1
|
Liu Z, Chen X, Ruan Z, Wang C, Yuan D, Xiao W, Li Y, Zhao S. Genetic analysis of comorbidities between osteoarthritis, sarcopenia, and osteoporosis. Exp Gerontol 2025; 206:112788. [PMID: 40389141 DOI: 10.1016/j.exger.2025.112788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/22/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Osteoarthritis (OA), sarcopenia (SCP), and osteoporosis (OP) pose a substantial global morbidity and mortality burden, and previous studies have observed potential associations among them. This study aims to comprehensively characterize the common genetic structure, biological basis, and underlying causal relationship among OA, SCP, and OP. METHODS We used pooled statistics from the largest European genome-wide association study to investigate the genetic overlap and underlying causal relationships among OA, SCP, and OP. LD Score Regression (LDSC) was first used for estimating global and local genetic associations, cross-trait meta-analysis was then conducted to identify shared loci, and mendelian randomization (MR) analysis was performed to test causal association. RESULTS In global and local genetic correlation analysis, we found strong positive correlations among OA, SCP, and OP. Cross-trait meta-analysis revealed 9 novel pleiotropic loci for HandOA_SCP trait-pairs, 1 for ThumbOA_SCP (females), and 6 for KneeOA_SCP (males)0.10 novel pleiotropic loci were also identified for HipOA_TBMD, while none for WLM_FinOP. Bidirectional MR analyses indicated significant causal associations between HandOA and SCP(Forward: OR: 1.41, 95 % CI: 1.25-1.60, p < 0.01,Reverse: OR: 1.77, 95 % CI: 1.34-2.35, p < 0.01). Reverse analyses suggested that ThumbOA.female (OR: 1.92, 95 % CI:1.18-3.13, p < 0.01) and KneeOA.male (OR: 1.58, 95 % CI: 1.13-2.12, p < 0.01) were positively correlated with SCP, while TBMD was positively correlated with HipOA (OR: 1.23, 95 % CI: 1.16-1.31, p < 0.01). CONCLUSIONS Our work demonstrates a shared genetic basis, pleiotropic loci, and putative causal relationships among OA, SCP, and OP, highlighting the intrinsic links behind these three complex skeletal diseases.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - XiangMing Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhe Ruan
- Department of Orthopaedics, The First Hospital of Changsha, Changsha 410005, PR China; The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410008, PR China
| | - Chao Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Dongliang Yuan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Wenfeng Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Shushan Zhao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| |
Collapse
|
2
|
Libedinsky I, Helwegen K, Boonstra J, Simón LG, Gruber M, Repple J, Kircher T, Dannlowski U, van den Heuvel MP. Polyconnectomic Scoring of Functional Connectivity Patterns Across Eight Neuropsychiatric and Three Neurodegenerative Disorders. Biol Psychiatry 2025; 97:1045-1058. [PMID: 39424166 DOI: 10.1016/j.biopsych.2024.10.007] [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: 03/22/2024] [Revised: 09/09/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Neuropsychiatric and neurodegenerative disorders involve diverse changes in brain functional connectivity. As an alternative to approaches that search for specific mosaic patterns of affected connections and networks, we used polyconnectomic scoring to quantify disorder-related whole-brain connectivity signatures into interpretable, personalized scores. METHODS The polyconnectomic score (PCS) measures the extent to which an individual's functional connectivity mirrors the whole-brain circuitry characteristics of a trait. We computed PCSs for 8 neuropsychiatric conditions (attention-deficit/hyperactivity disorder, anxiety-related disorders, autism spectrum disorder, obsessive-compulsive disorder, bipolar disorder, major depressive disorder, schizoaffective disorder, and schizophrenia) and 3 neurodegenerative conditions (Alzheimer's disease, frontotemporal dementia, and Parkinson's disease) across 22 datasets with resting-state functional magnetic resonance imaging data from 10,667 individuals (5325 patients, 5342 control participants). We also examined PCSs in 26,673 individuals from the population-based UK Biobank cohort. RESULTS PCSs were consistently higher in out-of-sample patients across 6 of the 8 neuropsychiatric and across all 3 investigated neurodegenerative disorders ([minimum, maximum]: area under the receiver operating characteristic curve = [0.55, 0.73], false discovery rate-corrected p [pFDR] = [1.8 × 10-16, 4.5 × 10-2]). Individuals with elevated PCS levels for neuropsychiatric conditions exhibited higher neuroticism (pFDR < 9.7 × 10-5), lower cognitive performance (pFDR < 5.3 × 10-5), and lower general well-being (pFDR < 9.7 × 10-4). CONCLUSIONS Our findings reveal generalizable whole-brain connectivity alterations in brain disorders. Polyconnectomic scoring effectively aggregates disorder-related signatures across the entire brain into an interpretable, participant-specific metric. A toolbox is provided for PCS computation.
Collapse
Affiliation(s)
- Ilan Libedinsky
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Koen Helwegen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Jackson Boonstra
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Laura Guerrero Simón
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Marius Gruber
- Institute for Translational Psychiatry, University of Münster, Münster, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany; Center for Mind, Brain and Behavior, University of Marburg and Justus Liebig University Giessen, Giessen, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Martijn P van den Heuvel
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Child and Adolescent Psychiatry and Psychology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Xia C, Alliey-Rodriguez N, Tamminga CA, Keshavan MS, Pearlson GD, Keedy SK, Clementz B, McDowell JE, Parker D, Lencer R, Hill SK, Bishop JR, Ivleva EI, Wen C, Dai R, Chen C, Liu C, Gershon ES. Genetic analysis of psychosis Biotypes: shared Ancestry-adjusted polygenic risk and unique genomic associations. Mol Psychiatry 2025; 30:2673-2685. [PMID: 39709506 DOI: 10.1038/s41380-024-02876-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
The Bipolar-Schizophrenia Network for Intermediate Phenotypes (B-SNIP) created psychosis Biotypes based on neurobiological measurements in a multi-ancestry sample. These Biotypes cut across DSM diagnoses of schizophrenia, schizoaffective disorder, and bipolar disorder with psychosis. Two recently developed post hoc ancestry adjustment methods of Polygenic Risk Scores (PRSs) generate Ancestry-Adjusted PRSs (AAPRSs), which allow for PRS analysis of multi-ancestry samples. Applied to schizophrenia PRS, we found the Khera AAPRS method to show superior portability and comparable prediction accuracy as compared with the Ge method. The three Biotypes of psychosis disorders had similar AAPRSs across ancestries. In genomic analysis of Biotypes, 12 genes, and isoforms showed significant genomic associations with specific Biotypes in a Transcriptome-Wide Association Study (TWAS) of genetically regulated expression (GReX) in the adult brain and fetal brain. TWAS inflation was addressed by the inclusion of genotype principal components in the association analyses. Seven of these 12 genes/isoforms satisfied Mendelian Randomization (MR) criteria for putative causality, including four genes TMEM140, ARTN, C1orf115, CYREN, and three transcripts ENSG00000272941, ENSG00000257176, ENSG00000287733. These genes are enriched in the biological pathways of Rearranged during Transfection (RET) signaling, Neural Cell Adhesion Molecule 1 (NCAM1) interactions, and NCAM signaling for neurite out-growth. The specific associations with Biotypes suggest that pharmacological clinical trials and biological investigations might benefit from analyzing Biotypes separately.
Collapse
Affiliation(s)
- Cuihua Xia
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Ney Alliey-Rodriguez
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL, USA
- Institute of Neuroscience, University of Texas Rio Grande Valley, Harlingen, TX, USA
| | - Carol A Tamminga
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Godfrey D Pearlson
- Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Institute of Living, Hartford Healthcare Corp, Hartford, CT, USA
| | - Sarah K Keedy
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL, USA
| | - Brett Clementz
- Departments of Psychology and Neuroscience, BioImaging Research Center, University of Georgia, Athens, GA, USA
| | - Jennifer E McDowell
- Departments of Psychology and Neuroscience, BioImaging Research Center, University of Georgia, Athens, GA, USA
| | - David Parker
- Departments of Psychology and Neuroscience, BioImaging Research Center, University of Georgia, Athens, GA, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebekka Lencer
- Institute for Translational Psychiatry, Münster University, Münster, Germany
- Department of Psychiatry and Psychotherapy, Lübeck University, Lübeck, Germany
| | - S Kristian Hill
- Department of Psychology, Rosalind Franklin University of Medicine and Science, Chicago, IL, USA
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology and Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Elena I Ivleva
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Cindy Wen
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rujia Dai
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Chao Chen
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
- National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunyu Liu
- MOE Key Laboratory of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Elliot S Gershon
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL, USA.
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Daniel LL, Nepal P, Zanussi J, Dickson AL, Straub P, Miller‐Fleming TW, Wei W, Hung AM, Cox NJ, Kawai VK, Mosley JD, Stein CM, Feng Q, Liu G, Tao R, Chung CP. PTPN2 and Leukopenia in Individuals With Normal TPMT and NUDT15 Metabolizer Status Taking Azathioprine. Clin Transl Sci 2025; 18:e70220. [PMID: 40442974 PMCID: PMC12122386 DOI: 10.1111/cts.70220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 06/02/2025] Open
Abstract
Leukopenia is a common dose-dependent side effect of azathioprine, often leading to drug discontinuation. Variants in TPMT and NUDT15 are associated with azathioprine-induced leukopenia but only explain 25% of cases. Thus, we aimed to identify novel genetic risk factors among TPMT and NUDT15 normal metabolizers through a genome-wide association study (GWAS). Using BioVU, Vanderbilt's electronic health record linked to genetic data, we assembled a discovery cohort of new users of azathioprine. The analysis was conducted in 1184 new users of azathioprine who had no history of prior thiopurine use or an organ transplant. A replication cohort of 521 patients was derived from All of Us, an NIH-funded project that links healthcare data and genetics. The GWAS was adjusted for sex, age, indication (inflammatory bowel disease, systemic lupus erythematosus, other autoimmune condition, or unknown), concurrent use of xanthine oxidase inhibitors (allopurinol or febuxostat) or immunosuppressants, prior TPMT or NUDT15 testing, and 10 principal components of ancestry. In BioVU, 65% of patients were female with a median age of 44 [IQR: 30, 57] and 125 patients developed leukopenia. In All of Us, 69% were female with a median age of 51 [36, 61], and 44 patients developed leukopenia. An intronic variant in PTPN2, rs11664064, reached genome-wide significance in BioVU (OR = 3.61; p = 1.96E-8) and replicated in All of Us (OR = 2.42, p = 0.039). Our finding suggests an association between rs11664064 in PTPN2 and azathioprine-induced leukopenia. PTPN2 plays a role in immune cell development and differentiation, providing a plausible mechanism for this association.
Collapse
Affiliation(s)
- Laura L. Daniel
- Department of MedicineUniversity of MiamiCoral GablesFloridaUSA
| | - Puran Nepal
- Department of MedicineUniversity of MiamiCoral GablesFloridaUSA
| | - Jacy Zanussi
- Department of MedicineUniversity of MiamiCoral GablesFloridaUSA
| | - Alyson L. Dickson
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Peter Straub
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - Wei‐Qi Wei
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Adriana M. Hung
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- VA Tennessee Valley Healthcare SystemNashvilleTennesseeUSA
| | - Nancy J. Cox
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vivian K. Kawai
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jonathan D. Mosley
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - QiPing Feng
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- VA Tennessee Valley Healthcare SystemNashvilleTennesseeUSA
| | - Ge Liu
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ran Tao
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Cecilia P. Chung
- Department of MedicineUniversity of MiamiCoral GablesFloridaUSA
- Miami VA Healthcare SystemMiamiFloridaUSA
| |
Collapse
|
5
|
Tan G, Wang J, Duan J, Xing W. Genetic associations of sex hormones with cerebral aneurysm formation and subarachnoid hemorrhage: A two-sample Mendelian randomization analysis. J Clin Neurosci 2025; 136:111244. [PMID: 40280082 DOI: 10.1016/j.jocn.2025.111244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/19/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND AND AIM Women are more likely than men to develop aneurysmal subarachnoid hemorrhage, and this difference is more pronounced in women after menopause, suggesting a possible correlation between sex hormone levels and cerebral aneurysm formation and rupture. METHODS AND RESULTS We selected genetic variants closely related to estrogen (estradiol), bioavailable testosterone (Bio T), and sex hormone-binding globulin (SHBG) as instrumental variables from the pooled data of the IEU Open GWAS project and cerebral aneurysm (CA) and subarachnoid hemorrhage (SAH) data from two independent datasets from the same study. Two-sample Mendelian randomization was subsequently performed to determine whether the relevant sex hormones and SHBG are causally associated with the formation and rupture of CA. We identified 14 causal associations of related sex hormones and their binding proteins with cerebral aneurysm formation and rupture. Inverse-variance weighting revealed that genetically predicted increased BioT levels reduced the risk of SAH development and genetically predicted increased levels of SHBG in females influenced reduced the risk of cerebral aneurysm formation. After excluding sex differences, weighted mode revealed opposite results, but there was no difference in the IVW, MR-Egger regression, weighted median, or simple mode analyses. No significant effects of the concentrations of other relevant sex hormones or SHBG on the risk of cerebral aneurysm formation or rupture were found. CONCLUSIONS Our study may explain the mechanisms underlying the increased incidence of cerebral aneurysms in menopausal women and provide new directions for intervention in cerebral aneurysm formation and rupture, but further studies are needed.
Collapse
Affiliation(s)
- Guanping Tan
- Department of Cerebrovascular Diseases, Suining Central Hospital, Sichuan Province, China
| | - Jing Wang
- Department of Oncology, Suining Central Hospital, Sichuan Province, China
| | - Jia Duan
- Department of Cerebrovascular Diseases, Suining Central Hospital, Sichuan Province, China
| | - Wenli Xing
- Department of Cerebrovascular Diseases, Suining Central Hospital, Sichuan Province, China.
| |
Collapse
|
6
|
Zhenyu W, Mengyu L, Dongdong D, Jinyi H, Chuanmin Q, Hao Z, Xinjian L, Shenping Z, Wenshui X. A meta-analysis of genome-wide association studies revealed significant QTL and candidate genes for loin muscle area in three breeding pigs. Sci Rep 2025; 15:18758. [PMID: 40436882 PMCID: PMC12119988 DOI: 10.1038/s41598-025-00819-4] [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: 11/19/2024] [Accepted: 03/31/2025] [Indexed: 06/01/2025] Open
Abstract
Loin muscle area (LMA) is an important production trait in pigs and is highly correlated with lean meat percentage. However, the genetic architecture of LMA has not yet been fully elucidated. This study conducted genome-wide association studies (GWAS) and meta-analyses of LMA in Duroc (n = 337), Landrace (n = 662), and Yorkshire pigs (n = 3,176) using imputed whole-genome sequencing to identify new QTLs and candidate genes associated with LMA traits. A total of 108, 34, and 232 significant variants were identified in the Duroc, Landrace, and Yorkshire populations, respectively. The meta-analysis revealed 143 genome-wide significant SNPs and 276 suggestive SNPs, among which 213 were not identified in single population GWAS. Notably, 229 and 413 SNPs were located on SSC16 in the Yorkshire population and meta-analysis, respectively. Based on the 2-LOD drop-off interval, the SSC16 QTL in the Yorkshire population was further narrowed to a 679.835 kb interval (from 32.818 Mb to 33.498 Mb). The most significant variant within this QTL, 16_33228254 (P = 4.45 × 10-9), explained 1.11% phenotypic variance, representing a potential novel locus for LMA. Further bioinformatics analysis determined seven promising candidate genes (NDUFS4, ARL15, FST, ADAM12, DAB2, PLPP1, and SGMS2) with biological processes such as myoblast fusion and positive regulation of transforming growth factor beta receptor signaling pathway. Among them, ARL15 was previously reported in LMA studies, while the other six genes represent novel candidate genes. These findings reveal potential functional genes and pathways associated with LMA, providing valuable insights for future genetic improvement in pigs.
Collapse
Affiliation(s)
- Wang Zhenyu
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Li Mengyu
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Duan Dongdong
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Han Jinyi
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Qiao Chuanmin
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China
| | - Zhou Hao
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Li Xinjian
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China
| | - Zhou Shenping
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China.
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China.
| | - Xin Wenshui
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China.
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China.
| |
Collapse
|
7
|
Miao J, Song G, Wu Y, Hu J, Wu Y, Basu S, Andrews JS, Schaumberg K, Fletcher JM, Schmitz LL, Lu Q. PIGEON: a statistical framework for estimating gene-environment interaction for polygenic traits. Nat Hum Behav 2025:10.1038/s41562-025-02202-9. [PMID: 40410536 DOI: 10.1038/s41562-025-02202-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/02/2025] [Indexed: 05/25/2025]
Abstract
Understanding gene-environment interaction (GxE) is crucial for deciphering the genetic architecture of human complex traits. However, current statistical methods for GxE inference face challenges in both scalability and interpretability. Here we introduce PIGEON-a unified statistical framework for quantifying polygenic GxE using a variance component analytical approach. Based on this framework, we outline the main objectives in GxE studies and introduce an estimation procedure that requires only summary statistics data as input. We demonstrate the effectiveness of PIGEON through theoretical and empirical analyses, including a quasi-experimental gene-by-education study of health outcomes and gene-by-sex interaction for 530 traits using UK Biobank. We also identify genetic interactors that explain the treatment effect heterogeneity in a clinical trial on smoking cessation. PIGEON suggests a path towards polygenic, summary statistics-based inference in future GxE studies.
Collapse
Affiliation(s)
- Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Gefei Song
- University of Wisconsin-Madison, Madison, WI, USA
| | - Yixuan Wu
- University of Wisconsin-Madison, Madison, WI, USA
| | - Jiaxin Hu
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuchang Wu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Shubhashrita Basu
- Department of Economics, Southern Utah University, Cedar City, UT, USA
| | - James S Andrews
- Department of Rheumatology, University of Alabama, Birmingham, AL, USA
| | | | - Jason M Fletcher
- Robert M. La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA
- Department of Population Health Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Lauren L Schmitz
- Robert M. La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
8
|
Qi G, Li Y, Zhang W, Han Z, Chen J, Zhang Z, Xuan L, Chen R, Fang L, Hu Y, Zhang T. Reveal genomic insights into cotton domestication and improvement using gene level functional haplotype-based GWAS. Nat Commun 2025; 16:4734. [PMID: 40399334 PMCID: PMC12095648 DOI: 10.1038/s41467-025-59983-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025] Open
Abstract
Genome-wide association studies (GWAS) are widely used to detect associations between genetic variants and phenotypes. However, few studies have thoroughly analyzed genes, the fundamental and most crucial functional units. Here, we develop an innovative strategy to translate genomic variants into gene-level functional haplotypes (FHs), effectively reducing the interference from complex genome structure and linkage disequilibrium (LD) present in the conventional genetic mapping framework. Using refined mixed linear models, gene-level FH is regressed with 20 cotton agronomic traits across 245 sets of phenotypic values in 3,724 accessions, directly identifying 532 quantitative trait genes (QTGs) with significant breeding potential. The biological function of a superior fiber quality QTG encoding ferulic acid 5-hydroxylase 1 is experimentally validated. Thereafter, we systematically analyze the genetic basis of cotton domestication and improvement at the gene level. This report provides genomic insight into the genetic dissection and efficient mapping of functional genes in plants.
Collapse
Affiliation(s)
- Guoan Qi
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Sanya, Hainan, 572025, China
| | - Yiqian Li
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Wanying Zhang
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zegang Han
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jinwen Chen
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Ziqian Zhang
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Lisha Xuan
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Rui Chen
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Lei Fang
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Sanya, Hainan, 572025, China
| | - Yan Hu
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Sanya, Hainan, 572025, China
| | - Tianzhen Zhang
- The Advanced Seed Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Sanya, Hainan, 572025, China.
| |
Collapse
|
9
|
Hu R, Liang Y, He T, Zhou Y, Lv Y. Causal association of hypertension in family members with preeclampsia-eclampsia in pregnant women: A two-sample Mendelian randomization study. Pregnancy Hypertens 2025; 40:101223. [PMID: 40403523 DOI: 10.1016/j.preghy.2025.101223] [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: 11/13/2024] [Revised: 02/01/2025] [Accepted: 05/11/2025] [Indexed: 05/24/2025]
Abstract
OBJECTIVES The genetic risk factors for hypertension are also high-risk factors for preeclampsia-eclampsia. This study examined the association of hypertension in family members with preeclampsia-eclampsia in pregnant women through two-sample Mendelian randomization (MR). STUDY DESIGN Mendelian randomization. MAIN OUTCOME MEASURES The data for hypertension in siblings, mother, and father were from the UK Biobank, including 364,661, 426,391, and 402,899 individuals, respectively. The data for preeclampsia-eclampsia were FinnGEN R9 (7217 cases and 194,266 controls). Inverse-variance weighted was used as the main analysis method. Weighted median, MR-Egger, simple mode, and weighted mode were complementary MR methods. Heterogeneity was detected using Cochran's Q-test, horizontal pleiotropy using MR-Egger regression, and driving single-nucleotide polymorphisms (SNPs) using the leave-one-out method. RESULTS Mendelian randomization analysis showed that hypertension in family members was positively correlated with preeclampsia-eclampsia risk. The risk of preeclampsia-eclampsia in pregnant women who have siblings with hypertension was the highest (OR = 179.41, 95 % CI: 23.10-1393.65, P = 6.98E-07), followed by hypertension in the mothers (OR = 26.83, 95 % CI: 5.42-132.87, P = 5.56E-05) and the fathers (OR = 18.97, 95 % CI: 1.28-281.29, P = 0.032). The MR-Egger regression test indicated no horizontal pleiotropy (P > 0.05). Cochran's Q-test showed that the effects of the included SNPs exhibited heterogeneity (P < 0.05). The leave-one-out analysis did not reveal SNPs driving the results by themselves. CONCLUSION The risk of preeclampsia-eclampsia in pregnant women who have siblings with hypertension was the highest, followed by pregnant women with a mother or father with hypertension. Having siblings with hypertension should be considered as a high-risk factor for the early prediction of preeclampsia-eclampsia.
Collapse
Affiliation(s)
- Rui Hu
- Department of Obstetrics and Gynecology Intensive Care Unit, Northwest Women's and Children's Hospital, Xi'an, Shaanxi Province 710061, China
| | - Yan Liang
- Department of Obstetrics and Gynecology Intensive Care Unit, Northwest Women's and Children's Hospital, Xi'an, Shaanxi Province 710061, China
| | - Tongqiang He
- Department of Obstetrics and Gynecology Intensive Care Unit, Northwest Women's and Children's Hospital, Xi'an, Shaanxi Province 710061, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology Intensive Care Unit, Northwest Women's and Children's Hospital, Xi'an, Shaanxi Province 710061, China
| | - Yanxiang Lv
- Department of Obstetrics and Gynecology Intensive Care Unit, Northwest Women's and Children's Hospital, Xi'an, Shaanxi Province 710061, China.
| |
Collapse
|
10
|
Sasikumar S, Kumar SP, Bhatt NP, Sinha H. Genome-scale metabolic modelling identifies reactions mediated by SNP-SNP interactions associated with yeast sporulation. NPJ Syst Biol Appl 2025; 11:50. [PMID: 40394077 PMCID: PMC12092771 DOI: 10.1038/s41540-025-00503-3] [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: 09/06/2024] [Accepted: 02/16/2025] [Indexed: 05/22/2025] Open
Abstract
Genome-scale metabolic models (GEMs) are powerful tools used to understand the functional effects of genetic variants. However, the impact of single nucleotide polymorphisms (SNPs) in transcription factors and their interactions on metabolic fluxes remains largely unexplored. Using gene expression data from a yeast allele replacement panel grown during sporulation, we constructed co-expression networks and SNP-specific GEMs. Analysis of co-expression networks revealed that during sporulation, SNP-SNP interactions impact the connectivity of metabolic regulators involved in glycolysis, steroid and histidine biosynthesis, and amino acid metabolism. Further, genome-scale differential flux analysis identified reactions within six major metabolic pathways associated with sporulation efficiency variation. Notably, autophagy was predicted to act as a pentose pathway-dependent compensatory mechanism supplying critical precursors like nucleotides and amino acids, enhancing sporulation. Our study highlights how transcription factor polymorphisms interact to shape metabolic pathways in yeast, offering insights into genetic variants associated with metabolic traits in genome-wide association studies.
Collapse
Affiliation(s)
- Srijith Sasikumar
- Systems Genetics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- Centre for Integrative Biology and Systems Medicine (IBSE), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- Wadhwani School of Data Science and Artificial Intelligence (WSAI), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - S Pavan Kumar
- Centre for Integrative Biology and Systems Medicine (IBSE), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- Wadhwani School of Data Science and Artificial Intelligence (WSAI), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- BioSystems Engineering and Control (BiSECt) Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Nirav Pravinbhai Bhatt
- Centre for Integrative Biology and Systems Medicine (IBSE), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- Wadhwani School of Data Science and Artificial Intelligence (WSAI), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- BioSystems Engineering and Control (BiSECt) Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
- Department of Data Science and Artificial Intelligence, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Himanshu Sinha
- Systems Genetics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
- Centre for Integrative Biology and Systems Medicine (IBSE), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
- Wadhwani School of Data Science and Artificial Intelligence (WSAI), Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
| |
Collapse
|
11
|
Cao Z, Tan Q, Yang H, Xu C. Shared genetic architecture between leukocyte telomere length and Alzheimer's disease. Alzheimers Res Ther 2025; 17:108. [PMID: 40382655 PMCID: PMC12085009 DOI: 10.1186/s13195-025-01757-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 05/07/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Epidemiological and clinical studies have reported an association between leukocyte telomere length (LTL) and Alzheimer's disease (AD). However, genetic association between the two phenotypes remains largely unknown. We aimed to elucidate the potential shared genetic architecture between LTL and AD. METHODS Summary statistics from genome-wide association studies were obtained from large-scale biobank in European-ancestry populations for LTL (N = 472,174) and AD (71,880 cases, 383,378 controls). We examined the global and local genetic correlation between LTL and AD using linkage-disequilibrium score regression and ρ-HESS. We applied the bivariate causal mixture model (MiXeR) to calculate the number of shared genetic causal variants, and the conditional/conjunctional false discovery rate (condFDR/conjFDR) framework to identify specific shared loci between LTL and AD. Bidirectional two-sample Mendelian randomization (MR) were used to explore the causal associations between LTL and AD. RESULTS We detected a significant genetic correlation between LTL and AD (rg = -0.168). Partitioning the whole genome into 1703 almost independent regions, we observed a significant local genetic correlation for LTL and AD at 19q13.32. MiXeR estimated a total of 360 variants affecting LTL, of which 16 was estimated to influence AD. The condFDR revealed an essential genetic enrichment in LTL conditional on associations with AD, and vice versa. We next identified 8 shared genomic loci between LTL and AD using conjFDR method, of which 4 are novel loci for both the phenotypes. Moreover, 3 shared loci were identified as eQTLs (rs3098168, rs4780338 and rs2680702). All shared loci mapped a subset of 48 credible genes, including USP8, DEXI and APOE. Gene-set analysis identified 18 putative gene sets enriched with the genes mapped to the shared loci. MR analysis suggested that genetically determined AD was causally associated with LTL. CONCLUSION Our study identified specific shared loci between LTL and AD, providing new insights for polygenic overlap and molecular mechanisms, and highlighting new opportunities for future experimental validation.
Collapse
Affiliation(s)
- Zhi Cao
- Department of Psychiatry, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Public Health, Hangzhou Normal University, NO.2318, Yuhangtang Road, Yuhang District, Hangzhou, 311121, China
| | - Qilong Tan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongxi Yang
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chenjie Xu
- School of Public Health, Hangzhou Normal University, NO.2318, Yuhangtang Road, Yuhang District, Hangzhou, 311121, China.
| |
Collapse
|
12
|
Xu J, Jiang X, Yin X, Zhao X, Chen N, Pan L, Fu C, Jiao Y, Ma J, Yuan M, Chi X. Genome-wide association analysis in peanut accessions uncovers the genetic basis regulating oil and fatty acid variation. BMC PLANT BIOLOGY 2025; 25:651. [PMID: 40380082 PMCID: PMC12082984 DOI: 10.1186/s12870-025-06690-9] [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] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND The cultivated peanut, Arachis hypogaea L., is a critical oil and food crop worldwide. Improving seed oil quality in peanut has long been an aim of breeders. However, our knowledge of the genetic basis of selecting for seed nutritional traits is limited. Based on AhFAD2A and AhFAD2B, scientists have now developed higher oleic acid (80-84%) in peanut. Decoding the genetic makeup behind natural variation in kernel oil and fatty acid concentrations is crucial for molecular breeding-based nutrient quantity and quality manipulation. RESULTS Herein, we recognized 87 quantitative trait loci (QTLs) in 45 genomic regions for the concentrations of oil, oleic acid, and linoleic acid, as well as the oleic acid to linoleic acid (O/L) ratio via a genome-wide association study (GWAS) involving 499 peanut accessions. Eight QTLs explained more than 15% of the phenotypic variation in peanut accessions. Among the 45 potential genes significantly related to the four traits, only three genes displayed annotation to the fatty acid pathway. Furthermore, on the basis of pleiotropism or linkage data belonging to the identified singular QTLs, we generated a trait-locus axis to better elucidate the genetic background behind the observed oil and fatty acid concentration association. Expression analysis indicated that arahy.AV6GAN and arahy.NNA8KD have higher expressions in the seeds. CONCLUSION This natural population consisting of 499 peanut accessions combined with high-density SNPs will provide a better choice for identifying peanut QTLs/genes in the future. Together, our results provide strong evidence for the genetic mechanism behind oil biosynthesis in peanut, facilitating future advances in multiple fatty acid component generation via pyramiding of desirable QTLs.
Collapse
Grants
- ZR2021QC172, ZR2023QC146 Natural Science Foundation of Shangdong Province
- ZR2021QC172, ZR2023QC146 Natural Science Foundation of Shangdong Province
- 2024LZGC035 Key R&D Program of Shandong Province
- KF2024007 Open Project of Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs, P. R. China
- CXGC2023F20, CXGC2024F20, CXGC2024G20 the innovation Project of SAAS
- CXGC2023F20, CXGC2024F20, CXGC2024G20 the innovation Project of SAAS
- tstp20240523, tsqn202312292 Taishan Scholars Program
- tstp20240523, tsqn202312292 Taishan Scholars Program
- 2022E10012 Open Project of Key Laboratory of Digital Upland Crops of Zhejiang Province
- 2018GNC110036, 2022TZXD0031 Key research and development plan of Shandong Province
- 2018GNC110036, 2022TZXD0031 Key research and development plan of Shandong Province
- 2022A02008-3 Major scientific and technological project in Xinjiang
- CARS-13 China Agriculture Research System of MOF and MARA
- Key R&D Program of Shandong Province
Collapse
Affiliation(s)
- Jing Xu
- Shandong Peanut Research Institute, Qingdao, CN-266100, China
| | - Xiao Jiang
- Shandong Peanut Research Institute, Qingdao, CN-266100, China
| | - Xiangzhen Yin
- Shandong Peanut Research Institute, Qingdao, CN-266100, China
| | - Xuhong Zhao
- Shandong Peanut Research Institute, Qingdao, CN-266100, China
| | - Na Chen
- Shandong Peanut Research Institute, Qingdao, CN-266100, China
| | - Lijuan Pan
- Shandong Peanut Research Institute, Qingdao, CN-266100, China
| | - Chun Fu
- Weifang Academy of Agricultural Sciences, Weifang, CN-261071, China
| | - Yanlin Jiao
- Yantai Academy of Agricultural Sciences, Yantai, CN-265500, China
| | - Junqing Ma
- Shandong Peanut Research Institute, Qingdao, CN-266100, China
| | - Mei Yuan
- Shandong Peanut Research Institute, Qingdao, CN-266100, China.
| | - Xiaoyuan Chi
- Shandong Peanut Research Institute, Qingdao, CN-266100, China.
| |
Collapse
|
13
|
Akoth M, Odhiambo J, Omolo B. Genome-wide association studies on malaria in Sub-Saharan Africa: A scoping review. PLoS One 2025; 20:e0309268. [PMID: 40378106 PMCID: PMC12083797 DOI: 10.1371/journal.pone.0309268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 04/02/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Malaria remains one of the leading causes of death in Sub-Saharan Africa (SSA). The scoping review mapped evidence in research on existing studies on malaria genome-wide association studies (GWAS) in SSA. METHODS A scoping review was conducted to map existing studies in genome-wide association on malaria in SSA, with a review period between 1st January 2000 and 31st December 2024. The searches were made with the last search done in January 2025. The extracted data were analyzed using R software and SRplot. Relevant studies were identified through electronic searching of Google Scholar, Pubmed, Scopus, and Web of Science databases. Two independent reviewers followed the inclusion-exclusion criteria to extract relevant studies. Data from the studies were collected and synthesized using Excel and Zotero software. RESULTS We identified 89 studies for inclusion. Most of these studies (n = 42, [Formula: see text]) used a case-control study design, while the rest used cross-sectional, cohort, longitudinal, family-based, and experimental study designs. These studies were conducted between 2000 and 2024, with a noticeable increase in publications from 2012. Most studies were carried out in Kenya (n = 23), Gambia (n = 18), Cameroon (n = 15), and Tanzania (n = 9), primarily exploring genetic variants associated with malaria susceptibility, resistance, and severity. CONCLUSION Many case-control studies in Kenya and Gambia reported genetic variants in malaria susceptibility, resistance, and severity. GWAS on malaria is scarce in SSA, and even fewer studies are model-based. Consequently, there is a pressing need for more genome-wide research on malaria in SSA.
Collapse
Affiliation(s)
- Morine Akoth
- Strathmore Institute of Mathematical Sciences, Strathmore University, Ole Sangale Road, Nairobi, Kenya
| | - John Odhiambo
- Strathmore Institute of Mathematical Sciences, Strathmore University, Ole Sangale Road, Nairobi, Kenya
| | - Bernard Omolo
- Strathmore Institute of Mathematical Sciences, Strathmore University, Ole Sangale Road, Nairobi, Kenya
- Division of Mathematics & Computer Science, University of South Carolina-Upstate, Spartanburg, South Carolina, USA
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, South Africa
| |
Collapse
|
14
|
Paye SM, Edge MD. Mathematical bounds on r 2 and the effect size in case-control genome-wide association studies. Theor Popul Biol 2025; 164:1-11. [PMID: 40381956 DOI: 10.1016/j.tpb.2025.04.003] [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: 12/17/2024] [Revised: 04/19/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025]
Abstract
Case-control genome-wide association studies (GWAS) are often used to find associations between genetic variants and diseases. When case-control GWAS are conducted, researchers must make decisions regarding how many cases and how many controls to include in the study. Connections between variants and diseases are made using association statistics, including χ2. Previous work in population genetics has shown that LD statistics, including r2, are bounded by the allele frequencies in the population being studied. Since varying the case fraction changes sample allele frequencies, we use the known bounds on r2 to explore how the fraction of cases included in a study can affect statistical power to detect associations. We analyze a simple mathematical model and use simulations to study a quantity proportional to the χ2 noncentrality parameter, which is closely related to r2, under various conditions. Varying the case fraction changes the χ2 noncentrality parameter, and by extension the statistical power, with effects depending on the dominance, penetrance, and frequency of the risk allele. Our framework explains previously observed results, such as asymmetries in power to detect risk vs. protective alleles, and the fact that a balanced sample of cases and controls does not always give the best power to detect associations, particularly for highly penetrant minor risk alleles that are either dominant or recessive. We show by simulation that our results can be used as a rough guide to statistical power for association tests other than χ2 tests of independence.
Collapse
Affiliation(s)
- Sanjana M Paye
- Department of Quantitative and Computational Biology, University of Southern California, United States of America; University of Michigan Medical Scientist Training Program, United States of America
| | - Michael D Edge
- Department of Quantitative and Computational Biology, University of Southern California, United States of America.
| |
Collapse
|
15
|
Li R, Su K, Wu T, Xu L, Song W, Sun D, Zeng T, Chen J, Xin H, Li Y, Zang M, Hu M. Genome-wide enhancer-gene regulatory maps of liver reveal novel regulatory mechanisms underlying NAFLD pathogenesis. BMC Genomics 2025; 26:493. [PMID: 40375105 PMCID: PMC12082939 DOI: 10.1186/s12864-025-11668-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) represents the most widespread liver disease globally, ranging from non-alcoholic fatty liver (NAFL) and steatohepatitis (NASH) to fibrosis/cirrhosis, with potential progression to hepatocellular carcinoma (HCC). Genome-wide association studies (GWASs) have identified several single nucleotide polymorphisms (SNPs) associated with NAFLD. However, numerous GWAS signals associated with NAFLD locate in non-coding regions, posing a challenge for interpreting their functional annotation. RESULTS In this study, we utilized the Activity-by-Contact (ABC) model to construct the enhancer-gene maps of liver by integrating epigenomic data from 15 liver tissues and cell lines. We constructed the most comprehensive genome-wide regulatory maps of the liver, identifying 543,486 enhancer-gene connections, including 267,857 enhancers and 16,872 target genes. Enrichment analyses revealed that the ABC SNPs are significantly enriched in active chromatin regions and active chromatin state. By combining the ABC regulatory maps and NAFLD GWAS data, we systematically identified ABC SNPs associated with NAFLD risk. Through the functional annotations, such as pathway enrichment and drug-gene interaction analyses, we identified 6 genes (GGT1, ACTG1, SPP1, EPHA2, PROZ and SHMT1) as candidate NAFLD genes, with SHMT1 previously reported. Among the SNPs connected to the candidate genes, the ABC SNP rs2017869 (odds ratio [OR] for the C allele = 1.10, 95% CI = 1.04-1.16, P = 5.97 × 10- 4) had the highest ABC score. According to the ABC maps, rs2017869 links to GGT1, and several drugs targeting this gene, such as liothyronine, showed potential benefits to patients with NAFLD. Furthermore, we identified that another novel gene, EPHA2, may play a crucial role in NAFLD by regulating the GGT levels. CONCLUSIONS Our study provides the most comprehensive ABC regulatory maps of the liver to date. This resource offers a valuable reference for identifying regulatory variants and prioritizing susceptibility genes of liver diseases, such as NAFLD.
Collapse
Affiliation(s)
- Ruofan Li
- Medical School of Chinese People's Liberation Army (PLA), 28 Fuxing Road, 100853, Beijing, China
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Kaiyan Su
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, 1,838 North Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Tianzhun Wu
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Li Xu
- Department of Hepatopancreatobiliary Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wenyu Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Dandan Sun
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Tao Zeng
- Medical School of Chinese People's Liberation Army (PLA), 28 Fuxing Road, 100853, Beijing, China
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Jinzhang Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, 1,838 North Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| | - Haibei Xin
- Department of Hepatobiliary Surgery, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Yuanfeng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| | - Mengya Zang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, 1,838 North Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| | - Minggen Hu
- Medical School of Chinese People's Liberation Army (PLA), 28 Fuxing Road, 100853, Beijing, China.
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
16
|
Han Y, Du Q, Dai Y, Gu S, Lei M, Liu W, Zhang W, Zhu M, Feng L, Si H, Liu J, Zan Y. EasyOmics: A graphical interface for population-scale omics data association, integration, and visualization. PLANT COMMUNICATIONS 2025; 6:101293. [PMID: 40017036 DOI: 10.1016/j.xplc.2025.101293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/16/2025] [Accepted: 02/26/2025] [Indexed: 03/01/2025]
Abstract
The rapid growth of population-scale whole-genome resequencing, RNA sequencing, bisulfite sequencing, and metabolomic and proteomic profiling has led quantitative genetics into the era of big omics data. Association analyses of omics data, such as genome-, transcriptome-, proteome-, and methylome-wide association studies, along with integrative analyses of multiple omics datasets, require various bioinformatics tools, which rely on advanced programming skills and command-line interfaces and thus pose challenges for wet-lab biologists. Here, we present EasyOmics, a stand-alone R Shiny application with a user-friendly interface that enables wet-lab biologists to perform population-scale omics data association, integration, and visualization. The toolkit incorporates multiple functions designed to meet the increasing demand for population-scale omics data analyses, including data quality control, heritability estimation, genome-wide association analysis, conditional association analysis, omics quantitative trait locus mapping, omics-wide association analysis, omics data integration, and visualization. A wide range of publication-quality graphs can be prepared in EasyOmics by pointing and clicking. EasyOmics is a platform-independent software that can be run under all operating systems, with a docker container for quick installation. It is freely available to non-commercial users at Docker Hub https://hub.docker.com/r/yuhan2000/easyomics.
Collapse
Affiliation(s)
- Yu Han
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266000, China; Key Laboratory for Bio-Resource and Eco-Environment of Ministry of Education & Sichuan Zoige Alpine Wetland Ecosystem National Observation and Research Station, College of Life Science, Sichuan University, Chengdu 610065, China; Department of Plant Physiology, Umeå Plant Science Center and Integrated Science Lab, Umeå University, Umeå, Sweden
| | - Qiao Du
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266000, China
| | - Yifei Dai
- Biostatistics Department, School of Public Health, University of Michigan, Ann Arbor, MI 48105, USA
| | - Shaobo Gu
- Key Laboratory for Bio-Resource and Eco-Environment of Ministry of Education & Sichuan Zoige Alpine Wetland Ecosystem National Observation and Research Station, College of Life Science, Sichuan University, Chengdu 610065, China
| | - Mengyu Lei
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266000, China
| | - Wei Liu
- Key Laboratory for Bio-Resource and Eco-Environment of Ministry of Education & Sichuan Zoige Alpine Wetland Ecosystem National Observation and Research Station, College of Life Science, Sichuan University, Chengdu 610065, China
| | - Wenjia Zhang
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266000, China
| | - Mingjia Zhu
- State Key Laboratory of Herbage Innovation and Grassland Agro-ecosystem, College of Ecology, Lanzhou University, Lanzhou 730000, China
| | - Landi Feng
- Key Laboratory for Bio-Resource and Eco-Environment of Ministry of Education & Sichuan Zoige Alpine Wetland Ecosystem National Observation and Research Station, College of Life Science, Sichuan University, Chengdu 610065, China
| | - Huan Si
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266000, China
| | - Jianquan Liu
- State Key Laboratory of Herbage Innovation and Grassland Agro-ecosystem, College of Ecology, Lanzhou University, Lanzhou 730000, China.
| | - Yanjun Zan
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266000, China; Department of Plant Physiology, Umeå Plant Science Center and Integrated Science Lab, Umeå University, Umeå, Sweden.
| |
Collapse
|
17
|
Xu L, He R, Ye X, Wang Y, Hui S, Li H, Chen H, Huang P. Leveraging transcriptome-wide association studies identifies the relationship between upper respiratory flora and cell type-specific gene expression in severe respiratory disease. PLoS One 2025; 20:e0322864. [PMID: 40343915 PMCID: PMC12063895 DOI: 10.1371/journal.pone.0322864] [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: 11/15/2024] [Accepted: 03/30/2025] [Indexed: 05/11/2025] Open
Abstract
ObjectivesThe upper respiratory tract flora may influence host immunity and modulate susceptibility to viral respiratory infections. This study aimed to investigate the associations between upper respiratory tract flora and immune cells in severe ILI, identify specific microbial taxa and immune response pathways contributing to disease severity, and elucidate how flora influences ILI progression by modulating immune cell functions.MethodsHeritability of GWAS summary data was estimated using LDSC (v1.0.1). Gene-level genetic associations were analyzed with MAGMA. scRNA-seq data were integrated with genetic association data using scDRS. FUSION was used to construct cell type-specific expression quantitative trait locus models based on genotypes and scRNA-seq data from the onek1k project, which were combined with flora abundance-related GWAS data for a transcriptome-wide association study.ResultsFrom the LDSC analysis, data from 1195 severe ILI-associated GWASs with upper respiratory flora(h2 > 0.1) were included in subsequent analysis. TWAS identified 19 significant association pairs (Padj < 0.05), and 1226 differentially expressed genes between mild and severe ILI patients (Padj < 0.05 and | log2FC|>0.25). Functional enrichment analyses using GO, KEGG, and Reactome databases revealed that immune cells,such as CD4 + T effector memory cells, cDCs, NK cells, were enriched in multiple biological processes or pathways.ConclusionsThis study identified associations between severe ILI-related upper respiratory tract flora and cell type-specific gene expression, potentially explaining how differential flora influences ILI progression. CD16 + monocytes exhibited the most differentially expressed genes, followed by proliferating cells and cDCs, highlighting the significant role of immune cell-enriched pathways in ILI progression.
Collapse
Affiliation(s)
- Lei Xu
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Ran He
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Xiangyu Ye
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Yifan Wang
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Shirong Hui
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Haochang Li
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Hongbo Chen
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Peng Huang
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Costa KA, Araujo AC, Fonseca PADS, Silva HT, Menegatto LS, de Freitas LA, Cardoso CM, Carvalho Filho I, Otto PI, Costa RLDD, Stafuzza NB, Paz CCPD. Genetic parameters and haplotype-based genome-wide association study of indicator traits for gastrointestinal parasite resistance in Santa Ines sheep. Vet Parasitol 2025; 337:110498. [PMID: 40359809 DOI: 10.1016/j.vetpar.2025.110498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
Genetic parameters are of great importance in animal breeding since they determine the strategies necessary to increase the genetic progress in economic traits, for example indicator traits for resistance to endoparasites in sheep. Furthermore, genetic markers have been used to identify genomic regions associated with economically important traits, which can help increase the genetic response through genomic selection. Therefore, this study aimed to estimate genetic parameters and perform a haplotype-based genome wide association study (GWAS) to identify genomic regions associated with indicator traits for endoparasite resistance in Santa Ines sheep. Haplotype GWAS was performed using linkage disequilibrium blocks (haploblocks) defined by the Haploview software. Records from 1725 animals for Famacha© (FAM, N = 5560), packed cell volume (PCV, N = 5135), total plasma protein (TPP, N = 4356), and fecal egg count (FEC, N = 4248) were used for analysis. The pedigree file contained information from 4821 animals; of these, 638 animals were genotyped using the Ovine SNP50 Genotyping BeadChip. Heritability estimates were moderate for FAM (0.26), PCV (0.26), TPP (0.16), and log-transformed FEC (0.19). Genomic regions that explained more than 0.3 % of the total additive genetic variance of the traits were defined as significant. These regions overlapped with quantitative traits loci associated with eosinophil number, fecal egg count, hematocrit, and immunoglobulin levels. In the top regions for the traits evaluated we found genes individually involved in inflammatory response, immunity, macrophage function in host-pathogen interactions, and other biological functions. All indicator traits for resistance to gastrointestinal parasites evaluated in this study exhibited sufficient genetic variability to respond to selection and can be used to improve the health and consequently the production of Santa Ines sheep.
Collapse
Affiliation(s)
- Karine Assis Costa
- Beef Cattle Research Center, Animal Science Institute, Sertãozinho 14174-000, Brazil.
| | | | - Pablo Augusto de Souza Fonseca
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph N1G 2W1, Canada.
| | - Hugo Teixeira Silva
- Department of Animal Science, Federal University of Viçosa, Viçosa 36570-900, Brazil.
| | - Leonardo Sartori Menegatto
- Department of Genetics, Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Ribeirão Preto 14049-900, Brazil.
| | - Luara Afonso de Freitas
- Department of Genetics, Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Ribeirão Preto 14049-900, Brazil.
| | - Cleyce Maiara Cardoso
- Beef Cattle Research Center, Animal Science Institute, Sertãozinho 14174-000, Brazil.
| | - Ivan Carvalho Filho
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, Brazil.
| | - Pamela Itajara Otto
- Department of Animal Science, University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil.
| | | | | | - Claudia Cristina Paro de Paz
- Department of Genetics, Ribeirão Preto Medical School (FMRP), University of São Paulo (USP), Ribeirão Preto 14049-900, Brazil; Sustainable Livestock Research Center, Animal Science Institute, São José do Rio Preto 15130-000, Brazil.
| |
Collapse
|
19
|
Dutta D, Chatterjee N. Expanding scope of genetic studies in the era of biobanks. Hum Mol Genet 2025:ddaf054. [PMID: 40312842 DOI: 10.1093/hmg/ddaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Biobanks have become pivotal in genetic research, particularly through genome-wide association studies (GWAS), driving transformative insights into the genetic basis of complex diseases and traits through the integration of genetic data with phenotypic, environmental, family history, and behavioral information. This review explores the distinct design and utility of different biobanks, highlighting their unique contributions to genetic research. We further discuss the utility and methodological advances in combining data from disease-specific study or consortia with that of biobanks, especially focusing on summary statistics based meta-analysis. Subsequently we review the spectrum of additional advantages offered by biobanks in genetic studies in representing population differences, calibration of polygenic scores, assessment of pleiotropy and improving post-GWAS in silico analyses. Advances in sequencing technologies, particularly whole-exome and whole-genome sequencing, have further enabled the discovery of rare variants at biobank scale. Among recent developments, the integration of large-scale multi-omics data especially proteomics and metabolomics, within biobanks provides deeper insights into disease mechanisms and regulatory pathways. Despite challenges like ascertainment strategies and phenotypic misclassification, biobanks continue to evolve, driving methodological innovation and enabling precision medicine. We highlight the contributions of biobanks to genetic research, their growing integration with multi-omics, and finally discuss their future potential for advancing healthcare and therapeutic development.
Collapse
Affiliation(s)
- Diptavo Dutta
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD, 20879, United States
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins University, 615 N Wolfe Street, Baltimore, MD, 21205, United States
- Department of Oncology, Johns Hopkins University, 615 N Wolfe Street, Baltimore, MD, 21205, United States
| |
Collapse
|
20
|
Yang Y, Zhang Y, Liu K, Liu M, Zhang H, Guo M. IFI27, a potential candidate molecular marker for primary Sjogren's syndrome. Clin Rheumatol 2025; 44:1949-1960. [PMID: 40146445 DOI: 10.1007/s10067-025-07409-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 03/04/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
OBJECTIVE The etiology of primary Sjogren's syndrome (pSS) is complex and not completely clear. This study was to identify key genes in pSS based on Gene Expression Omnibus (GEO). METHODS We downloaded the GSE40568, GSE80805, GSE127952, and GSE164885 mRNA expression profiles from GEO. Differentially expressed genes (DEGs) analyses were carried out by using the online analysis tool GEO2R and R. Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to elucidate the biological processes, molecular function, cellular component, and KEGG signaling pathways for the DEGs in salivary glands (SGs) and peripheral blood mononuclear cells (PBMCs). Genes co-expressed were found in PBMCs and SGs of pSS patients. RT-qPCR was performed for validation. Finally, clinical correlation analysis and receiver operator characteristic (ROC) curve analysis were performed. RESULTS A total of thirty-nine up-regulated and one down-regulated genes were identified in pSS SGs. GO and KEGG pathway revealed that these DEGs were related to response to virus, and type I interferon signaling pathway. It was verified that fourteen genes were up-regulated in the SGs of pSS by RT-qPCR. Twenty up-regulated genes were identified in pSS patients PBMCs. Two genes were up-regulated in SGs and PBMCs of pSS patients, including IFI27 and IFI44L. The mRNA level of IFI27 was positively correlated with the disease activity of pSS patients. Furthermore, ROC analyses proved IFI27 may have diagnostic value for pSS. CONCLUSION IFI27 might serve as a potential biomarker for the early diagnosis and therapy of pSS. Key Points • IFI27 expression was significantly increased in PBMCs and SGs of pSS patients. • IFI27 was positively correlated with disease activity in pSS patients. • IFI27 might have a good diagnostic value for pSS.
Collapse
Affiliation(s)
- Yiying Yang
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
- Postdoctoral Research Station of Biology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Ying Zhang
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Ke Liu
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Meidong Liu
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, 410000, Changsha, Hunan, China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Jiang L, Shen M, Zhang S, Zhang J, Shi Y, Gu Y, Yang T, Fu Q, Wang B, Chen Y, Xu K, Chen H. A regulatory variant rs9379874 in T1D risk region 6p22.2 affects BTN3A1 expression regulating T cell function. Acta Diabetol 2025; 62:695-706. [PMID: 39417845 DOI: 10.1007/s00592-024-02389-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Genome-wide association studies (GWAS) have identified that 6p22.2 region is associated with type 1 diabetes (T1D) risk in the Chinese Han population. This study aims to reveal associations between this risk region and T1D subgroups and related clinical features, and further identify causal variant(s) and target gene(s) in this region. METHODS 2608 T1D and 4814 healthy controls were recruited from East, Central, and South China. Baseline data and genotyping for rs4320356 were collected. The most likely causal variant and gene were identified by bioinformatics analysis, dual-luciferase reporter assays, expression quantitative trait loci (eQTL), and functional annotation of the non-coding region within the 6p22.2 region. RESULTS The leading variant rs4320356 in the 6p22.2 region was associated with T1D risk in the Chinese and Europeans. However, this variant was not significantly associated with islet function or autoimmunity. In silico analysis suggested rs9379874 was the most potential causal variant for T1D risk among thymus, spleen, and T cells, overlapping with the enhancer-related histone mark in multiple T cell subsets. Dual luciferase reporter assay and eQTL showed that the T allele of rs9379874 increased BTN3A1 expression by binding to FOXA1. Public single-cell RNA sequencing analysis indicated that BTN3A1 was related to T-cell activation, ATP metabolism, and cytokine metabolism pathways, which might contribute to T1D development. CONCLUSION This study indicates that a functional variant rs9379874 regulates BTN3A1 expression, expanding the genomic landscape of T1D risk and offering a potential target for developing novel therapies.
Collapse
Affiliation(s)
- Liying Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Department of Rehabilitation Medicine, Lishui People's Hospital, Lishui, 323000, Zhejiang, China
| | - Min Shen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Saisai Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jie Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yun Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yong Gu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qi Fu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bingwei Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yang Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Kuanfeng Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Heng Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
22
|
Leventhal L, Ruffley M, Exposito-Alonso M. Planting Genomes in the Wild: Arabidopsis from Genetics History to the Ecology and Evolutionary Genomics Era. ANNUAL REVIEW OF PLANT BIOLOGY 2025; 76:605-635. [PMID: 39971350 DOI: 10.1146/annurev-arplant-071123-095146] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The genetics model system Arabidopsis thaliana (L.) Heynh. lives across a vast geographic range with contrasting climates, in response to which it has evolved diverse life histories and phenotypic adaptations. In the last decade, the cataloging of worldwide populations, DNA sequencing of whole genomes, and conducting of outdoor field experiments have transformed it into a powerful evolutionary ecology system to understand the genomic basis of adaptation. Here, we summarize new insights on Arabidopsis following the coordinated efforts of the 1001 Genomes Project, the latest reconstruction of biogeographic and demographic history, and the systematic genomic mapping of trait natural variation through 15 years of genome-wide association studies. We then put this in the context of local adaptation across climates by summarizing insights from 73 Arabidopsis outdoor common garden experiments conducted to date. We conclude by highlighting how molecular and genomic knowledge of adaptation can help us to understand species' (mal)adaptation under ongoing climate change.
Collapse
Affiliation(s)
- Laura Leventhal
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California, USA
- Department of Biology, Stanford University, Stanford, California, USA
- Department of Integrative Biology, University of California, Berkeley, California, USA
| | - Megan Ruffley
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California, USA
- Department of Integrative Biology, University of California, Berkeley, California, USA
| | - Moises Exposito-Alonso
- Department of Plant Biology, Carnegie Institution for Science, Stanford, California, USA
- Department of Biology, Stanford University, Stanford, California, USA
- Department of Global Ecology, Carnegie Institution for Science, Stanford, California, USA
- Department of Integrative Biology, University of California, Berkeley, California, USA
- Howard Hughes Medical Institute, University of California, Berkeley, California, USA;
| |
Collapse
|
23
|
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, The PigGTEx Consortium, 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
|
24
|
Bang NN, Hayes BJ, Lyons RE, Randhawa IAS, Gaughan JB, Trach NX, McNeill DM. Genomic Prediction and Genome-Wide Association Studies for Productivity, Conformation and Heat Tolerance Traits in Tropical Smallholder Dairy Cows. J Anim Breed Genet 2025; 142:322-341. [PMID: 39462234 DOI: 10.1111/jbg.12907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/28/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
Genomic selection (GS) and genome-wide association studies (GWAS) have not been investigated in Vietnamese dairy cattle, even for basic milk production traits, largely due to the scarcity of individual phenotype recording in smallholder dairy farms (SDFs). This study aimed to estimate heritability (h 2) and test the applicability of GS and GWAS for milk production, body conformation and novel heat tolerance traits using single test day phenotypic data. Thirty-two SDFs located in either the north (a lowland vs. a highland) or the south (a lowland vs. a highland) of Vietnam were each visited for an afternoon and the next morning to collect phenotype data of all lactating cows (n = 345). Tail hair from each cow was sampled for subsequent genotyping with a 50K SNP chip at that same visit. Milk production traits (single-test day) were milk yield (MILK, kg/cow/day), energy corrected milk yield adjusted for body weight (ECMbw, kg/100 kg BW/day), fat (mFA, %), protein (mPR, %) and dry matter (mDM, %). Conformation traits were body weight (BW, kg) and body condition score (BCS, 1 = thin to 5 = obese). Heat tolerance traits were panting score (PS, 0 = normal to 4.5 = extremely heat-stressed) and infrared temperatures (IRTs, °C) at 11 areas on the external body surface of the cow (inner vulval lip, outer vulval surface, inner tail base surface, ocular area, muzzle, armpit area, paralumbar fossa area, fore udder, rear udder, forehoof and hind hoof), assessed by an Infrared Camera. Univariate linear mixed models and a 10-fold cross-validation approach were applied for GS. Univariate single SNP mixed linear models were applied for the GWAS. Estimated h 2 (using the genotype information to build relationships among animals) were moderate (0.20-0.37) for ECMbw, mFA, mPR, mRE, BW, BCS and IRT at rear udder; low (0.08-0.19) for PS and other IRTs; and very low (≤ 0.07) for MILK, ECM and mDM. Accuracy of genomic estimated breeding values (GEBVs) was low (≤ 0.12) for MILK, ECM, mDM and IRT at hind hoof; and moderate to high (0.32-0.46) for all other traits. The most significant regions on chromosomes (BTA) associated with milk production traits were 0.47-1.18 Mb on BTA14. Moderate to high h 2 and moderate accuracies of GEBVs for mFA, mPR, ECMbw, BCS, BW, PS and IRTs at rear udder and outer vulval surface suggested that GS using single test day phenotypic data could be applied for these traits. However, a greater sample size is required to decrease the bias of GEBVs by GS and increase the power of detecting significant quantitative trait loci (QTLs) by GWAS.
Collapse
Affiliation(s)
- Nguyen N Bang
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
- Faculty of Animal Science, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Ben J Hayes
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, Queensland, Australia
| | - Russell E Lyons
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Imtiaz A S Randhawa
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - John B Gaughan
- School of Agriculture and Food Sciences, The University of Queensland, Gatton, Queensland, Australia
| | - Nguyen X Trach
- Faculty of Animal Science, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - David M McNeill
- School of Environmental and Rural Science, University of New England, Armidale, New South Wales, Australia
| |
Collapse
|
25
|
Keskitalo S, Seppänen MRJ, Del Sol A, Varjosalo M. From rare to more common: The emerging role of omics in improving understanding and treatment of severe inflammatory and hyperinflammatory conditions. J Allergy Clin Immunol 2025; 155:1435-1450. [PMID: 39978687 DOI: 10.1016/j.jaci.2025.02.011] [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: 10/07/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Inflammation is a pathogenic driver of many diseases, including atherosclerosis and rheumatoid arthritis. Hyperinflammation can be seen as any inflammatory response that is deleterious to the host, regardless of cause. In medicine, hyperinflammation is defined as severe, deleterious, and fluctuating systemic or local inflammation with presence of a cytokine storm. It has been associated with rare autoinflammatory disorders. However, advances in omics technologies, including genomics, proteomics, and metabolomics, have revealed it to be more common, occurring in sepsis and severe coronavirus disease 2019. With a focus on proteomics, this review highlights the key role of omics in this shift. Through an exploration of research, we present how omics technologies have contributed to improved diagnostics, prognostics, and targeted therapeutics in the field of hyperinflammation. We also discuss the integration of advanced technologies, multiomics approaches, and artificial intelligence in analyzing complex datasets to develop targeted therapies, and we address their potential for revolutionizing the clinical aspects of hyperinflammation. We emphasize personalized medicine approaches for effective treatments and outline challenges, including the need for standardized methodologies, robust bioinformatics tools, and ethical considerations regarding data privacy. This review aims to provide a comprehensive overview of the molecular mechanisms underpinning hyperinflammation and underscores the potential of omics technologies in enabling successful clinical management.
Collapse
Affiliation(s)
- Salla Keskitalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Mikko R J Seppänen
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland; Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; European Reference Network Rare Immunodeficiency Autoinflammatory and Autoimmune Diseases Network (ERN RITA) Core Center, Helsinki, The Netherlands
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Computational Biology Group, Basque Research and Technology Alliance (CIC bioGUNE-BRTA), Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland; Department of Biochemistry and Developmental Biology and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
26
|
Ryzhkov FV, Ryzhkova YE, Elinson MN. Machine learning: Python tools for studying biomolecules and drug design. Mol Divers 2025:10.1007/s11030-025-11199-2. [PMID: 40301135 DOI: 10.1007/s11030-025-11199-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025]
Abstract
The increasing adoption of computational methods and artificial intelligence in scientific research has led to a growing interest in versatile tools like Python. In the fields of medical chemistry, biochemistry, and bioinformatics, Python has emerged as a key language for tackling complex challenges. It is used to solve various tasks, such as drug discovery, high-throughput and virtual screening, protein and genome analysis, and predicting drug efficacy. This review presents a list of tools for these tasks, including scripts, libraries, and ready-made programs, and serves as a starting point for scientists wishing to apply automation or optimization to routine tasks in medical chemistry and bioinformatics.
Collapse
Affiliation(s)
- Fedor V Ryzhkov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia.
| | - Yuliya E Ryzhkova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia
| | - Michail N Elinson
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Prospekt, 119991, Moscow, Russia
| |
Collapse
|
27
|
Yang G, Du X, Jiang X, Wang J, Shi S, Zhong VW. Biological age acceleration and interaction with genetic predisposition in the risk of type 2 diabetes and coronary artery disease. GeroScience 2025:10.1007/s11357-025-01671-0. [PMID: 40299261 DOI: 10.1007/s11357-025-01671-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Biological age (BA), compared to chronological age, offers a more accurate reflection of aging status. In this prospective UK Biobank study, BA acceleration was measured using the Klemera-Doubal method BA (KDM-BA) and Phenotypic age (PhenoAge). Cox models estimated associations of BA acceleration with incident T2D (n = 271,885) and CAD (n = 270,054). Both additive and multiplicative interactions between BA acceleration and polygenic risk score (PRS) were examined. Predictive performance was assessed by adding BA, PRS, and their interactions to traditional risk models. BA acceleration was positively associated with incident T2D (HRQ4 vs Q1 for KDM-BA: 2.38 [95% CI, 2.22-2.56]; HRQ4 vs Q1 for PhenoAge: 1.85 [95% CI, 1.72-1.99]) and CAD (HRQ4 vs Q1 for KDM-BA: 1.67 [95% CI, 1.58-1.76]; HRQ4 vs Q1 for PhenoAge: 1.33 [95% CI, 1.27-1.39]). Significant multiplicative interactions were observed between BA acceleration and PRS (all P for multiplicative interaction ≤ 0.002). Individuals with highest BA acceleration and PRS had strongest risk elevation for T2D (HR for KDM-BA, 6.89 [95% CI, 6.03-7.87]; HR for PhenoAge, 6.28 [95% CI, 5.28-7.46]) and CAD (HR for KDM-BA, 2.80 [95% CI, 2.59-3.02]; HR for PhenoAge, 2.25 [95% CI, 2.07-2.45]). Additive interactions were observed for T2D with 18-28% of risk attributable to BA-genetic interaction. Adding BA measures and PRS to traditional risk models significantly improved prediction for both diseases (Δ C-statistic 0.024-0.034). In conclusion, BA acceleration was positively associated with incident T2D and CAD, especially in individuals with high genetic predisposition, and improved T2D and CAD prediction beyond traditional risk factors.
Collapse
Affiliation(s)
- Guangrui Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building, 227 South Chongqing Rd, Shanghai, 200025, China
| | - Xihao Du
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building, 227 South Chongqing Rd, Shanghai, 200025, China
| | - Xuanwei Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building, 227 South Chongqing Rd, Shanghai, 200025, China
| | - Jingxuan Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building, 227 South Chongqing Rd, Shanghai, 200025, China
| | - Shuxiao Shi
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building, 227 South Chongqing Rd, Shanghai, 200025, China
| | - Victor W Zhong
- Department of Epidemiology and Biostatistics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 415 East No. 1 Building, 227 South Chongqing Rd, Shanghai, 200025, China.
| |
Collapse
|
28
|
Heng TH, Walter K, Huang QQ, Karjalainen J, Daly MJ, Heyne HO, Malawsky DS, Kalantzis G, Finer S, van Heel DA, Martin HC. Widespread recessive effects on common diseases in a cohort of 44,000 British Pakistanis and Bangladeshis with high autozygosity. Am J Hum Genet 2025:S0002-9297(25)00141-7. [PMID: 40306283 DOI: 10.1016/j.ajhg.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Genetic association studies have focused on testing additive models in cohorts with European ancestry. Little is known about recessive effects on common diseases, specifically for non-European ancestry. Genes & Health is a cohort of British Pakistani and Bangladeshi individuals with elevated rates of consanguinity and endogamy, making it suitable to study recessive effects. We imputed variants into a genotyped dataset (n = 44,190) by using two reference panels: a set of 4,982 whole-exome sequences from within the cohort and the Trans-Omics for Precision Medicine (TOPMed-r2) panel. We performed association testing with 898 diseases from electronic health records. 185 independent loci reached genome-wide significance (p < 5 × 10-8) under the recessive model, with p values lower than under the additive model, and >40% of these were novel. 140 loci demonstrated nominally significant (p < 0.05) dominance deviation p values, confirming a recessive association pattern. Sixteen loci in three clusters were significant at a Bonferroni threshold, accounting for multiple phenotypes tested (p < 5.4 × 10-12). In FinnGen, we replicated 44% of the expected number of Bonferroni-significant loci we were powered to replicate, at least one from each cluster, including an intronic variant in patatin-like phospholipase domain-containing protein 3 (PNPLA3; rs66812091) and non-alcoholic fatty liver disease, a previously reported additive association. We present evidence suggesting that the association is recessive instead (odds ratio [OR] = 1.3, recessive p = 2 × 10-12, additive p = 2 × 10-11, dominance deviation p = 3 × 10-2, and FinnGen recessive OR = 1.3 and p = 6 × 10-12). We identified a novel protective recessive association between a missense variant in SGLT4 (rs61746559), a sodium-glucose transporter with a possible role in the renin-angiotensin-aldosterone system, and hypertension (OR = 0.2, p = 3 × 10-8, dominance deviation p = 7 × 10-6). These results motivate interrogating recessive effects on common diseases more widely.
Collapse
Affiliation(s)
- Teng Hiang Heng
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK.
| | - Klaudia Walter
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Qin Qin Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | | | - Mark J Daly
- Broad Institute, 415 Main Street, Cambridge, MA 02142, USA
| | - Henrike O Heyne
- Broad Institute, 415 Main Street, Cambridge, MA 02142, USA; Hasso Plattner Institute, 14482 Potsdam, Germany
| | - Daniel S Malawsky
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | | | - Sarah Finer
- Wolfson Institute for Population Health, Queen Mary University of London, London E1 4NS, UK
| | - David A van Heel
- Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Hilary C Martin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK.
| |
Collapse
|
29
|
Eichemberger Rius F, Santa Cruz Guindalini R, Viana D, Salomão J, Gallo L, Freitas R, Bertolacini C, Taniguti L, Imparato D, Antunes F, Sousa G, Achjian R, Fukuyama E, Gregório C, Ventura I, Gomes J, Taniguti N, Maistro S, Krieger JE, Zheng Y, Huo D, Olopade OI, Azevedo Koike Folgueira MA, Schlesinger D. A Breast Cancer Polygenic Risk Score Validation in 15,490 Brazilians Using Exome Sequencing. Diagnostics (Basel) 2025; 15:1098. [PMID: 40361916 PMCID: PMC12071591 DOI: 10.3390/diagnostics15091098] [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: 02/25/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Brazil has a highly admixed population. Polygenic risk scores (PRSs) have mostly been developed from European population studies, and their application to other populations is challenging. To assess the use of PRS for breast cancer (BC) risk in Brazil, we evaluated four PRSs in the Brazilian population. Methods: We analyzed a Brazilian cohort composed of 6206 women with a history of breast cancer and 8878 unphenotyped adults as controls. Genomic variants were imputed from exomes, and scores were calculated for all samples. Results: After individuals with known pathogenic or likely pathogenic variants in BRCA1, BRCA2, PALB2, PTEN, or TP53 genes, and first-degree relatives of the probands were excluded, 5598 cases and 8767 controls remained. Four PRS models were compared, and PRS3820 achieved the best performance, with an odds ratio (OR) of 1.43 per standard deviation increase (p value < 0.001) and an OR of 1.88 (p value < 0.001) for the top decile. PRS3820 also performed well for different ancestry groups: East Asian majority (OR 1.59, p value 0.004), Non-European majority (OR 1.45, p value < 0.001), and European majority (OR 1.43, p value < 0.001). Conclusions: Among the different PRSs, PRS313 and PRS3820 could be validated in our Brazilian cohort, with the latter exhibiting the best performance. While further clinical studies are necessary to guide clinical practice, this work represents an important step toward improving BC precision medicine in Brazil.
Collapse
Affiliation(s)
- Flávia Eichemberger Rius
- Mendelics, Sao Paulo 02511-000, SP, Brazil
- Comprehensive Center for Precision Oncology (C2PO), Centro de Investigação Translacional em Oncologia (CTO), Departamento de Radiologia e Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-000, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Simone Maistro
- Comprehensive Center for Precision Oncology (C2PO), Centro de Investigação Translacional em Oncologia (CTO), Departamento de Radiologia e Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-000, SP, Brazil
| | - José Eduardo Krieger
- Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo 05403-900, SP, Brazil
| | - Yonglan Zheng
- Medicine and Human Genetics, Center for Clinical Cancer Genetics and Global Health, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Olufunmilayo I. Olopade
- Medicine and Human Genetics, Center for Clinical Cancer Genetics and Global Health, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Maria Aparecida Azevedo Koike Folgueira
- Comprehensive Center for Precision Oncology (C2PO), Centro de Investigação Translacional em Oncologia (CTO), Departamento de Radiologia e Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-000, SP, Brazil
| | | |
Collapse
|
30
|
Lou J, Xiang Z, Zhu X, Fan Y, Li J, Jin G, Cui S, Huang N. A bidirectional mendelian-randomization analyses of genetically predicted circulating levels of systemic inflammatory regulators with risk of sepsis. Medicine (Baltimore) 2025; 104:e42199. [PMID: 40295284 PMCID: PMC12040038 DOI: 10.1097/md.0000000000042199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Whether there is a causal relationship between circulating levels of systemic inflammatory regulators and sepsis remains unclear. To determine whether genetically predicted circulating levels of cytokines are associated with risk of sepsis, a bidirectional two-sample Mendelian randomization (MR) analysis based on the a STROBE-compliant cross-sectional observational study was conducted utilizing gene-wide association study (GWAS) data. Selected with rigor, single-nucleotide polymorphisms served as instrumental variables for subsequent MR analysis. The preferred method for the MR analysis was the inverse-variance weighted approach. However, for comprehensive sensitivity analyses, 6 additional MR methods were employed. Cochrane's Q test was performed to examine heterogeneity. A leave-one-out method ensured the stability of MR results. Our findings suggest an inverse association between the levels of beta-nerve growth factor (BNGF) and the risk of sepsis development (OR = 0.769, 95% CI = 0.599-0.987, P = .039). In contrast, higher levels of TNF-related apoptosis-inducing ligand and vascular endothelial growth factor A (VEGF-A) are positively correlated with sepsis risk (OR = 1.094, 95% CI = 1.012-1.183, P = .025; OR = 1.182, 95% CI = 1.016-1.375, P = .031, respectively). Reverse MR Analysis indicated that sepsis risk is linked with lower circulating levels of adenosine deaminase and Interleukin-17A (β = -0.043, 95% CI = -0.085 to -0.002, P = .042; β = -0.061, 95% CI = -0.108 to -0.013, P = .012, respectively), and also with higher circulating levels of BNGF, delta/notchlike epidermal growth factor-related receptor, fibroblast growth factor 23, leukemia inhibitory factor, monocyte chemoattractant protein-1, and osteoprotegerin (β = 0.056, 95% CI = 0.015-0.096, P = .007; β = 0.137, 95% CI = 0.035-0.240, P = .009; β = 0.118, 95% CI = 0.020-0.216, P = .018; β = 0.136, 95% CI = 0.020-0.252, P = .022; β = 0.143, 95% CI = 0.043-0.242, P = .005; β = 0.116, 95% CI = 0.010-0.222, P = .031, respectively). Sum up, our study provides evidence supporting a bidirectional causal relationship between sepsis and genetically predicted circulating levels of systemic inflammatory regulators.
Collapse
Affiliation(s)
- Jiaqi Lou
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Ziyi Xiang
- Institute of Pathology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Xiaoyu Zhu
- Health Science Center, Ningbo University, Ningbo, China
| | - Youfen Fan
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Jiliang Li
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Guoying Jin
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Shengyong Cui
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Neng Huang
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| |
Collapse
|
31
|
Zhou M, Ling C, Xiao H, Zhang Z. Identification of Gene Expression and Splicing QTLs in Porcine Muscle Associated with Meat Quality Traits. Animals (Basel) 2025; 15:1209. [PMID: 40362025 PMCID: PMC12071002 DOI: 10.3390/ani15091209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/10/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Understanding the genetic regulation of gene expression and splicing in muscle tissues is critical for elucidating the molecular mechanisms of meat quality traits. In this study, we integrated large-scale whole-genome sequencing and strand-specific RNA-seq data from 582 F2 hybrid pigs (White Duroc × Erhualian) to characterize the expression and splicing quantitative trait loci (eQTLs/sQTL) in longissimus dorsi muscle. We identified 11,058 cis-eQTL-associated genes (eGenes) and 5139 cis-sQTL-associated genes (sGenes), of which 29% of eGenes and 80% of sGenes were previously unreported in the PigGTEx database. Functional analyses revealed distinct genomic features: eQTLs were enriched near transcription start sites (TSSs) and associated with active TSS-proximal transcribed regions and enhancers, whereas sQTLs clustered at splice junctions, underscoring their distinct roles in gene expression and splicing regulation. Colocalization analysis of e/sQTLs with GWAS signals prioritized PHKG1 as a key candidate gene (PPH4 > 0.9) for glycogen metabolism. Notably, we confirmed that an sQTL-driven alternative splicing event in exon 10 of PHKG1 was significantly correlated with phenotypic variation (R = -0.39, p = 9.5 × 10-21). Collectively, this study provides novel insights into the genetic regulation of gene expression and alternative splicing in porcine muscle tissue, advancing our understanding of the molecular mechanisms underlying economically important meat quality traits.
Collapse
Affiliation(s)
- Meng Zhou
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation Technology, Jiangxi Agricultural University, Nanchang 330045, China; (C.L.); (H.X.); (Z.Z.)
| | | | | | | |
Collapse
|
32
|
Stanley J, Rabot E, Reddy S, Belilovsky E, Mottron L, Bzdok D. Large language models deconstruct the clinical intuition behind diagnosing autism. Cell 2025; 188:2235-2248.e10. [PMID: 40147442 DOI: 10.1016/j.cell.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/28/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025]
Abstract
Efforts to use genome-wide assays or brain scans to diagnose autism have seen diminishing returns. Yet the clinical intuition of healthcare professionals, based on longstanding first-hand experience, remains the gold standard for diagnosis of autism. We leveraged deep learning to deconstruct and interrogate the logic of expert clinician intuition from clinical reports to inform our understanding of autism. After pre-training on hundreds of millions of general sentences, we finessed large language models (LLMs) on >4,000 free-form health records from healthcare professionals to distinguish confirmed versus suspected autism cases. By introducing an explainability strategy, our extended language model architecture could pin down the most salient single sentences in what drives clinical thinking toward correct diagnoses. Our framework flagged the most autism-critical DSM-5 criteria to be stereotyped repetitive behaviors, special interests, and perception-based behaviors, which challenges today's focus on deficits in social interplay, suggesting necessary revision of long-trusted diagnostic criteria in gold-standard instruments.
Collapse
Affiliation(s)
- Jack Stanley
- Mila - Québec Artificial Intelligence Institute, Montréal, QC H2S3H1, Canada; The Neuro - Montréal Neurological Institute (MNI), McConnell Brain Imaging Centre, Department of Biomedical Engineering, Faculty of Medicine, School of Computer Science, McGill University, Montréal, QC H3A2B4, Canada
| | - Emmett Rabot
- Research Center, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Ile-de-Montréal (CIUSSS-NIM), Montréal, QC H4K1B3, Canada; Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Siva Reddy
- Mila - Québec Artificial Intelligence Institute, Montréal, QC H2S3H1, Canada
| | - Eugene Belilovsky
- Mila - Québec Artificial Intelligence Institute, Montréal, QC H2S3H1, Canada; Department of Computer Science and Software Engineering, Concordia University, Montreal, QC H3G 1M8, Canada
| | - Laurent Mottron
- Research Center, Centre Intégré Universitaire de Santé et de Services Sociaux du Nord-de-l'Ile-de-Montréal (CIUSSS-NIM), Montréal, QC H4K1B3, Canada; Université de Montréal, Montréal, QC H3C3J7, Canada
| | - Danilo Bzdok
- Mila - Québec Artificial Intelligence Institute, Montréal, QC H2S3H1, Canada; The Neuro - Montréal Neurological Institute (MNI), McConnell Brain Imaging Centre, Department of Biomedical Engineering, Faculty of Medicine, School of Computer Science, McGill University, Montréal, QC H3A2B4, Canada.
| |
Collapse
|
33
|
Ben-Ari Y, Danchin ÉÉ. Limitations of genomics to predict and treat autism: a disorder born in the womb. J Med Genet 2025; 62:303-310. [PMID: 40081874 PMCID: PMC12015019 DOI: 10.1136/jmg-2024-110224] [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: 07/24/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Brain development involves the sequential expression of vulnerable biological processes including cell proliferation, programmed cell death, neuronal migration, synapse and functional unit formation. All these processes involve gene and activity-dependent events that can be distorted by many extrinsic and intrinsic environmental factors, including stress, microbiota, inflammatory signals, hormonal signals and epigenetic factors, hence leading to disorders born in the womb that are manifested later in autism spectrum disorders (ASDs) and other neurodevelopmental disorders. Predicting and treating such disorders call for a conceptual framework that includes all aspects of developmental biology. Here, taking the high incidence of ASDs as an example, we first discuss the intrinsic limitations of the genetic approach, notably the widely used twin studies and SNPs. We then review the long list of in utero events that can deviate developmental sequences, leading to persistent aberrant activity generated by immature misplaced and misconnected neuronal ensembles that are the direct cause of ASD. In a clinical perspective, we suggest analysing non-genetic maternity data to enable an early prediction of babies who will develop ASD years later, thereby facilitating early psycho-educative techniques. Subsequently, agents capable of selectively silencing malformed immature networks offer promising therapeutic perspectives. In summary, understanding developmental processes is critical to predicting, understanding and treating ASD, as well as most other disorders that arise in the womb.
Collapse
Affiliation(s)
| | - Étienne É Danchin
- Centre de biologie integrative, Centre de recherches sur la cognition animale, Toulouse University, Toulouse, France
| |
Collapse
|
34
|
Hamazaki K, Iwata H, Mary-Huard T. A novel genome-wide association study method for detecting quantitative trait loci interacting with complex population structures in plant genetics. Genetics 2025; 229:iyaf038. [PMID: 40091626 DOI: 10.1093/genetics/iyaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
In plant genetics, most modern association analyses are performed on panels that bring together individuals from several populations, including admixed individuals whose genomes comprise chromosomal regions from different populations. These panels can identify quantitative trait loci (QTLs) with population-specific effects and epistatic interactions between QTLs and polygenic backgrounds. However, analyzing a diverse panel constitutes a challenge for statistical analysis. The statistical model must account for possible interactions between a QTL and the panel structure while strictly controlling the detection error rate. Although models to detect population-specific QTLs have already been developed, they rely on prior information about the population structure. In practice, this prior information may be missing as many genome-wide association study (GWAS) panels exhibit complex population structures. The present study introduces 2 new models for detecting QTLs interacting with complex population structures. Both incorporate an interaction term between single nucleotide polymorphism/haplotype block and genetic background into conventional GWAS models. The proposed models were compared with state-of-the-art models through simulation studies that considered QTLs with different levels of interaction with their genetic backgrounds. Results showed that models matching simulation settings were most effective for detecting corresponding QTLs while the proposed models outperformed classical models in detecting QTLs interacting with polygenes. Additionally, when applied to a soybean dataset, one of our models identified putative associated QTLs that conventional models failed to detect. The new models, implemented in the RAINBOWR package available on CRAN, are expected to help uncover complex trait genetic architectures.
Collapse
Affiliation(s)
- Kosuke Hamazaki
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroyoshi Iwata
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Tristan Mary-Huard
- MIA-Paris Saclay, INRAE, AgroParisTech, Université Paris-Saclay, Palaiseau 91120, France
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, Génétique Quantitative et Evolution-Le Moulon, Gif-sur-Yvette 91190, France
| |
Collapse
|
35
|
Kinstler E, Gorelik AJ, Paul SE, Aggarwal A, Johnson EC, Cyders MA, Agrawal A, Bogdan R, Miller AP. Genetic influences for distinct impulsivity domains are differentially associated with early substance use initiation: Results from the ABCD Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.14.25325687. [PMID: 40321268 PMCID: PMC12047936 DOI: 10.1101/2025.04.14.25325687] [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: 05/11/2025]
Abstract
Background Impulsivity is among the strongest correlates of substance involvement and its distinct domains (e.g., sensation seeking, urgency) are differentially correlated, phenotypically and genetically, with unique substance involvement stages. Understanding whether polygenic influences for distinct impulsivity domains are differentially predictive of early substance use initiation, a major risk factor for later problematic use, will improve our understanding of the role of impulsivity in addiction etiology. Methods Data collected from participants (n=4,808) of genetically-inferred European ancestry enrolled in the Adolescent Brain Cognitive Development StudySM were used to estimate associations between polygenic scores (PGS) for UPPS-P impulsivity domains (i.e., sensation seeking, lack of premeditation/perseverance, and negative/positive urgency) and substance (i.e., any, alcohol, nicotine, cannabis) use initiation before age 15. Mediation models examined whether child impulsivity (ages 9-11) mediated links between PGSs and substance use initiation. Results Sensation seeking PGS was significantly associated with any substance and alcohol use initiation (ORs>1.10, psFDR<0.006). Lack of perseverance and urgency (negative/positive) PGSs were nominally associated with alcohol and nicotine use initiation, respectively (ORs>1.06, ps<0.05, psFDR>0.05). No significant associations were observed for lack of premeditation PGS or cannabis use initiation. Measured impulsivity domains accounted for 5-9% of associations between UPPS-P PGSs and substance use initiation. Conclusions Genetic influences for distinct impulsivity domains have differential associations with early substance use initiation with sensation seeking showing the most robust associations. Evaluating genetic influences for distinct impulsivity domains can yield valuable etiologic insight into the earliest stages of substance involvement that may be missed when adopting broad impulsivity definitions.
Collapse
Affiliation(s)
- Ethan Kinstler
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, St. Louis, MO
| | - Aaron J. Gorelik
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, St. Louis, MO
| | - Sarah E. Paul
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, St. Louis, MO
| | - Adamya Aggarwal
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, St. Louis, MO
| | - Emma C. Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | - Melissa A. Cyders
- Department of Psychology, Indiana University Indianapolis, Indianapolis, IN
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, St. Louis, MO
| | - Alex P. Miller
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
36
|
van Alten S, Domingue BW, Faul J, Galama T, Marees AT. Correcting for volunteer bias in GWAS increases SNP effect sizes and heritability estimates. Nat Commun 2025; 16:3578. [PMID: 40234401 PMCID: PMC12000612 DOI: 10.1038/s41467-025-58684-8] [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: 02/13/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
Selection bias in genome-wide association studies (GWASs) due to volunteer-based sampling (volunteer bias) is poorly understood. The UK Biobank (UKB), one of the largest and most widely used cohorts, is highly selected. Using inverse probability (IP) weights we estimate inverse probability weighted GWAS (WGWAS) to correct GWAS summary statistics in the UKB for volunteer bias. Our IP weights were estimated using UK Census data - the largest source of population-representative data - made representative of the UKB's sampling population. These weights have a substantial SNP-based heritability of 4.8% (s.e. 0.8%), suggesting they capture volunteer bias in GWAS. Across ten phenotypes, WGWAS yields larger SNP effect sizes, larger heritability estimates, and altered gene-set tissue expression, despite decreasing the effective sample size by 62% on average, compared to GWAS. The impact of volunteer bias on GWAS results varies by phenotype. Traits related to disease, health behaviors, and socioeconomic status are most affected. We recommend that GWAS consortia provide population weights for their data sets, or use population-representative samples.
Collapse
Affiliation(s)
- Sjoerd van Alten
- Vrije Universiteit Amsterdam, Amsterdam, Netherlands.
- Tinbergen Institute, Amsterdam, Netherlands.
| | | | | | - Titus Galama
- Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Tinbergen Institute, Amsterdam, Netherlands
- University of Southern California, Dornsife Center for Economic and Social Research and Department of Economics, California, US
| | | |
Collapse
|
37
|
Lee I, Wallace ZS, Wang Y, Park S, Nam H, Majithia AR, Ideker T. A genotype-phenotype transformer to assess and explain polygenic risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.23.619940. [PMID: 40291728 PMCID: PMC12026415 DOI: 10.1101/2024.10.23.619940] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Genome-wide association studies have linked millions of genetic variants to biomedical phenotypes, but their utility has been limited by lack of mechanistic understanding and widespread epistatic interactions. Recently, Transformer models have emerged as a powerful machine learning architecture with potential to address these and other challenges. Accordingly, here we introduce the Genotype-to-Phenotype Transformer (G2PT), a framework for modeling hierarchical information flow among variants, genes, multigenic systems, and phenotypes. As proof-of-concept, we use G2PT to model the genetics of TG/HDL (triglycerides to high-density lipoprotein cholesterol), an indicator of metabolic health. G2PT predicts this trait via attention to 1,395 variants underlying at least 20 systems, including immune response and cholesterol transport, with accuracy exceeding state-of-the-art. It implicates 40 epistatic interactions, including epistasis between APOA4 and CETP in phospholipid transfer, a target pathway for cholesterol modification. This work positions hierarchical graph transformers as a next-generation approach to polygenic risk.
Collapse
|
38
|
Dudek MF, Wenz BM, Brown CD, Voight BF, Almasy L, Grant SFA. Characterization of non-coding variants associated with transcription-factor binding through ATAC-seq-defined footprint QTLs in liver. Am J Hum Genet 2025:S0002-9297(25)00140-5. [PMID: 40250421 DOI: 10.1016/j.ajhg.2025.03.019] [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: 09/24/2024] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/20/2025] Open
Abstract
Non-coding variants discovered by genome-wide association studies (GWASs) are enriched in regulatory elements harboring transcription factor (TF) binding motifs, strongly suggesting a connection between disease association and the disruption of cis-regulatory sequences. Occupancy of a TF inside a region of open chromatin can be detected in ATAC-seq where bound TFs block the transposase Tn5, leaving a pattern of relatively depleted Tn5 insertions known as a "footprint." Here, we sought to identify variants associated with TF binding, or "footprint quantitative trait loci" (fpQTLs), in ATAC-seq data generated from 170 human liver samples. We used computational tools to scan the ATAC-seq reads to quantify TF binding likelihood as "footprint scores" at variants derived from whole-genome sequencing generated in the same samples. We tested for association between genotype and footprint score and observed 809 fpQTLs associated with footprint-inferred TF binding (FDR < 5%). Given that Tn5 insertion sites are measured with base-pair resolution, we show that fpQTLs can aid GWAS and QTL fine-mapping by precisely pinpointing TF activity within broad trait-associated loci where the underlying causal variant is unknown. Liver fpQTLs were strongly enriched across ChIP-seq peaks, liver expression QTLs (eQTLs), and liver-related GWAS loci, and their inferred effect on TF binding was concordant with their effect on underlying sequence motifs in 78% of cases. We conclude that fpQTLs can reveal causal GWAS variants, define the role of TF binding-site disruption in complex traits, and provide functional insights into non-coding variants, ultimately informing novel treatments for common diseases.
Collapse
Affiliation(s)
- Max F Dudek
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon M Wenz
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher D Brown
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin F Voight
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 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, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura Almasy
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Lifespan Brain Institute, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biomedical and Health Informatics, 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 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
39
|
Wang L, Markus H, Chen D, Chen S, Zhang F, Gao S, Khunsriraksakul C, Chen F, Olsen N, Foulke G, Jiang B, Carrel L, Liu DJ. An atlas of single-cell eQTLs dissects autoimmune disease genes and identifies novel drug classes for treatment. CELL GENOMICS 2025; 5:100820. [PMID: 40154479 PMCID: PMC12008810 DOI: 10.1016/j.xgen.2025.100820] [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: 02/27/2024] [Revised: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Most variants identified from genome-wide association studies (GWASs) are non-coding and regulate gene expression. However, many risk loci fail to colocalize with expression quantitative trait loci (eQTLs), potentially due to limited GWAS and eQTL analysis power or cellular heterogeneity. Population-scale single-cell RNA-sequencing (scRNA-seq) datasets are emerging, enabling mapping of eQTLs in different cell types (sc-eQTLs). Compared to eQTL data from bulk tissues (bk-eQTLs), sc-eQTL datasets are smaller. We propose a joint model of bk-eQTLs as a weighted sum of sc-eQTLs (JOBS) from constituent cell types to improve power. Applying JOBS to One1K1K and eQTLGen data, we identify 586% more eQTLs, matching the power of 4× the sample sizes of OneK1K. Integrating sc-eQTLs with GWAS data creates an atlas for 14 immune-mediated disorders, colocalizing 29.9% or 32.2% more loci than using sc-eQTL or bk-eQTL alone. Extending JOBS, we develop a drug-repurposing pipeline and identify novel drugs validated by real-world data.
Collapse
Affiliation(s)
- Lida Wang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Havell Markus
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Dieyi Chen
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Siyuan Chen
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Fan Zhang
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Shuang Gao
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Chachrit Khunsriraksakul
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Fang Chen
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nancy Olsen
- Department of Medicine, Penn State University, College of Medicine, Hershey, PA 17033, USA
| | - Galen Foulke
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Dermatology, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Bibo Jiang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Laura Carrel
- Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Dajiang J Liu
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Bioinformatics and Genomics PhD Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
40
|
Shao Z, Tang W, Wu H, Kong Y, Hao X. Incorporating multiple functional annotations to improve polygenic risk prediction accuracy. CELL GENOMICS 2025:100850. [PMID: 40239655 DOI: 10.1016/j.xgen.2025.100850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/21/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025]
Abstract
We present OmniPRS, a scalable biobank-scale framework that improves genetic risk prediction for complex traits by integrating genome-wide association study (GWAS) summary statistics and functional annotations. It employs a mixed model incorporating tissue-specific genetic variance components from annotations to re-estimate single-nucleotide polymorphism (SNP) effects and constructs tissue-specific polygenic risk scores (PRSs) and aggregates them into the final OmniPRS. Our experiments, encompassing 135 simulation scenarios and 11 representative traits, demonstrate that OmniPRS is flexible and robust, delivering efficient and accurate predictions comparable to ten leading PRS methods. For quantitative (binary) traits, OmniPRS achieved an average improvement of 52.31% (19.83%) versus the clumping and thresholding (C+T) method, 3.92% (1.31%) versus the annotation-integrated PRSs (LDpred-funct), and 8.44% (2.27%) versus the Bayesian-based PRSs (PRScs). Notably, it achieved 35× faster computation than the PRScs. This rapid, precise framework enables efficient polygenic risk scoring with multi-annotation integration for large-scale genomic studies.
Collapse
Affiliation(s)
- Zhonghe Shao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wangxia Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hongji Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yifan Kong
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
41
|
Huang M, Zhang W, Dong J, Hu Z, Tan X, Li H, Sun K, Zhao A, Huang T. Genome-Wide Association Studies of Body Weight and Average Daily Gain in Chinese Dongliao Black Pigs. Int J Mol Sci 2025; 26:3453. [PMID: 40244387 PMCID: PMC11989284 DOI: 10.3390/ijms26073453] [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: 03/04/2025] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/18/2025] Open
Abstract
In the domain of swine production, body weight (BW) and average daily gain (ADG) are recognized as the primary performance indicators. Nevertheless, the genetic architecture of ADG and BW in Dongliao black (DLB) pigs remains to be fully elucidated. In this study, we performed a genome-wide association analysis of BW, ADG, and body mass index (BMI) in 358 DLB pigs of different days of age. The genome-wide association study (GWAS) showed the following: (1) The most significant single nucleotide polymorphism (SNP) detected for BW was on Sus scrofa chromosome (SSC) 11:100,808 (p-value = 1.16 × 10-6) that was also the most significant SNP for ADG. (2) The most significant SNP associated with BMI was SSC17:51,463,521 (p-value = 5.16 × 10-8). (3) SNPs SSC10:6,523,844 and SSC17:23,852,682 were identified in both BW and ADG. A meta-analysis was conducted on BW at different days and demonstrated SSC5:39,028,335 (p-value = 8.37 × 10-6) which was not identified in the results of each single trait. The regions of two SNPs (SSC11:100,808, SSC4:10,703,277) exhibited considerable influence on both BW and ADG and the related regions were selected for linkage disequilibrium (LD) analyses that exhibited a notable linkage. In addition, several genes were identified that are associated with obesity and play roles in lipid metabolism, including MACROD2, PHLPP2, CYP2E1, and STT3B.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ayong Zhao
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China; (M.H.); (W.Z.); (J.D.); (Z.H.); (X.T.); (H.L.); (K.S.)
| | - Tao Huang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China; (M.H.); (W.Z.); (J.D.); (Z.H.); (X.T.); (H.L.); (K.S.)
| |
Collapse
|
42
|
Tian C. No Associations Between Genetically Predicted Chronotype, Insomnia, Daytime Sleepiness, or Physical Activity and Acne Vulgaris: A Two-Sample Mendelian Randomization Study. Clin Cosmet Investig Dermatol 2025; 18:827-835. [PMID: 40225312 PMCID: PMC11986648 DOI: 10.2147/ccid.s510739] [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: 01/07/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025]
Abstract
Purpose The exact factors leading to the development of acne vulgaris are poorly understood. Besides diet, lifestyle habits like sleep and physical activity have received attention. This study explored the causal associations between genetically predicted sleep traits and exercise and acne vulgaris. Patients and Methods The genome-wide association study (GWAS) data for sleep, physical activity, and acne vulgaris were retrieved from the FinnGen Project (1092/211,139 patients/controls) to carry out a two-sample Mendelian randomization (MR) analysis. Validation was performed using a dataset from the Estonian Biobank (34,422/364,991 patients/controls). The inverse variance weighted (IVW) method was the primary analytical method, with robustness tested using the weighted median, weighted mode, and MR-Egger analyses. Heterogeneity was tested using Cochran's Q-test, horizontal pleiotropy using MR-Egger regression, outliers using MR-PRESSO, and driving SNPs using the leave-one-out method. Results The results revealed that genetically predicted chronotype (OR=1.021, 95% CI: 0.786-1.326, P=0.875), insomnia (OR=1.475, 95% CI: 0.676-3.216, P=0.329), daytime sleepiness (OR=0.466, 95% CI: 0.046-4.708, P=0.518), or physical activity (OR=0.990, 95% CI: 0.925-1.059, P=0.767) were not causally associated with acne vulgaris. Cochran's Q-test detected no heterogeneity (all P>0.05). No horizontal pleiotropy was detected (all P>0.05), indicating that the selected IVs met the third MR assumption. MR-PRESSO revealed no outliers. No single SNP drove the results according to the leave-one-out analysis. These results were validated through the use of additional datasets. Conclusion Our study found no causal associations between sleep traits and physical activity and acne vulgaris. However, further research is needed to explore other potential factors and validate these results in more diverse populations.
Collapse
Affiliation(s)
- Chaoqun Tian
- Department of Dermatology, Chongqing Yubei District People`s Hospital, Chongqing, 401120, People’s Republic of China
| |
Collapse
|
43
|
Khalil M, Kalyoncu A, Bellon A. Genetics of Suicide. Genes (Basel) 2025; 16:428. [PMID: 40282388 PMCID: PMC12027201 DOI: 10.3390/genes16040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Over the past two decades, suicide has consistently ranked among the leading causes of death in the United States. While suicide deaths are closely associated with uicidal ideation and attempts, these are not good predictors of future suicide deaths. Establishing who is at risk of suicide remains a challenge that is mostly hampered by the lack of understanding of its pathophysiology. Nonetheless, evidence continues to accumulate suggesting that suicide is driven by a complex and dynamic interaction between environmental factors and genetics. The identification of genes that place people at risk of suicide remains elusive, but data are rapidly evolving. In this narrative review, we describe how Tryptophan hydroxylase (TPH) genes, particularly TPH1 and TPH2, have been associated with suicide in various publications. There is also replicated evidence linking the brain-derived neurotrophic factor gene to suicide, with its most consistent results originating from epigenetic studies. Not surprisingly, many genes involved in the hypothalamic-pituitary-adrenal axis have been connected with suicide, but these data require replication. Finally, among the inflammatory genes studied in suicide, only specific polymorphisms in TNF-alpha and IL-6 may increase susceptibility to suicidal behavior. In conclusion, significant work remains to be performed as inconsistencies undermine the reliability of genetic results in suicide. Potential avenues for future research are proposed.
Collapse
Affiliation(s)
- Mostafa Khalil
- Brown University, Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Providence, RI 02912, USA;
| | - Anil Kalyoncu
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA 17033, USA;
| | - Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA 17033, USA;
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA 17033, USA
| |
Collapse
|
44
|
Sun Q, Horimoto ARVR, Chen B, Ockerman F, Mohlke KL, Blue E, Raffield LM, Li Y. Opportunities and challenges of local ancestry in genetic association analyses. Am J Hum Genet 2025; 112:727-740. [PMID: 40185073 DOI: 10.1016/j.ajhg.2025.03.004] [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: 11/24/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/07/2025] Open
Abstract
Recently, admixed populations make up an increasing percentage of the US and global populations, and the admixture is not uniform over space or time or across genomes. Therefore, it becomes indispensable to evaluate local ancestry in addition to global ancestry to improve genetic epidemiological studies. Recent advances in representing human genome diversity, coupled with large-scale whole-genome sequencing initiatives and improved tools for local ancestry inference, have enabled studies to demonstrate that incorporating local ancestry information enhances both genetic association analyses and polygenic risk predictions. Along with the opportunities that local ancestry provides, there exist challenges preventing its full usage in genetic analyses. In this review, we first summarize methods for local ancestry inference and illustrate how local ancestry can be utilized in various analyses, including admixture mapping, association testing, and polygenic risk score construction. In addition, we discuss current challenges in research involving local ancestry, both in terms of the inference itself and its role in genetic association studies. We further pinpoint some future study directions and methodology development opportunities to help more effectively incorporate local ancestry in genetic analyses. It is worth the effort to pursue those future directions and address these analytical challenges because the appropriate use of local ancestry estimates could help mitigate inequality in genomic medicine and improve our understanding of health and disease outcomes.
Collapse
Affiliation(s)
- Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Andrea R V R Horimoto
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank Ockerman
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth Blue
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA; Brotman Baty Institute, Seattle, WA 98195, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
45
|
Long E, Williams J, Zhang H, Choi J. An evolving understanding of multiple causal variants underlying genetic association signals. Am J Hum Genet 2025; 112:741-750. [PMID: 39965570 DOI: 10.1016/j.ajhg.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
Understanding how genetic variation contributes to phenotypic variation is a fundamental question in genetics. Genome-wide association studies (GWASs) have discovered numerous genetic associations with various human phenotypes, most of which contain co-inherited variants in strong linkage disequilibrium (LD) with indistinguishable statistical significance. The experimental and analytical difficulty in identifying the "causal variant" among the co-inherited variants has traditionally led mechanistic studies to focus on relatively simple loci, where a single functional variant is presumed to explain most of the association signal and affect a target gene. The notion that a single causal variant is responsible for an association signal, while other variants in LD are merely correlated, has often been assumed in functional studies. However, emerging evidence powered by high-throughput experimental tools and context-specific functional databases argues that even a single independent signal may involve multiple functional variants in strong LD, each contributing to the observed genetic association. In this perspective, we articulate this evolving understanding of causal variants through examples from both traditional locus-by-locus approaches and more recent high-throughput functional studies. We then discuss the implications and prospects of this notion in understanding the genetic architecture of complex traits and interpreting the variant-level causality in GWAS follow-up studies.
Collapse
Affiliation(s)
- Erping Long
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jacob Williams
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
46
|
Liu Y, Li C, Cui X, Li M, Liu S, Wang Z. Potentially diagnostic and prognostic roles of piRNAs/PIWIs in pancreatic cancer: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189286. [PMID: 39952623 DOI: 10.1016/j.bbcan.2025.189286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with limited early diagnostic methods and therapeutic options, contributing to its poor prognosis. Recent advances in high-throughput sequencing have highlighted the critical roles of noncoding RNAs (ncRNAs), particularly PIWI-interacting RNAs (piRNAs), in cancer biology. In this review, we systematically summarize the emerging roles of piRNAs and their associated PIWI proteins in PDAC pathogenesis, progression, and prognosis. We provide a comprehensive analysis of the molecular mechanisms by which piRNAs/PIWIs regulate gene expression and cellular signaling pathways in PDAC. Furthermore, we discuss their potential as novel biomarkers for early diagnosis and therapeutic targets. Importantly, this review identifies key piRNAs/PIWIs involved in PDAC and proposes innovative strategies for improving diagnosis and treatment outcomes. Our work not only consolidates current knowledge but also offers new perspectives for future research and clinical applications in PDAC management.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changlei Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miaomiao Li
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Shiguo Liu
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China.
| | - Zusen Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
47
|
Choi J, Tang Z, Dong W, Ulibarri J, Mehinovic E, Thomas S, Höke A, Jin SC. Unleashing the Power of Multiomics: Unraveling the Molecular Landscape of Peripheral Neuropathy. Ann Clin Transl Neurol 2025; 12:674-685. [PMID: 40126913 PMCID: PMC12040521 DOI: 10.1002/acn3.70019] [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: 09/27/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 03/26/2025] Open
Abstract
Peripheral neuropathies (PNs) affect over 20 million individuals in the United States, manifesting as a wide range of sensory, motor, and autonomic nerve symptoms. While various conditions such as diabetes, metabolic disorders, trauma, autoimmune disease, and chemotherapy-induced neurotoxicity have been linked to PN, approximately one-third of PN cases remain idiopathic, underscoring a critical gap in our understanding of these disorders. Over the years, considerable efforts have focused on unraveling the complex molecular pathways underlying PN to advance diagnosis and treatment. Traditional methods such as linkage analysis, fluorescence in situ hybridization, polymerase chain reaction, and Sanger sequencing identified initial genetic variants associated with PN. However, the establishment and application of next-generation sequencing (NGS) and, more recently, long-read/single-cell sequencing have revolutionized the field, accelerating the discovery of novel disease-causing variants and challenging previous assumptions about pathogenicity. This review traces the evolution of genomic technologies in PN research, emphasizing the pivotal role of NGS in uncovering genetic complexities. We provide a comprehensive analysis of established genomic approaches such as genome-wide association studies, targeted gene panel sequencing, and whole-exome/genome sequencing, alongside emerging multiomic technologies including RNA sequencing and proteomics. Integrating these approaches promises holistic insights into PN pathophysiology, potentially revealing new biomarkers and therapeutic targets. Furthermore, we discuss the clinical implications of genomic and multiomic integration, highlighting their potential to enhance diagnostic accuracy, prognostic assessment, and personalized treatment strategies for PN. Challenges and questions in standardizing these technologies for clinical use are raised, underscoring the need for robust guidelines to maximize their clinical utility.
Collapse
Affiliation(s)
- Julie Choi
- Department of GeneticsSchool of Medicine, Washington UniversitySt. LouisMissouriUSA
| | - Zitian Tang
- Department of GeneticsSchool of Medicine, Washington UniversitySt. LouisMissouriUSA
| | - Wendy Dong
- Department of GeneticsSchool of Medicine, Washington UniversitySt. LouisMissouriUSA
| | - Jenna Ulibarri
- Department of GeneticsSchool of Medicine, Washington UniversitySt. LouisMissouriUSA
| | - Elvisa Mehinovic
- Department of GeneticsSchool of Medicine, Washington UniversitySt. LouisMissouriUSA
| | - Simone Thomas
- Department of Neurology, Neuromuscular DivisionJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ahmet Höke
- Department of Neurology, Neuromuscular DivisionJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeuroscienceJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Sheng Chih Jin
- Department of GeneticsSchool of Medicine, Washington UniversitySt. LouisMissouriUSA
- Department of PediatricsSchool of Medicine, Washington UniversitySt. LouisMissouriUSA
| |
Collapse
|
48
|
Cheng XX, Lin GW, Zhou YQ, Li YQ, He S, Liu Y, Zeng YN, Guo YM, Liu SQ, Peng W, Wei PP, Luo CL, Bei JX. A rare KLHDC4 variant Glu510Lys is associated with genetic susceptibility and promotes tumor metastasis in nasopharyngeal carcinoma. J Genet Genomics 2025; 52:559-569. [PMID: 39706520 DOI: 10.1016/j.jgg.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Various genetic association studies have identified numerous single nucleotide polymorphisms (SNPs) associated with nasopharyngeal carcinoma (NPC) risk. However, these studies have predominantly focused on common variants, leaving the contribution of rare variants to the "missing heritability" largely unexplored. Here, we integrate genotyping data from 3925 NPC cases and 15,048 healthy controls to identify a rare SNP, rs141121474, resulting in a Glu510Lys mutation in KLHDC4 gene linked to increased NPC risk. Subsequent analyses reveal that KLHDC4 is highly expressed in NPC and correlates with poorer prognosis. Functional characterizations demonstrate that KLHDC4 acts as an oncogene in NPC cells, enhancing their migratory and metastatic capabilities, with these effects being further augmented by the Glu510Lys mutation. Mechanistically, the Glu510Lys mutant exhibits increased interaction with Vimentin compared to the wild-type KLHDC4 (KLHDC4-WT), leading to elevated Vimentin protein stability and modulation of the epithelial-mesenchymal transition process, thereby promoting tumor metastasis. Moreover, Vimentin knockdown significantly mitigates the oncogenic effects induced by overexpression of both KLHDC4-WT and the Glu510Lys variant. Collectively, our findings highlight the critical role of the rare KLHDC4 variant rs141121474 in NPC progression and propose its potential as a diagnostic and therapeutic target for NPC patients.
Collapse
Affiliation(s)
- Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Guo-Wang Lin
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Yan-Ni Zeng
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yun-Miao Guo
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, Guangdong 524045, China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Wan Peng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China; Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, China; Department of Medical Oncology, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
49
|
Mohanty JK, Yadav A, Narnoliya L, Thakro V, Nayyar H, Dixit GP, Jha UC, Vara Prasad PV, Agarwal P, Parida SK. A Next-Generation Combinatorial Genomic Strategy Scans Genomic Loci Governing Heat Stress Tolerance in Chickpea. PLANT, CELL & ENVIRONMENT 2025; 48:2706-2726. [PMID: 39360859 DOI: 10.1111/pce.15186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024]
Abstract
In the wake of rising earth temperature, chickpea crop production is haunted by the productivity crisis. Chickpea, a cool season legume manifests tolerance in several agro-physiological level, which is complex quantitative in nature, and regulated by multiple genes and genetic networks. Understanding the molecular genetic basis of this tolerance and identifying key regulators can leverage chickpea breeding against heat stress. This study employed a genomics-assisted breeding strategy utilizing multi-locus GWAS to identify 10 key genomic regions linked to traits contributing to heat stress tolerance in chickpea. These loci subsequently delineated few key candidates and hub regulatory genes, such as RAD23b, CIPK25, AAE19, CK1 and WRKY40, through integrated genomics, transcriptomics and interactive analyses. The differential transcript accumulation of these identified candidates in contrasting chickpea accessions suggests their potential role in heat stress tolerance. Differential ROS accumulation along with their scavengers' transcript abundance aligning with the expression of identified candidates in the contrasting chickpea accessions persuade their regulatory significance. Additionally, their functional significance is ascertained by heterologous expression and subsequent heat stress screening. The high confidence genomic loci and the superior genes and natural alleles delineated here has great potential for swift genomic interventions to enhance heat resilience and yield stability in chickpea.
Collapse
Affiliation(s)
- Jitendra K Mohanty
- Genomics-Assisted Breeding and Crop Improvement Laboratory, National Institute of Plant Genome Research (NIPGR), New Delhi, India
| | - Antima Yadav
- Genomics-Assisted Breeding and Crop Improvement Laboratory, National Institute of Plant Genome Research (NIPGR), New Delhi, India
| | - Laxmi Narnoliya
- Genomics-Assisted Breeding and Crop Improvement Laboratory, National Institute of Plant Genome Research (NIPGR), New Delhi, India
| | - Virevol Thakro
- Genomics-Assisted Breeding and Crop Improvement Laboratory, National Institute of Plant Genome Research (NIPGR), New Delhi, India
| | - Harsh Nayyar
- Department of Botany, Panjab University, Chandigarh, India
| | - Girish P Dixit
- Indian Institute of Pulses Research (IIPR), Kanpur, Uttar Pradesh, India
| | - Uday Chand Jha
- Indian Institute of Pulses Research (IIPR), Kanpur, Uttar Pradesh, India
- Sustainable Intensification Innovation Lab, Department of Agronomy, Kansas State University, Manhattan, Kansas, USA
| | - P V Vara Prasad
- Sustainable Intensification Innovation Lab, Department of Agronomy, Kansas State University, Manhattan, Kansas, USA
| | - Pinky Agarwal
- Genomics-Assisted Breeding and Crop Improvement Laboratory, National Institute of Plant Genome Research (NIPGR), New Delhi, India
| | - Swarup K Parida
- Genomics-Assisted Breeding and Crop Improvement Laboratory, National Institute of Plant Genome Research (NIPGR), New Delhi, India
| |
Collapse
|
50
|
Li Y, Pawitan Y, Shen X. An enhanced framework for local genetic correlation analysis. Nat Genet 2025; 57:1053-1058. [PMID: 40065165 DOI: 10.1038/s41588-025-02123-3] [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: 06/12/2024] [Accepted: 02/10/2025] [Indexed: 04/12/2025]
Abstract
Genetic correlation is a key parameter in the joint genetic model of complex traits, but it is usually estimated on a global genomic scale. Understanding local genetic correlations provides more detailed insight into the shared genetic architecture of complex traits. However, a state-of-the-art tool for local genetic correlation analysis, LAVA, is prone to false inference. Here we extend the high-definition likelihood (HDL) method to a local version, HDL-L, which performs genetic correlation analysis in small, approximately independent linkage disequilibrium blocks. HDL-L allows a more granular estimation of genetic variances and covariances. Simulations show that HDL-L offers more consistent heritability estimates and more efficient genetic correlation estimates compared with LAVA. HDL-L demonstrated robust performance across a wide range of simulations conducted under varying parameter settings. In the analysis of 30 phenotypes from the UK Biobank, HDL-L identified 109 significant local genetic correlations and showed a notable computational advantage. HDL-L proves to be a powerful tool for uncovering the detailed genetic landscape that underlies complex human traits, offering both accuracy and computational efficiency.
Collapse
Affiliation(s)
- Yuying Li
- Greater Bay Area Institute of Precision Medicine, State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Xia Shen
- Greater Bay Area Institute of Precision Medicine, State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, China.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|