treatment of AA men and other minorities are hurdles in improving the outcomes of PCa patients. This review provides the most up-to-date information on various biological and molecular contributing factors, such as the single nucleotide polymorphisms (SNPs), mutational spectrum, altered chromosomal loci, differential gene expression, transcriptome analysis, epigenetic factors, tumor microenvironment (TME), and immune modulation of PCa racial disparities. This review also highlights future research avenues to explore the underlying biological factors contributing to PCa disparities, particularly in men of African ancestry.
Collapse
Affiliation(s)
- Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Pratima Raut
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Zahraa W Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA
| | - Sujit S Nair
- Department of Urology and the Tisch Cancer Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ashutosh K Tewari
- Department of Urology and the Tisch Cancer Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Division of Urology, Department of Surgery, University of Nebraska Medical Center, Omaha, NE-68198, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE-68198, USA.
| |
Collapse
|
29
|
Chakraborty S, Sarma J, Roy SS, Mitra S, Bagchi S, Das S, Saha S, Mahapatra S, Bhattacharjee S, Maulik M, Acharya M. Functional investigation suggests CNTNAP5 involvement in glaucomatous neurodegeneration obtained from a GWAS in primary angle closure glaucoma. PLoS Genet 2024; 20:e1011502. [PMID: 39637236 DOI: 10.1371/journal.pgen.1011502] [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: 05/08/2024] [Revised: 12/17/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Primary angle closure glaucoma (PACG) affects more than 20 million people worldwide, with an increased prevalence in south-east Asia. In a prior haplotype-based Genome Wide Association Study (GWAS), we identified a novel CNTNAP5 genic region, significantly associated with PACG. In the current study, we have extended our perception of CNTNAP5 involvement in glaucomatous neurodegeneration in a zebrafish model, through investigating phenotypic consequences pertinent to retinal degeneration upon knockdown of cntnap5 by translation-blocking morpholinos. While cntnap5 knockdown was successfully validated using an antibody, immunofluorescence followed by western blot analyses in cntnap5-morphant (MO) zebrafish revealed increased expression of acetylated tubulin indicative of perturbed cytoarchitecture of retinal layers. Moreover, significant loss of Nissl substance is observed in the neuro-retinal layers of cntnap5-MO zebrafish eye, indicating neurodegeneration. Additionally, in spontaneous movement behavioural analysis, cntnap5-MO zebrafish have a significantly lower average distance traversed in light phase compared to mismatch-controls, whereas no significant difference was observed in the dark phase, corroborating with vision loss in the cntnap5-MO zebrafish. This study provides the first direct functional evidence of a putative role of CNTNAP5 in visual neurodegeneration.
Collapse
Affiliation(s)
- Sudipta Chakraborty
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Jyotishman Sarma
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Shantanu Saha Roy
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Sukanya Mitra
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Sayani Bagchi
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Sankhadip Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Sreemoyee Saha
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Surajit Mahapatra
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Samsiddhi Bhattacharjee
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Mahua Maulik
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
| | - Moulinath Acharya
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, India
- Regional Centre for Biotechnology, Faridabad, India
| |
Collapse
|
30
|
Lou S, Zhu G, Xing C, Hao S, Lin J, Xu J, Li D, Du Y, Mi C, Sun L, Wang L, Wang M, Du M, Pan Y. Transcriptome-wide association identifies KLC1 as a regulator of mitophagy in non-syndromic cleft lip with or without palate. IMETA 2024; 3:e262. [PMID: 39742305 PMCID: PMC11683466 DOI: 10.1002/imt2.262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025]
Abstract
This study investigated pathogenic genes associated with non-syndromic cleft lip with or without cleft palate (NSCL/P) through transcriptome-wide association studies (TWAS). By integrating expression quantitative trait loci (eQTL) data with genome-wide association study (GWAS) data, we identified key susceptibility genes, including KLC1. Notably, the variant rs12884809 G>A was associated with an increased risk of NSCL/P by enhancing the binding of the transcription factor ELK1 to the KLC1 promoter, thereby activating its expression. This alteration in KLC1 expression subsequently impacted mitophagy, leading to significant changes in cellular behavior and zebrafish morphology. Our findings illuminate the genetic mechanisms underlying NSCL/P and provide valuable insights for future prevention strategies and a deeper understanding of this condition.
Collapse
Affiliation(s)
- Shu Lou
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Department of Orthodontics, Affiliated Hospital of StomatologyNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Guirong Zhu
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Changyue Xing
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Shushu Hao
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Junyan Lin
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Jiayi Xu
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Dandan Li
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Department of Orthodontics, Affiliated Hospital of StomatologyNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Yifei Du
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Congbo Mi
- The First Affiliated Hospital of Xinjiang Medical UniversityWulumuqiChina
| | - Lian Sun
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Department of Orthodontics, Affiliated Hospital of StomatologyNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
| | - Lin Wang
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Department of Orthodontics, Affiliated Hospital of StomatologyNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Meilin Wang
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Mulong Du
- Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
- Department of Biostatistics, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Yongchu Pan
- State Key Laboratory of Cultivation Base of Research, Prevention and Treatment for Oral DiseasesNanjing Medical UniversityNanjingChina
- Department of Orthodontics, Affiliated Hospital of StomatologyNanjing Medical UniversityNanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| |
Collapse
|
31
|
Kong L, Cheng H, Zhu K, Song B. LOGOWheat: deep learning-based prediction of regulatory effects for noncoding variants in wheats. Brief Bioinform 2024; 26:bbae705. [PMID: 39789857 PMCID: PMC11717721 DOI: 10.1093/bib/bbae705] [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: 05/29/2024] [Revised: 10/18/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Identifying the regulatory effects of noncoding variants presents a significant challenge. Recently, the accumulation of epigenomic profiling data in wheat has provided an opportunity to model the functional impacts of these variants. In this study, we introduce Language of Genome for Wheat (LOGOWheat), a deep learning-based tool designed to predict the regulatory effects of noncoding variants in wheat. LOGOWheat initially employs a self-attention-based, contextualized pretrained language model to acquire bidirectional representations of the unlabeled wheat reference genome. Epigenomic profiling data are also collected and utilized to fine-tune the model, enabling it to discern the regulatory code inherent in genomic sequences. The test results suggest that LOGOWheat is highly effective in predicting multiple chromatin features, achieving an average area under the receiver operating characteristic (AUROC) of 0.8531 and an average area under the precision-recall curve (AUPRC) of 0.7633. Two case studies illustrate and demonstrate the main functions provided by LOGOWheat: assigning scores and prioritizing causal variants within a given variant set and constructing a saturated mutagenesis map in silico to discover high-impact sites or functional motifs in a given sequence. Finally, we propose the concept of extracting potential functional variations from the wheat population by integrating evolutionary conservation information. LOGOWheat is available at http://logowheat.cn/.
Collapse
Affiliation(s)
- Lingpeng Kong
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, No. 97 Buxin Road, Dapeng New District, Shenzhen 518124, China
| | - Hong Cheng
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, No. 97 Buxin Road, Dapeng New District, Shenzhen 518124, China
| | - Kun Zhu
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, No. 97 Buxin Road, Dapeng New District, Shenzhen 518124, China
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, No. 379 Mingli Road (North Section), Zhengzhou 450046, China
| | - Bo Song
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, No. 97 Buxin Road, Dapeng New District, Shenzhen 518124, China
| |
Collapse
|
32
|
Prida E, Pérez-Lois R, Jácome-Ferrer P, Muñoz-Moreno D, Brea-García B, Villalón M, Pena-Leon V, Vázquez-Cobela R, Aguilera CM, Conde-Aranda J, Costas J, Estany-Gestal A, Quiñones M, Leis R, Seoane LM, Al-Massadi O. The PTK2B gene is associated with obesity, adiposity, and leptin levels in children and adolescents. iScience 2024; 27:111120. [PMID: 39498303 PMCID: PMC11533559 DOI: 10.1016/j.isci.2024.111120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/06/2024] [Accepted: 10/03/2024] [Indexed: 11/07/2024] Open
Abstract
Previous studies determined that Pyk2 is involved in several diseases in which the symptomatology presents alterations in energy balance. However, its role in obesity is poorly understood. To evaluate the metabolic role of the Pyk2 gene (PTK2B) in children and adolescents with obesity we measured its mRNA expression levels in peripheral blood mononuclear cells. For that we performed a cross-sectional study involving 130 Caucasian subjects that was divided into two groups according to BMI. Data showed increased PTK2B mRNA expression in children and adolescents with obesity. Interestingly, a positive correlation has been found between the levels of PTK2B with weight, BMI, BMI Z score, fat mass, waist circumference, waist to height ratio, diastolic blood pressure, and leptin. In addition, it is indicated that high levels of PTK2B gene expression might be a predictor of obesity development. This work provides important insights into the previously undescribed role of Pyk2 in obesity.
Collapse
Affiliation(s)
- Eva Prida
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)/Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
| | - Raquel Pérez-Lois
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Pablo Jácome-Ferrer
- Psychiatric Genetics group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
- Universidade de Santiago de Compostela (USC), Rua san francisco s/n, 15782 Santiago de Compostela, Galicia, Spain
| | - Diego Muñoz-Moreno
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)/Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
| | - Beatriz Brea-García
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)/Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - María Villalón
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Verónica Pena-Leon
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Rocío Vázquez-Cobela
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
- Pediatric Nutrition Research Group. Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS). Santiago de Compostela Spain Unit of Investigation in Human Nutrition, Growth and Development of Galicia (GALINUT), University of Santiago de Compostela (USC), Santiago de Compostela, Galicia, Spain
- Unit of Pediatric Gastroenterology, Hepatology and Nutrition. Pediatric Service. University Clinical Hospital of Santiago (CHUS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Concepción M. Aguilera
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Armilla, Granada, Spain
- Biosanitary Research Institute (IBS), University of Granada, Av de Madrid 15, 18012 Granada, Andalusia, Spain
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology Group, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706 Galicia, Spain
| | - Javier Costas
- Psychiatric Genetics group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Ana Estany-Gestal
- Plataforma de Metodología de la Investigación, Instituto de Investigación de Santiago (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Mar Quiñones
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Rosaura Leis
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
- Pediatric Nutrition Research Group. Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS). Santiago de Compostela Spain Unit of Investigation in Human Nutrition, Growth and Development of Galicia (GALINUT), University of Santiago de Compostela (USC), Santiago de Compostela, Galicia, Spain
- Unit of Pediatric Gastroenterology, Hepatology and Nutrition. Pediatric Service. University Clinical Hospital of Santiago (CHUS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Luisa María Seoane
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
| | - Omar Al-Massadi
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS)/Complexo Hospitalario Universitario de Santiago (SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Galicia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av Monforte de Lemos3-5, 28029 Madrid, Spain
| |
Collapse
|
33
|
Thakur R, Xu M, Sowards H, Yon J, Jessop L, Myers T, Zhang T, Chari R, Long E, Rehling T, Hennessey R, Funderburk K, Yin J, Machiela MJ, Johnson ME, Wells AD, Chesi A, Grant SF, Iles MM, Landi MT, Law MH, Choi J, Brown KM. Mapping chromatin interactions at melanoma susceptibility loci and cell-type specific dataset integration uncovers distant gene targets of cis-regulation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.14.24317204. [PMID: 39802764 PMCID: PMC11722502 DOI: 10.1101/2024.11.14.24317204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Genome-wide association studies (GWAS) of melanoma risk have identified 68 independent signals at 54 loci. For most loci, specific functional variants and their respective target genes remain to be established. Capture-HiC is an assay that links fine-mapped risk variants to candidate target genes by comprehensively mapping cell-type specific chromatin interactions. We performed a melanoma GWAS region-focused capture-HiC assay in human primary melanocytes to identify physical interactions between fine-mapped risk variants and potential causal melanoma susceptibility genes. Overall, chromatin interaction data alone nominated potential causal genes for 61 of the 68 melanoma risk signals, identifying many candidates beyond those reported by previous studies. We further integrated these data with cell-type specific epigenomic (chromatin state, accessibility), gene expression (eQTL/TWAS), DNA methylation (meQTL/MWAS), and massively parallel reporter assay (MPRA) data to prioritize potentially cis-regulatory variants and their respective candidate gene targets. From the set of fine-mapped variants across these loci, we identified 140 prioritized candidate causal variants linked to 195 candidate genes at 42 risk signals. In addition, we developed an integrative scoring system to facilitate candidate gene prioritization, integrating melanocyte and melanoma datasets. Notably, at several GWAS risk signals we observed long-range chromatin connections (500 kb to >1 Mb) with distant candidate target genes. We validated several such cis-regulatory interactions using CRISPR inhibition, providing evidence for known cancer driver genes MDM4 and CBL, as well as the SRY-box transcription factor SOX4, as likely melanoma risk genes.
Collapse
Affiliation(s)
- Rohit Thakur
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Hayley Sowards
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Joshuah Yon
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Lea Jessop
- Laboratory of Genomic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Timothy Myers
- Laboratory of Genomic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Tongwu Zhang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Lab for Cancer Research, Frederick, MD, USA
| | - Erping Long
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Thomas Rehling
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Rebecca Hennessey
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Karen Funderburk
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jinhu Yin
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mitchell J. Machiela
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Matthew E. Johnson
- Division of Human Genetics, Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mark M. Iles
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Maria Teresa Landi
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Matthew H. Law
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, University fo Queensland, Brisbane, QLD, Australia
| | | | - Jiyeon Choi
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin M. Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
34
|
Zainab A, Anzawa H, Kinoshita K. Identifying key genes in COPD risk via multiple population data integration and gene prioritization. PLoS One 2024; 19:e0305803. [PMID: 39509417 PMCID: PMC11542775 DOI: 10.1371/journal.pone.0305803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent disease, making it a leading cause of death worldwide. Several genome-wide association studies (GWAS) have been conducted to identify loci associated with COPD. However, different ancestral genetic compositions for the same disease across various populations present challenges in studies involving multi-population data. In this study, we aimed to identify protein-coding genes associated with COPD by prioritizing genes for each population's GWAS data, and then combining these results instead of performing a common meta-GWAS due to significant sample differences in different population cohorts. Lung function measurements are often used as indicators for COPD risk prediction; therefore, we used lung function GWAS data from two populations, Japanese and European, and re-evaluated them using a multi-population gene prioritization approach. This study identified significant single nucleotide variants (SNPs) in both Japanese and European populations. The Japanese GWAS revealed nine significant SNPs and four lead SNPs in three genomic risk loci. In comparison, the European population showed five lead SNPs and 17 independent significant SNPs in 21 genomic risk loci. A comparative analysis of the results found 28 similar genes in the prioritized gene lists of both populations. We also performed a standard meta-analysis for comparison and identified 18 common genes in both populations. Our approach demonstrated that trans-ethnic linkage disequilibrium (LD) could detect some significant novel associations and genes that have yet to be reported or were missed in previous analyses. The study suggests that a gene prioritization approach for multi-population analysis using GWAS data may be a feasible method to identify new associations in data with genetic diversity across different populations. It also highlights the possibility of identifying generalized and population-specific treatment and diagnostic options.
Collapse
Affiliation(s)
- Afeefa Zainab
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Hayato Anzawa
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kengo Kinoshita
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| |
Collapse
|
35
|
Nguyen PT, Coetzee SG, Silacheva I, Hazelett DJ. Genome wide association studies are enriched for interacting genes. RESEARCH SQUARE 2024:rs.3.rs-5189487. [PMID: 39502771 PMCID: PMC11537335 DOI: 10.21203/rs.3.rs-5189487/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
Background With recent advances in single cell technology, high-throughput methods provide unique insight into disease mechanisms and more importantly, cell type origin. Here, we used multi-omics data to understand how genetic variants from genome-wide association studies influence development of disease. We show in principle how to use genetic algorithms with normal, matching pairs of single-nucleus RNA- and ATAC-seq, genome annotations, and protein-protein interaction data to describe the genes and cell types collectively and their contribution to increased risk. Results We used genetic algorithms to measure fitness of gene-cell set proposals against a series of objective functions that capture data and annotations. The highest information objective function captured protein-protein interactions. We observed significantly greater fitness scores and subgraph sizes in foreground vs.matching sets of control variants. Furthermore, our model reliably identified known targets and ligand-receptor pairs, consistent with prior studies. Conclusions Our findings suggested that application of genetic algorithms to association studies can generate a coherent cellular model of risk from a set of susceptibility variants. Further, we showed, using breast cancer as an example, that such variants have a greater number of physical interactions than expected due to chance.
Collapse
|
36
|
Ghura S, Beratan NR, Shi X, Alvarez-Periel E, Bond Newton SE, Akay-Espinoza C, Jordan-Sciutto KL. Genetic knock-in of EIF2AK3 variants reveals differences in PERK activity in mouse liver and pancreas under endoplasmic reticulum stress. Sci Rep 2024; 14:23812. [PMID: 39394239 PMCID: PMC11470120 DOI: 10.1038/s41598-024-74362-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: 04/08/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024] Open
Abstract
Common single-nucleotide variants (SNVs) of eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3) slightly increase the risk of disorders in the periphery and the central nervous system. EIF2AK3 encodes protein kinase RNA-like endoplasmic reticulum kinase (PERK), a key regulator of ER stress. Three exonic EIF2AK3 SNVs form the PERK-B haplotype, which is present in 28% of the global population. Importantly, the precise impact of these SNVs on PERK activity remains elusive. In this study, we demonstrate that PERK-B SNVs do not alter PERK expression or basal activity in vitro and in the novel triple knock-in mice expressing the exonic PERK-B SNVs in vivo. However, the kinase activity of PERK-B protein is higher than that of PERK-A in a cell-free assay and in mouse liver homogenates. Pancreatic tissue in PERK-B/B mice also exhibit increased susceptibility to apoptosis under acute ER stress. Monocyte-derived macrophages from PERK-B/B mice exhibit higher PERK activity than those from PERK-A/A mice, albeit with minimal functional consequences at acute timepoints. The subtle PERK-B-driven effects observed in liver and pancreas during acute stress implicate PERK as a contributor to disease susceptibility. The novel PERK-B mouse model provides valuable insights into ER stress-induced PERK activity, aiding the understanding of the genetic basis of disorders associated with ER stress.
Collapse
Affiliation(s)
- Shivesh Ghura
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy, Philadelphia, PA, 19104, USA
| | - Noah R Beratan
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy, Philadelphia, PA, 19104, USA
| | - Xinglong Shi
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy, Philadelphia, PA, 19104, USA
| | - Elena Alvarez-Periel
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy, Philadelphia, PA, 19104, USA
| | - Sarah E Bond Newton
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy, Philadelphia, PA, 19104, USA
- Department of Neuroscience, Weinberg ALS Center, Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Cagla Akay-Espinoza
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy, Philadelphia, PA, 19104, USA
| | - Kelly L Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy, Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
Reus LM, Jansen IE, Tijms BM, Visser PJ, Tesi N, van der Lee SJ, Vermunt L, Peeters CFW, De Groot LA, Hok-A-Hin YS, Chen-Plotkin A, Irwin DJ, Hu WT, Meeter LH, van Swieten JC, Holstege H, Hulsman M, Lemstra AW, Pijnenburg YAL, van der Flier WM, Teunissen CE, del Campo Milan M. Connecting dementia risk loci to the CSF proteome identifies pathophysiological leads for dementia. Brain 2024; 147:3522-3533. [PMID: 38527854 PMCID: PMC11449142 DOI: 10.1093/brain/awae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
Genome-wide association studies have successfully identified many genetic risk loci for dementia, but exact biological mechanisms through which genetic risk factors contribute to dementia remains unclear. Integrating CSF proteomic data with dementia risk loci could reveal intermediate molecular pathways connecting genetic variance to the development of dementia. We tested to what extent effects of known dementia risk loci can be observed in CSF levels of 665 proteins [proximity extension-based (PEA) immunoassays] in a deeply-phenotyped mixed memory clinic cohort [n = 502, mean age (standard deviation, SD) = 64.1 (8.7) years, 181 female (35.4%)], including patients with Alzheimer's disease (AD, n = 213), dementia with Lewy bodies (DLB, n = 50) and frontotemporal dementia (FTD, n = 93), and controls (n = 146). Validation was assessed in independent cohorts (n = 99 PEA platform, n = 198, mass reaction monitoring-targeted mass spectroscopy and multiplex assay). We performed additional analyses stratified according to diagnostic status (AD, DLB, FTD and controls separately), to explore whether associations between CSF proteins and genetic variants were specific to disease or not. We identified four AD risk loci as protein quantitative trait loci (pQTL): CR1-CR2 (rs3818361, P = 1.65 × 10-8), ZCWPW1-PILRB (rs1476679, P = 2.73 × 10-32), CTSH-CTSH (rs3784539, P = 2.88 × 10-24) and HESX1-RETN (rs186108507, P = 8.39 × 10-8), of which the first three pQTLs showed direct replication in the independent cohorts. We identified one AD-specific association between a rare genetic variant of TREM2 and CSF IL6 levels (rs75932628, P = 3.90 × 10-7). DLB risk locus GBA showed positive trans effects on seven inter-related CSF levels in DLB patients only. No pQTLs were identified for FTD loci, either for the total sample as for analyses performed within FTD only. Protein QTL variants were involved in the immune system, highlighting the importance of this system in the pathophysiology of dementia. We further identified pQTLs in stratified analyses for AD and DLB, hinting at disease-specific pQTLs in dementia. Dissecting the contribution of risk loci to neurobiological processes aids in understanding disease mechanisms underlying dementia.
Collapse
Affiliation(s)
- Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, CA 90095 CA, USA
| | - Iris E Jansen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Niccoló Tesi
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Lisa Vermunt
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Carel F W Peeters
- Mathematical and Statistical Methods group (Biometris), Wageningen University and Research, Wageningen, 6708 PB Wageningen, The Netherlands
| | - Lisa A De Groot
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Rutgers-RWJ Medical School, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08901, USA
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Henne Holstege
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Marc Hulsman
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Marta del Campo Milan
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, 28003 Madrid, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, 08005 Barcelona, Spain
| |
Collapse
|
38
|
Mayerhofer L, Nes RB, Yu B, Ayorech Z, Lan X, Ystrom E, Røysamb E. Stability and change in maternal wellbeing and illbeing from pregnancy to three years postpartum. Qual Life Res 2024; 33:2797-2808. [PMID: 38992240 PMCID: PMC11452533 DOI: 10.1007/s11136-024-03730-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] [Accepted: 06/27/2024] [Indexed: 07/13/2024]
Abstract
PURPOSE Motherhood affects women's mental health, encompassing aspects of both wellbeing and illbeing. This study investigated stability and change in wellbeing (i.e., relationship satisfaction and positive affect) and illbeing (i.e., depressive and anxiety symptoms) from pregnancy to three years postpartum. We further investigated the mutual and dynamic relations between these constructs over time and the role of genetic propensities in their time-invariant stability. DATA AND METHODS This four-wave longitudinal study included 83,124 women from the Norwegian Mother, Father, and Child Cohort Study (MoBa) linked to the Medical Birth Registry of Norway. Data were collected during pregnancy (30 weeks) and at 6, 18 and 36 months postpartum. Wellbeing and illbeing were based on the Relationship Satisfaction Scale, the Differential Emotions Scale and Hopkins Symptoms Checklist-8. Genetics were measured by the wellbeing spectrum polygenic index. Analyses were based on random intercept cross-lagged panel models using R. RESULTS All four outcomes showed high stability and were mutually interconnected over time, with abundant cross-lagged predictions. The period of greatest instability was from pregnancy to 6 months postpartum, followed by increasing stability. Prenatal relationship satisfaction played a crucial role in maternal mental health postpartum. Women's genetic propensity to wellbeing contributed to time-invariant stability of all four constructs. CONCLUSION Understanding the mutual relationship between different aspects of wellbeing and illbeing allows for identifying potential targets for health promotion interventions. Time-invariant stability was partially explained by genetics. Maternal wellbeing and illbeing develop in an interdependent way from pregnancy to 36 months postpartum.
Collapse
Affiliation(s)
| | - Ragnhild Bang Nes
- PROMENTA Research Center, Oslo University, Oslo, Norway
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Philosophy, Classics, and History of Arts and Ideas, University of Oslo, Oslo, Norway
| | - Baeksan Yu
- Gwangju National University of Education, Gwangju, South Korea
| | - Ziada Ayorech
- PROMENTA Research Center, Oslo University, Oslo, Norway
| | - Xiaoyu Lan
- PROMENTA Research Center, Oslo University, Oslo, Norway
| | - Eivind Ystrom
- PROMENTA Research Center, Oslo University, Oslo, Norway
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Espen Røysamb
- PROMENTA Research Center, Oslo University, Oslo, Norway
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
39
|
Barry T, Roeder K, Katsevich E. Exponential family measurement error models for single-cell CRISPR screens. Biostatistics 2024; 25:1254-1272. [PMID: 38649751 PMCID: PMC11471999 DOI: 10.1093/biostatistics/kxae010] [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: 03/24/2022] [Revised: 01/10/2024] [Accepted: 03/11/2024] [Indexed: 04/25/2024] Open
Abstract
CRISPR genome engineering and single-cell RNA sequencing have accelerated biological discovery. Single-cell CRISPR screens unite these two technologies, linking genetic perturbations in individual cells to changes in gene expression and illuminating regulatory networks underlying diseases. Despite their promise, single-cell CRISPR screens present considerable statistical challenges. We demonstrate through theoretical and real data analyses that a standard method for estimation and inference in single-cell CRISPR screens-"thresholded regression"-exhibits attenuation bias and a bias-variance tradeoff as a function of an intrinsic, challenging-to-select tuning parameter. To overcome these difficulties, we introduce GLM-EIV ("GLM-based errors-in-variables"), a new method for single-cell CRISPR screen analysis. GLM-EIV extends the classical errors-in-variables model to responses and noisy predictors that are exponential family-distributed and potentially impacted by the same set of confounding variables. We develop a computational infrastructure to deploy GLM-EIV across hundreds of processors on clouds (e.g. Microsoft Azure) and high-performance clusters. Leveraging this infrastructure, we apply GLM-EIV to analyze two recent, large-scale, single-cell CRISPR screen datasets, yielding several new insights.
Collapse
Affiliation(s)
- Timothy Barry
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Building 2 435, 655 Huntington Ave, Boston, MA 02115, United States
| | - Kathryn Roeder
- Department of Statistics and Data Science, Carnegie Mellon University, Baker Hall 228B, 4909 Frew St, Pittsburgh, PA 15213, United States
| | - Eugene Katsevich
- Department of Statistics and Data Science, University of Pennsylvania, Academic Research Building 311, 265 South 37th Street Philadelphia, PA 19104, United States
| |
Collapse
|
40
|
Amini M, Bagheri A, P Paulus M, Delen D. Multimorbidity in neurodegenerative diseases: a network analysis. Inform Health Soc Care 2024; 49:212-226. [PMID: 39363570 DOI: 10.1080/17538157.2024.2405869] [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: 10/05/2024]
Abstract
The socioeconomic costs of neurodegenerative diseases (NDs) are highly affected by comorbidities. This study aims to enhance our understanding of the prevalent complications of NDs through the lens of network analysis. A multimorbidity network (MN) was constructed based on a longitudinal EHR dataset of 93,647,498 diagnoses of 824,847 patients. The association between the conditions was measured by two metrics, i.e. Phi-correlation and Cosine Index (CI). Based on multiple network centrality measures, a fused ranking list of the prevalent multimorbidities was provided. Finally, class-level networks depicting the prevalence and strength of diseases in different classes were constructed. The general MN included 928 diseases and 337,253 associations. Considering a 99% confidence level, two networks of 575 relationships were constructed based on Phi-correlations (73 diseases) and CI (102 diseases). Five out of 19 ICD-9 categories did not appear in either of the networks. Also, ND's immediate MNs for the top 50% of the significant associations included 42 relationships, whereas the Phi-correlation and CI networks included 36 and 34 diseases, respectively. Thirteen diseases were identified as the most notable multimorbidities based on various centrality measures. The analysis framework helps practitioners toward better resource allocations, more effective preventive screenings, and improved quality of life for ND patients and caregivers.
Collapse
Affiliation(s)
- Mostafa Amini
- Department of Information Systems, College of Business, California State University, Long Beach, California, USA
| | - Ali Bagheri
- Department of Management Science and Information Systems, Spears School of Business, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA
- Oxley College of Health and Natural Sciences, University of Tulsa, Tulsa, Oklahoma, USA
| | - Dursun Delen
- Center for Health Systems Innovation, Department of Management Science and Information Systems, Spears School of Business, Oklahoma State University, Tulsa, Oklahoma, USA
- Department of Industrial Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer/Istanbul, Türkiye
| |
Collapse
|
41
|
Xavier JM, Magno R, Russell R, de Almeida BP, Jacinta-Fernandes A, Besouro-Duarte A, Dunning M, Samarajiwa S, O'Reilly M, Maia AM, Rocha CL, Rosli N, Ponder BAJ, Maia AT. Identification of candidate causal variants and target genes at 41 breast cancer risk loci through differential allelic expression analysis. Sci Rep 2024; 14:22526. [PMID: 39341862 PMCID: PMC11438911 DOI: 10.1038/s41598-024-72163-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: 02/01/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
Understanding breast cancer genetic risk relies on identifying causal variants and candidate target genes in risk loci identified by genome-wide association studies (GWAS), which remains challenging. Since most loci fall in active gene regulatory regions, we developed a novel approach facilitated by pinpointing the variants with greater regulatory potential in the disease's tissue of origin. Through genome-wide differential allelic expression (DAE) analysis, using microarray data from 64 normal breast tissue samples, we mapped the variants associated with DAE (daeQTLs). Then, we intersected these with GWAS data to reveal candidate risk regulatory variants and analysed their cis-acting regulatory potential. Finally, we validated our approach by extensive functional analysis of the 5q14.1 breast cancer risk locus. We observed widespread gene expression regulation by cis-acting variants in breast tissue, with 65% of coding and noncoding expressed genes displaying DAE (daeGenes). We identified over 54 K daeQTLs for 6761 (26%) daeGenes, including 385 daeGenes harbouring variants previously associated with BC risk. We found 1431 daeQTLs mapped to 93 different loci in strong linkage disequilibrium with risk-associated variants (risk-daeQTLs), suggesting a link between risk-causing variants and cis-regulation. There were 122 risk-daeQTL with stronger cis-acting potential in active regulatory regions with protein binding evidence. These variants mapped to 41 risk loci, of which 29 had no previous report of target genes and were candidates for regulating the expression levels of 65 genes. As validation, we identified and functionally characterised five candidate causal variants at the 5q14.1 risk locus targeting the ATG10 and ATP6AP1L genes, likely acting via modulation of alternative transcription and transcription factor binding. Our study demonstrates the power of DAE analysis and daeQTL mapping to identify causal regulatory variants and target genes at breast cancer risk loci, including those with complex regulatory landscapes. It additionally provides a genome-wide resource of variants associated with DAE for future functional studies.
Collapse
Affiliation(s)
- Joana M Xavier
- Cintesis@Rise, Universidade do Algarve, Faro, Portugal.
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Faro, Portugal.
| | - Ramiro Magno
- Cintesis@Rise, Universidade do Algarve, Faro, Portugal
- Pattern Institute PT, Faro, Portugal
| | - Roslin Russell
- Cambridge Institute - CRUK, University of Cambridge, Cambridge, UK
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Bernardo P de Almeida
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Faro, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
- InstaDeep, Paris, France
| | - Ana Jacinta-Fernandes
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Faro, Portugal
| | | | - Mark Dunning
- Cambridge Institute - CRUK, University of Cambridge, Cambridge, UK
- Sheffield Bioinformatics Core, The School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Shamith Samarajiwa
- Medical Research Council (MRC) Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
- Genetics and Genomics Section, Imperial College London, London, UK
| | - Martin O'Reilly
- Cambridge Institute - CRUK, University of Cambridge, Cambridge, UK
| | | | - Cátia L Rocha
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Faro, Portugal
- Faculty of Medicine, Instituto de Saúde Ambiental (ISAMB), University of Lisbon, Lisbon, Portugal
| | - Nordiana Rosli
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Faro, Portugal
- Training Division, Ministry of Health Malaysia, Putrajaya, Malaysia
- Biometrology Group, Division of Chemical and Biological Metrology, Korea Research Institute of Standards and Science, Daejeon, South Korea
| | - Bruce A J Ponder
- Cambridge Institute - CRUK, University of Cambridge, Cambridge, UK
| | - Ana-Teresa Maia
- Cintesis@Rise, Universidade do Algarve, Faro, Portugal.
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Faro, Portugal.
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Faro, Portugal.
| |
Collapse
|
42
|
Aman A, Slob EAW, Ward J, Sattar N, Strawbridge RJ. Investigating the association of the effect of genetically proxied PCSK9i with mood disorders using cis-pQTLs: A drug-target Mendelian randomization study. PLoS One 2024; 19:e0310396. [PMID: 39325747 PMCID: PMC11426468 DOI: 10.1371/journal.pone.0310396] [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: 10/12/2023] [Accepted: 09/01/2024] [Indexed: 09/28/2024] Open
Abstract
PCSK9-inhibitors (PCSK9i) are new drugs recently approved to lower LDL-cholesterol levels. However, due to the lack of long-term clinical data, the potential adverse effects of long-term use are still unknown. The PCSK9 genetic locus has been recently implicated in mood disorders and hence we wanted to assess if the effect of PCSK9i that block the PCSK9 protein can lead to an increase in the incidence of mood disorders. We used genetically-reduced PCSK9 protein levels (pQTLs) in plasma, serum, cerebrospinal fluid as a proxy for the effect of PCSK9i. We performed Mendelian randomization analyses using PCSK9 levels as exposure and mood disorder traits major depressive disorder, mood instability, and neuroticism score as outcomes. We find no association of PCSK9 levels with mood disorder traits in serum, plasma, and cerebrospinal fluid. We can conclude that genetically proxied on-target effect of pharmacological PCSK9 inhibition is unlikely to contribute to mood disorders.
Collapse
Affiliation(s)
- Alisha Aman
- The Graduate School, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- School of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Eric A W Slob
- Department of Psychology, Education, and Child Studies, Erasmus University Rotterdam, Rotterdam, The Netherlands
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, Rotterdam, The Netherlands
- Erasmus University Rotterdam Institute for Behaviour and Biology, Erasmus School of Economics, Rotterdam, The Netherlands
| | - Joey Ward
- School of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Rona J Strawbridge
- School of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
43
|
Long E, Yin J, Shin JH, Li Y, Li B, Kane A, Patel H, Sun X, Wang C, Luong T, Xia J, Han Y, Byun J, Zhang T, Zhao W, Landi MT, Rothman N, Lan Q, Chang YS, Yu F, Amos CI, Shi J, Lee JG, Kim EY, Choi J. Context-aware single-cell multiomics approach identifies cell-type-specific lung cancer susceptibility genes. Nat Commun 2024; 15:7995. [PMID: 39266564 PMCID: PMC11392933 DOI: 10.1038/s41467-024-52356-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
Genome-wide association studies (GWAS) identified over fifty loci associated with lung cancer risk. However, underlying mechanisms and target genes are largely unknown, as most risk-associated variants might regulate gene expression in a context-specific manner. Here, we generate a barcode-shared transcriptome and chromatin accessibility map of 117,911 human lung cells from age/sex-matched ever- and never-smokers to profile context-specific gene regulation. Identified candidate cis-regulatory elements (cCREs) are largely cell type-specific, with 37% detected in one cell type. Colocalization of lung cancer candidate causal variants (CCVs) with these cCREs combined with transcription factor footprinting prioritize the variants for 68% of the GWAS loci. CCV-colocalization and trait relevance score indicate that epithelial and immune cell categories, including rare cell types, contribute to lung cancer susceptibility the most. A multi-level cCRE-gene linking system identifies candidate susceptibility genes from 57% of the loci, where most loci display cell-category-specific target genes, suggesting context-specific susceptibility gene function.
Collapse
Affiliation(s)
- Erping Long
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinhu Yin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ju Hye Shin
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yuyan Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bolun Li
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Alexander Kane
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Harsh Patel
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Xinti Sun
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Thong Luong
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jun Xia
- Department of Biomedical Sciences, Creighton University, Omaha, NE, USA
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wei Zhao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Yoon Soo Chang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Fulong Yu
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jin Gu Lee
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Eun Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
44
|
Wenz BM, He Y, Chen NC, Pickrell JK, Li JH, Dudek MF, Li T, Keener R, Voight BF, Brown CD, Battle A. Genotype inference from aggregated chromatin accessibility data reveals genetic regulatory mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.610850. [PMID: 39282458 PMCID: PMC11398312 DOI: 10.1101/2024.09.04.610850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Background Understanding the genetic causes for variability in chromatin accessibility can shed light on the molecular mechanisms through which genetic variants may affect complex traits. Thousands of ATAC-seq samples have been collected that hold information about chromatin accessibility across diverse cell types and contexts, but most of these are not paired with genetic information and come from diverse distinct projects and laboratories. Results We report here joint genotyping, chromatin accessibility peak calling, and discovery of quantitative trait loci which influence chromatin accessibility (caQTLs), demonstrating the capability of performing caQTL analysis on a large scale in a diverse sample set without pre-existing genotype information. Using 10,293 profiling samples representing 1,454 unique donor individuals across 653 studies from public databases, we catalog 23,381 caQTLs in total. After joint discovery analysis, we cluster samples based on accessible chromatin profiles to identify context-specific caQTLs. We find that caQTLs are strongly enriched for annotations of gene regulatory elements across diverse cell types and tissues and are often strongly linked with genetic variation associated with changes in expression (eQTLs), indicating that caQTLs can mediate genetic effects on gene expression. We demonstrate sharing of causal variants for chromatin accessibility and diverse complex human traits, enabling a more complete picture of the genetic mechanisms underlying complex human phenotypes. Conclusions Our work provides a proof of principle for caQTL calling from previously ungenotyped samples, and represents one of the largest, most diverse caQTL resources currently available, informing mechanisms of genetic regulation of gene expression and contribution to disease.
Collapse
Affiliation(s)
- Brandon M. Wenz
- Genetics and Epigenetics Program, Cell and Molecular Biology Graduate Group, Biomedical Graduate Studies, University of Pennsylvania - Perelman School of Medicine, Philadelphia PA 19104
| | - Yuan He
- Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD, 21218
| | - Nae-Chyun Chen
- Department of Computer Science, Johns Hopkins University; Baltimore, MD, 21218
| | | | | | - Max F. Dudek
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - Taibo Li
- Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD, 21218
| | - Rebecca Keener
- Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD, 21218
| | - Benjamin F. Voight
- Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia PA, 19104
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania – Perelman School of Medicine, Philadelphia, PA, 19104
| | - Christopher D. Brown
- Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD, 21218
- Department of Computer Science, Johns Hopkins University; Baltimore, MD, 21218
- Department of Genetic Medicine, Johns Hopkins University; Baltimore, MD, 21218
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, 21218
- Data Science and AI Institute, Johns Hopkins University, Baltimore, MD, 21218
| |
Collapse
|
45
|
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
|
46
|
Broadaway KA, Brotman SM, Rosen JD, Currin KW, Alkhawaja AA, Etheridge AS, Wright F, Gallins P, Jima D, Zhou YH, Love MI, Innocenti F, Mohlke KL. Liver eQTL meta-analysis illuminates potential molecular mechanisms of cardiometabolic traits. Am J Hum Genet 2024; 111:1899-1913. [PMID: 39173627 PMCID: PMC11393674 DOI: 10.1016/j.ajhg.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Understanding the molecular mechanisms of complex traits is essential for developing targeted interventions. We analyzed liver expression quantitative-trait locus (eQTL) meta-analysis data on 1,183 participants to identify conditionally distinct signals. We found 9,013 eQTL signals for 6,564 genes; 23% of eGenes had two signals, and 6% had three or more signals. We then integrated the eQTL results with data from 29 cardiometabolic genome-wide association study (GWAS) traits and identified 1,582 GWAS-eQTL colocalizations for 747 eGenes. Non-primary eQTL signals accounted for 17% of all colocalizations. Isolating signals by conditional analysis prior to coloc resulted in 37% more colocalizations than using marginal eQTL and GWAS data, highlighting the importance of signal isolation. Isolating signals also led to stronger evidence of colocalization: among 343 eQTL-GWAS signal pairs in multi-signal regions, analyses that isolated the signals of interest resulted in higher posterior probability of colocalization for 41% of tests. Leveraging allelic heterogeneity, we predicted causal effects of gene expression on liver traits for four genes. To predict functional variants and regulatory elements, we colocalized eQTL with liver chromatin accessibility QTL (caQTL) and found 391 colocalizations, including 73 with non-primary eQTL signals and 60 eQTL signals that colocalized with both a caQTL and a GWAS signal. Finally, we used publicly available massively parallel reporter assays in HepG2 to highlight 14 eQTL signals that include at least one expression-modulating variant. This multi-faceted approach to unraveling the genetic underpinnings of liver-related traits could lead to therapeutic development.
Collapse
Affiliation(s)
- K Alaine Broadaway
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah M Brotman
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jonathan D Rosen
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kevin W Currin
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Abdalla A Alkhawaja
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Amy S Etheridge
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Fred Wright
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA; Department of Statistics, North Carolina State University, Raleigh, NC 27695, USA
| | - Paul Gallins
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | - Dereje Jima
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | - Yi-Hui Zhou
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Federico Innocenti
- Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
47
|
Unger Avila P, Padvitski T, Leote AC, Chen H, Saez-Rodriguez J, Kann M, Beyer A. Gene regulatory networks in disease and ageing. Nat Rev Nephrol 2024; 20:616-633. [PMID: 38867109 DOI: 10.1038/s41581-024-00849-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
The precise control of gene expression is required for the maintenance of cellular homeostasis and proper cellular function, and the declining control of gene expression with age is considered a major contributor to age-associated changes in cellular physiology and disease. The coordination of gene expression can be represented through models of the molecular interactions that govern gene expression levels, so-called gene regulatory networks. Gene regulatory networks can represent interactions that occur through signal transduction, those that involve regulatory transcription factors, or statistical models of gene-gene relationships based on the premise that certain sets of genes tend to be coexpressed across a range of conditions and cell types. Advances in experimental and computational technologies have enabled the inference of these networks on an unprecedented scale and at unprecedented precision. Here, we delineate different types of gene regulatory networks and their cell-biological interpretation. We describe methods for inferring such networks from large-scale, multi-omics datasets and present applications that have aided our understanding of cellular ageing and disease mechanisms.
Collapse
Affiliation(s)
- Paula Unger Avila
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Tsimafei Padvitski
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ana Carolina Leote
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - He Chen
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Julio Saez-Rodriguez
- Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Martin Kann
- Department II of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andreas Beyer
- Cluster of Excellence on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
48
|
Malakhov MM, Dai B, Shen XT, Pan W. A bootstrap model comparison test for identifying genes with context-specific patterns of genetic regulation. Ann Appl Stat 2024; 18:1840-1857. [PMID: 39421855 PMCID: PMC11484521 DOI: 10.1214/23-aoas1859] [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] [Indexed: 10/19/2024]
Abstract
Understanding how genetic variation affects gene expression is essential for a complete picture of the functional pathways that give rise to complex traits. Although numerous studies have established that many genes are differentially expressed in distinct human tissues and cell types, no tools exist for identifying the genes whose expression is differentially regulated. Here we introduce DRAB (differential regulation analysis by bootstrapping), a gene-based method for testing whether patterns of genetic regulation are significantly different between tissues or other biological contexts. DRAB first leverages the elastic net to learn context-specific models of local genetic regulation and then applies a novel bootstrap-based model comparison test to check their equivalency. Unlike previous model comparison tests, our proposed approach can determine whether population-level models have equal predictive performance by accounting for the variability of feature selection and model training. We validated DRAB on mRNA expression data from a variety of human tissues in the Genotype-Tissue Expression (GTEx) Project. DRAB yielded biologically reasonable results and had sufficient power to detect genes with tissue-specific regulatory profiles while effectively controlling false positives. By providing a framework that facilitates the prioritization of differentially regulated genes, our study enables future discoveries on the genetic architecture of molecular phenotypes.
Collapse
Affiliation(s)
| | - Ben Dai
- Department of Statistics, The Chinese University of Hong Kong
| | | | - Wei Pan
- Division of Biostatistics and Health Data Science, University of Minnesota
| |
Collapse
|
49
|
Verardo LL, Carolino N, Duarte MR, Rodrigues Almeida EA, Dallago G, Braga Magalhães AF. Editorial: Omics applied to livestock genetics: volume II. Front Genet 2024; 15:1477826. [PMID: 39246573 PMCID: PMC11377329 DOI: 10.3389/fgene.2024.1477826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024] Open
Affiliation(s)
- Lucas Lima Verardo
- Laboratory of Animal Breeding, Department of Animal Science, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Nuno Carolino
- Instituto Nacional Investigação Agraria e Veterinaria (INIAV), Oeiras, Portugal
| | - Marcela Ramos Duarte
- Laboratory of Animal Breeding, Department of Animal Science, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Emily Alves Rodrigues Almeida
- Laboratory of Animal Breeding, Department of Animal Science, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Gabriel Dallago
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Ana Fabrícia Braga Magalhães
- Laboratory of Animal Breeding, Department of Animal Science, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| |
Collapse
|
50
|
Qin L, Qiu M, Tang J, Liu S, Lin Q, Huang Q, Wei X, Wen Q, Chen P, Zhou Z, Cao J, Liang X, Guo Q, Nong C, Gong Y, Wei Y, Jiang Y, Yu H, Liu Y. Genetic Variants in p53 Pathway Genes Affect Survival of Patients with HBV-Related Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:1541-1555. [PMID: 39156673 PMCID: PMC11328861 DOI: 10.2147/jhc.s459792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/26/2024] [Indexed: 08/20/2024] Open
Abstract
Purpose P53 is a suppressor gene closely related to carcinogenesis. However, the associations between genetic variants in the p53 signaling pathway and prognosis in hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) remain unknown. The current study aims to analyze associations between the single nucleotide polymorphisms (SNPs) in p53 pathway-related genes and survival of patients with HBV-HCC. Methods We evaluated the associations between 4698 SNPs in 70 genes of the p53 pathway and overall survival (OS) of 866 patients in additive genetic models by using Cox proportional hazards regression analysis. Stepwise multivariable Cox regression analysis was conducted to determine the independent effects of identified SNPs in single-locus analyses. The expression of quantitative trait loci (eQTL) was also analyzed using data from GTEx and 1000 Genomes Project, and functional prediction of SNPs was performed by using RegulomeDB v2.2, 3DSNP v2.0, HaploReg v4.2 and VannoPortal. Results We found that two novel SNPs of CD82 rs7925603 A > G and PMAIP1 rs4396625 A > T, were significantly and independently associated with OS [adjusted hazards ratios (HRs) and 95% confidence intervals (CI) were 1.27 (1.10-1.48) and 0.77 (0.66-0.91), respectively; P = 0.001 and = 0.002, respectively] and that the combined risk genotypes of these SNPs showed a significant association with OS in patients with HBV-HCC (P trend < 0.001). Further eQTL analysis in the GTEx dataset showed that the rs7925603 G allele was associated with lower CD82 mRNA expression levels, while the rs4396625 T allele was associated with higher PMAIP1 mRNA expression levels in whole blood cells. Conclusion We identified two observed survival-associated SNPs in CD82 and PMAIP1 in the p53 pathway, which influenced HBV-HCC survival possibly through a mechanism of altering mRNA expression. Large studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Liming Qin
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning, People’s Republic of China
| | - Moqin Qiu
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Jingmei Tang
- School of Public Health, Guangxi Medical University, Nanning, People’s Republic of China
| | - Shuyan Liu
- Key Laboratory of Biological Molecular Medicine Research, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, People’s Republic of China
| | - Qiuling Lin
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Qiongguang Huang
- School of Public Health, Guangxi Medical University, Nanning, People’s Republic of China
| | - Xiaoxia Wei
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Qiuping Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Peiqin Chen
- Editorial Department of Chinese Journal of Oncology Prevention and Treatment, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Zihan Zhou
- Department of Cancer Prevention and Control, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Ji Cao
- Department of Cancer Prevention and Control, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Xiumei Liang
- Department of Disease Process Management, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Qian Guo
- Liuzhou Worker’s Hospital, Liuzhou, People’s Republic of China
| | - Cunli Nong
- Liuzhou Worker’s Hospital, Liuzhou, People’s Republic of China
| | - Yizhen Gong
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Yuying Wei
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Yanji Jiang
- Department of Scientific Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Hongping Yu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People’s Republic of China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, People’s Republic of China
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health Commission, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Yingchun Liu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| |
Collapse
|