1
|
Ling Y, Yang YX, Chen YC, Wang JH, Feng DG, Xiang SJ, Zhang X, Lyu J, Li SS. Newly identified single-nucleotide polymorphism associated with the transition from nonalcoholic fatty liver disease to liver fibrosis: results from a nested case-control study in the UK biobank. Ann Med 2025; 57:2458201. [PMID: 39898988 PMCID: PMC11792139 DOI: 10.1080/07853890.2025.2458201] [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: 02/23/2024] [Revised: 12/06/2024] [Accepted: 12/14/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Genetic factors may have a significant influence on the likelihood of liver fibrosis in individuals with nonalcoholic fatty liver disease (NAFLD). The present study was conducted to explore how single-nucleotide polymorphism (SNP) impacts the development of fibrosis in those suffering from NAFLD. MATERIALS AND METHODS Utilizing the UK Biobank dataset, we conducted a nested case-control analysis among NAFLD participants, defining the case group as those with liver fibrosis and cirrhosis during follow-up. For our in vitro investigations, we employed the LX-2 human hepatic stellate cell line. Our procedures included cultivating these cells, employing SAMM50-rs2073080 plasmid techniques to enhance the expression of recently discovered SNPs, and conducting biochemical assays. To quantify gene expression, we used real-time PCR with fluorescence detection. RESULTS The study analyzed data from 5467 participants (1094 cases and 4373 controls). Genome-wide association analysis identified nine significant loci, including the novel rs2073080 variant, strongly associated with NAFLD-associated hepatic fibrosis. In vitro TGF-β modeling revealed significant upregulation of α-SMA and COL1A1, confirming model effectiveness. Oxidative stress markers like elevated malondialdehyde (MDA) and reduced catalase (CAT) and superoxide dismutase (SOD) levels indicated liver damage in the TGF-β group. SAMM50-rs2073080 was upregulated in the NAFLD-associated fibrosis model. In vitro experiments on LX-2 cells showed that SAMM50-rs2073080 overexpression led to increased fibrosis, as indicated by higher cellular MDA levels and lower CAT and SOD levels, compared to the vector group. CONCLUSION Our research highlights a significant association of SAMM50-rs2073080 with the progression of NAFLD to hepatic fibrosis, and the in vitro experiments further corroborated these findings.
Collapse
Affiliation(s)
- Yitong Ling
- Department of Neurology, Jinan University First Affiliated Hospital, Guangzhou, China
| | - Yu Xuan Yang
- Department of Pharmacy, Jinan University First Affiliated Hospital, Guangzhou, China
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Yan Chun Chen
- Department of Pharmacy, Jinan University First Affiliated Hospital, Guangzhou, China
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Jing Hao Wang
- Department of Pharmacy, Jinan University First Affiliated Hospital, Guangzhou, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, the First Affiliated Hospital, Jinan University, Guangzhou China
| | - Dong Ge Feng
- Department of Pharmacy, Jinan University First Affiliated Hospital, Guangzhou, China
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Shi Jian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaoyu Zhang
- Department of Rheumatology, Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Lyu
- Department of Clinical Research, Jinan University First Affiliated Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, China
| | - Sha Sha Li
- Department of Pharmacy, Jinan University First Affiliated Hospital, Guangzhou, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, the First Affiliated Hospital, Jinan University, Guangzhou China
| |
Collapse
|
2
|
Li J, Li J, Yu Y, Sun Y, Fu Y, Cai L, Shen W, Tan X, Wang N, Lu Y, Wang B. Data-driven discovery of midlife cardiometabolic profile associated with incident early-onset and late-onset dementia. Diabetes Obes Metab 2025; 27:2822-2832. [PMID: 40045775 DOI: 10.1111/dom.16292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Cardiometabolic risk factors have been associated with the risk of late-onset dementia. However, evidence regarding early-onset dementia was inconsistent, and the impact of clustered cardiometabolic risk factors was unclear. We aimed to investigate the associations of cardiometabolic profiles with incident early-onset and late-onset dementia. METHODS Among 289 494 UK Biobank participants, cluster analysis was built on 12 common cardiometabolic markers. Analyses were performed on those aged <65 years at baseline (n = 249 870) for early-onset dementia and those ≥65 at the end of follow-up (n = 191 213) for late-onset dementia. RESULTS During a median follow-up of 14.1 years, 279 early-onset dementia cases and 3167 late-onset dementia cases were documented. Among the five clusters of cardiometabolic profiles identified (cluster 1 [obesity-dyslipidemia pattern], cluster 2 [high blood pressure pattern], cluster 3 [high liver enzymes pattern], cluster 4 [inflammation pattern] and cluster 5 [relatively healthy pattern]), cluster 3 was significantly associated with higher risks of both early-onset and late-onset dementia; however, the risk estimate for early-onset dementia (hazard ratio 2.58, 95% CI 1.61-4.14) was larger than that for late-onset dementia (1.36, 1.09-1.71). Cluster 4 was associated with a higher risk of late-onset dementia (hazard ratio 1.39, 95% CI 1.13-1.72). No significant interactions were observed between cardiometabolic clusters and apolipoprotein E ε4 genotype. CONCLUSIONS Cardiometabolic patterns characterised by relatively high liver enzyme levels or systemic inflammation were associated with increased risks of early-onset and late-onset dementia. Identification of high-risk subgroups according to distinct cardiometabolic patterns might help develop more precise strategies for dementia prevention regardless of apolipoprotein E (APOE) ε4 status.
Collapse
Affiliation(s)
- Jiang Li
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Li
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuefeng Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Sun
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqi Fu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingli Cai
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqi Shen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Tan
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Big Data in Health Science, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Tanimura K, Aldrich MC, Jaworski J, Xing J, Okawa S, Chandra D, Nouraie SM, Nyunoya T. Identifying a Genetic Link Between Lung Function and Psoriasis. Ann Hum Genet 2025; 89:89-95. [PMID: 39718377 PMCID: PMC11982659 DOI: 10.1111/ahg.12587] [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: 07/09/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION The common genetic underpinnings of psoriasis and pulmonary comorbidities have yet to be explored. MATERIAL AND METHODS In this cross-sectional study, we investigated the single-nucleotide polymorphisms (SNPs) associated with psoriasis and their relationship with pulmonary function using data from the UK Biobank (UKBB) and the Vanderbilt University Medical Center Biobank (BioVU). RESULTS Out of the 63 psoriasis-associated SNPs identified in previous genome-wide association studies within the European population, we successfully identified 53 SNPs, including proxy SNPs in UKBB database. Following adjustments using age and sex, 31 SNPs displayed statistically significant associations with psoriasis. Among these, 16 SNPs exhibited significant associations with forced expiratory volume in 1 s (FEV1), 14 with forced vital capacity (FVC), and 5 with the FEV1/FVC ratio in the UKBB. In the validation analysis using the BioVU database, 27 of the 31 psoriasis-associated SNPs were available for examination. Notably, the minor allele of SNP rs8016947 was confirmed to be significant, indicating a reduced risk for psoriasis and improved FEV1. Similarly, the minor alleles of SNPs rs17716942 and rs8016947 were associated with a reduced risk of psoriasis and enhanced FVC. However, none of the 5 SNPs significantly associated with the FEV1/FVC ratio in the UKBB displayed significance in the BioVU dataset. CONCLUSION This study has unveiled genetic variants that bridge the realms of psoriasis and lung function. The genes associated with these variants, including IFIH1, Grancalcin gene (GCA), and NFKB inhibitor alpha gene (NFKBIA), regulate innate immune responses, which suggests that immunodysregulation, a central element in psoriasis pathogenesis, may also impact lung function, alluding to a "skin-lung axis."
Collapse
Affiliation(s)
- Kazuya Tanimura
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Respiratory MedicineNara Medical UniversityKashiharaNaraJapan
| | - Melinda C. Aldrich
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - James Jaworski
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jinchuan Xing
- Department of GeneticsHuman Genetic Institute of New Jersey, Rutgers, the State University of New JerseyPiscatawayNew JerseyUSA
| | - Satoshi Okawa
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Divay Chandra
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Seyed M. Nouraie
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Toru Nyunoya
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Medical Specialty Service LineVeterans Affairs Pittsburgh Healthcare SystemPittsburghPennsylvaniaUSA
| |
Collapse
|
4
|
Kawahara T, Nawa N, Murakami K, Tanaka T, Ohseto H, Takahashi I, Narita A, Obara T, Ishikuro M, Orui M, Noda A, Shinoda G, Nagata Y, Nagaie S, Ogishima S, Sugawara J, Kure S, Kinoshita K, Hozawa A, Fuse N, Tamiya G, Bennett WL, Taub MA, Surkan PJ, Kuriyama S, Fujiwara T. Genetic effects on gestational diabetes mellitus and their interactions with environmental factors among Japanese women. J Hum Genet 2025; 70:265-273. [PMID: 40119124 DOI: 10.1038/s10038-025-01330-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/28/2025] [Indexed: 03/24/2025]
Abstract
Gestational diabetes mellitus (GDM) is common in Japanese women, posing serious risks to mothers and offspring. This study investigated the influence of maternal genotypes on the risk of GDM and examined how these genotypes modify the effects of psychological and dietary factors during pregnancy. We analyzed data from 20,399 women in the Tohoku Medical Megabank Project Birth and Three-Generation Cohort. Utilizing two customized SNP arrays for the Japanese population (Affymetrix Axiom Japonica Array v2 and NEO), we performed a meta-analysis to combine the datasets. Gene-environment interactions were assessed by modeling interaction terms between genome-wide significant single nucleotide polymorphisms (SNPs) and psychological and dietary factors. Our analysis identified two SNP variants, rs7643571 (p = 9.14 × 10-9) and rs140353742 (p = 1.24 × 10-8), located in an intron of the MDFIC2 gene, as being associated with an increased risk of GDM. Additionally, although there were suggestive patterns for interactions between these SNPs and both dietary factors (e.g., carbohydrate and fruit intake) and psychological distress, none of the interaction terms remained significant after Bonferroni correction (p < 0.05/8). While nominal significance was observed in some models (e.g., psychological distress, p = 0.04), the data did not provide robust evidence of effect modification on GDM risk once adjusted for multiple comparisons. These findings reveal novel genetic associations with GDM in Japanese women and highlight the importance of gene-environment interactions in its etiology. Given that previous genome-wide association studies (GWAS) on GDM have primarily focused on Western populations, our study provides new insights by examining an Asian population using a population-specific array.
Collapse
Affiliation(s)
- Tomoki Kawahara
- Department of Public Health, Institute of Science Tokyo, Tokyo, Japan
- Department of Clinical Information Applied Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Nobutoshi Nawa
- Department of Public Health, Institute of Science Tokyo, Tokyo, Japan.
| | - Keiko Murakami
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Toshihiro Tanaka
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
- Bioresource Research Center, Institute of Science Tokyo, Tokyo, Japan
| | - Hisashi Ohseto
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Ippei Takahashi
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Akira Narita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Taku Obara
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Mami Ishikuro
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Masatsugu Orui
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Aoi Noda
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Genki Shinoda
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Yuki Nagata
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Satoshi Nagaie
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Soichi Ogishima
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Junichi Sugawara
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Shigeo Kure
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Atsushi Hozawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Nobuo Fuse
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Gen Tamiya
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Wendy L Bennett
- Division of General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Pamela J Surkan
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Shinichi Kuriyama
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Takeo Fujiwara
- Department of Public Health, Institute of Science Tokyo, Tokyo, Japan
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
5
|
Lin BD, Pries LK, Arias-Magnasco A, Klingenberg B, Linden DE, Blokland GA, van der Meer D, Luykx JJ, Rutten BP, Guloksuz S. Exposome-Wide Gene-By-Environment Interaction Study of Psychotic Experiences in the UK Biobank. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100460. [PMID: 40206033 PMCID: PMC11981733 DOI: 10.1016/j.bpsgos.2025.100460] [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: 10/29/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 04/11/2025] Open
Abstract
Background A previous study successfully identified 148 of 23,098 exposures associated with any psychotic experiences (PEs) in the UK Biobank using an exposome-wide association study (XWAS). Furthermore, research has shown that the polygenic risk score for schizophrenia (PRS-SCZ) is associated with PEs. However, the interaction of these exposures with PRS-SCZ remains unknown. Method To systematically investigate possible gene-by-environment interactions underlying PEs through data-driven agnostic analyses, we conducted 1) conditional XWAS adjusting for PRS-SCZ to estimate the main effects of the exposures and of PRS-SCZ, 2) exposome-wide interaction study (XWIS) to estimate multiplicative and additive interactions between PRS-SCZ and exposures, and 3) correlation analyses between PRS-SCZ and exposures. The study included 148,502 participants from the UK Biobank. Results In the conditional XWAS models, significant effects of PRS-SCZ and 148 exposures on PEs remained statistically significant. In the XWIS model, we found significant multiplicative (multiplicative scale, 1.23; 95% CI, 1.10-1.37; p = 4.0 × 10-4) and additive (relative excess risk due to interaction, 0.55; 95% CI, 0.32-0.77; synergy index, 0.22; 95% CI, 0.14-0.30; and attributable proportion, 1.59; 95% CI, 1.30-1.91; all ps < .05/148) interactions of PRS-SCZ and the variable serious medical conditions/disability with PEs. We additionally identified 6 additive gene-by-environment interactions for mental distress, help-/treatment-seeking behaviors (3 variables), sadness, and sleep problems. In the correlation test focused on 7 exposures that exhibited significant interactions with PRS-SCZ, nonsignificant or small (r < 0.04) gene-by-environment correlations were observed. Conclusions These findings reveal evidence for gene-by-environment interactions underlying PEs and suggest that intertwined pathways of genetic vulnerability and exposures may contribute to psychosis risk.
Collapse
Affiliation(s)
- Bochao Danae Lin
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Lotta-Katrin Pries
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Angelo Arias-Magnasco
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Boris Klingenberg
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - David E.J. Linden
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Gabriëlla A.M. Blokland
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Dennis van der Meer
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jurjen J. Luykx
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
- Department of Psychiatry, Amsterdam University Medical Center, Amsterdam, the Netherlands
- GGZ inGeest Mental Health Care, Amsterdam, the Netherlands
- Neuroscience Mood, Anxiety, Psychosis, Stress & Sleep Research Program, Amsterdam University Medical Center, Amsterdam, the Netherlands
- Public Health Mental Health Research Program, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bart P.F. Rutten
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Sinan Guloksuz
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine, and Life Sciences, Maastricht University Medical Centre, Maastricht, the Netherlands
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
6
|
Xia JW, Li JJ, Qian Y, Han J, Lin M, Wang MY, Chen T, Chai GL, Zhao YN, Hao JW. Observational and genetic evidence highlight the association of modifiable risk factors with the incidence and severity of neuroimmunological disorders. Brain Behav Immun Health 2025; 45:100975. [PMID: 40235834 PMCID: PMC11999315 DOI: 10.1016/j.bbih.2025.100975] [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: 09/26/2024] [Revised: 01/23/2025] [Accepted: 03/06/2025] [Indexed: 04/17/2025] Open
Abstract
Background Myasthenia gravis (MG), multiple sclerosis (MS), and neuromyelitis optica spectrum disorders (NMOSD) are a heterogeneous group of rare neuroimmunological disorders whose incidence rates have increased in recent years. The relationships between modifiable risk factors and neuroimmunological disorders are not fully understood. Methods We utilized multiple logistic regression to estimate the relationships between 38 modifiable risk factors and two neuroimmunological diseases using data from nearly 500,000 individuals in the UK Biobank. Additionally, we applied two-sample Mendelian Randomization (MR) analyses using genetic variants as instrumental variables to investigate the causal relationships of 32 modifiable lifestyle factors with 8 outcomes, representing risk and severity across three neuroimmunological diseases. To further explore the underlying mechanisms, mediation analysis was conducted to elucidate how significant associations might be mediated by intermediate variables. Results Our observational and MR analyses consistently found significant associations (P < 0.05) indicating the number of cigarettes smoked daily, television watching, waist circumference, and BMI are all positively associated with the risk of developing MG. In contrast, moderate-to-vigorous physical activity and higher vitamin D levels are associated with a reduced risk of MS. Moreover, we discovered that the impact of television watching on the risk of MG was mediated by BMI (observational mediation analysis: 26.22%; MR mediation analysis: 9.90%). Conclusions These findings underscore the importance of modifiable risk factors in the development of neuroimmune diseases and support the identification of personalized intervention and prevention strategies. Notably, BMI significantly mediates the relationship between television watching and MG, indicating potential for targeted interventions to mitigate the risk of MG.
Collapse
Affiliation(s)
- Jiang-wei Xia
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Jia-jian Li
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Yu Qian
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, 18 Shilongshan Road, Xihu District, Hangzhou, 310024, Zhejiang, China
| | - Jinmin Han
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Ming Lin
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Ming-yang Wang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Teng Chen
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Guo-liang Chai
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Yi-nan Zhao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
- Beijing Municipal Geriatric Medical Research Center, Beijing, 100053, China
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100069, China
| | - Jun-wei Hao
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
- Beijing Municipal Geriatric Medical Research Center, Beijing, 100053, China
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100069, China
| |
Collapse
|
7
|
Hosseinzadeh Taher MR, Haghighi F, Gotway MB, Liang J. Large-scale benchmarking and boosting transfer learning for medical image analysis. Med Image Anal 2025; 102:103487. [PMID: 40117988 DOI: 10.1016/j.media.2025.103487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 08/03/2024] [Accepted: 01/27/2025] [Indexed: 03/23/2025]
Abstract
Transfer learning, particularly fine-tuning models pretrained on photographic images to medical images, has proven indispensable for medical image analysis. There are numerous models with distinct architectures pretrained on various datasets using different strategies. But, there is a lack of up-to-date large-scale evaluations of their transferability to medical imaging, posing a challenge for practitioners in selecting the most proper pretrained models for their tasks at hand. To fill this gap, we conduct a comprehensive systematic study, focusing on (i) benchmarking numerous conventional and modern convolutional neural network (ConvNet) and vision transformer architectures across various medical tasks; (ii) investigating the impact of fine-tuning data size on the performance of ConvNets compared with vision transformers in medical imaging; (iii) examining the impact of pretraining data granularity on transfer learning performance; (iv) evaluating transferability of a wide range of recent self-supervised methods with diverse training objectives to a variety of medical tasks across different modalities; and (v) delving into the efficacy of domain-adaptive pretraining on both photographic and medical datasets to develop high-performance models for medical tasks. Our large-scale study (∼5,000 experiments) yields impactful insights: (1) ConvNets demonstrate higher transferability than vision transformers when fine-tuning for medical tasks; (2) ConvNets prove to be more annotation efficient than vision transformers when fine-tuning for medical tasks; (3) Fine-grained representations, rather than high-level semantic features, prove pivotal for fine-grained medical tasks; (4) Self-supervised models excel in learning holistic features compared with supervised models; and (5) Domain-adaptive pretraining leads to performant models via harnessing knowledge acquired from ImageNet and enhancing it through the utilization of readily accessible expert annotations associated with medical datasets. As open science, all codes and pretrained models are available at GitHub.com/JLiangLab/BenchmarkTransferLearning (Version 2).
Collapse
Affiliation(s)
| | - Fatemeh Haghighi
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ 85281, USA
| | | | - Jianming Liang
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ 85281, USA.
| |
Collapse
|
8
|
Liu C, Wu X, Li J, Song S, Guan J, Wang Q. Sleep-Associated Traits and Hearing Difficulties in Noise: A Bidirectional Mendelian Randomization Study. Ear Hear 2025; 46:817-826. [PMID: 39828915 PMCID: PMC11984542 DOI: 10.1097/aud.0000000000001625] [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] [Indexed: 01/22/2025]
Abstract
OBJECTIVES The aim of this study was to investigate the causal relationships between sleep-associated traits and hearing difficulties in noise (HDinN) by Mendelian randomization (MR) analysis. DESIGN Single nucleotide polymorphisms associated with chronotype, insomnia, sleep duration, daytime dozing or sleeping, and ease of getting up in the morning were extracted from European population genome-wide association study pooled data for bidirectional MR analysis. The MR-Egger regression, the inverse variance weighted technique, and the weighted median method were used for data analysis. The study was then expanded to include South Asian, East Asian, African, and Greater Middle Eastern populations. RESULTS MR analysis indicated that in European populations, ease of getting up in the morning is a protective factor for HDinN (odds ratio [OR] = 0.932, p = 4.22 × 10 -5 , pFDR = 5.62 × 10 -4 ), while shorter sleep duration was a risk factor (undersleepers: OR = 1.164, p = 0.002, pFDR = 0.014). In addition, there was an indicative causal association between daytime dozing and HDinN (OR = 1.089, p = 0.046, pFDR = 0.123). The conclusions were consistent in African populations (ease of getting up: OR = 0.696, p = 0.012, pFDR = 0.041, sleep duration: OR = 0.677, p = 0.032 pFDR = 0.091, daytime dozing: OR = 1.164, p = 0.002, pFDR = 0.014). In the reverse direction, there was a significant causal association between HDinN and both chronotype (OR = 1.413, p = 0.011, pFDR = 0.042) and ease of getting up in the morning (OR = 0.668, p = 1.75 × 10 -5 , pFDR = 3.49 × 10 -4 ) in European populations, with similar conclusions respectively reached in East Asian (OR = 1.085, p = 0.010, pFDR = 0.045) and African populations (OR = 0.936, p = 0.002, pFDR = 0.012). Furthermore, although not observed in European populations, exploratory studies in non-European populations suggested a potential association between insomnia and HDinN (East Asian: OR = 1.920, p = 0.011, pFDR = 0.043, African: OR = 2.080, p = 0.004, pFDR = 0.019, South Asian: OR = 1.981, p = 1.59 × 10 -4 , PFDR = 0.002, Greater Middle Eastern: OR = 2.394, p = 0.002, pFDR = 0.012), and vice versa (Greater Middle Eastern: OR = 1.056, p = 0.014, pFDR = 0.044). CONCLUSIONS This study identified a potential bidirectional causal relationship between sleep-associated traits and HDinN. However, the underlying mechanisms of the causal relationships reported here have yet to be elucidated.
Collapse
Affiliation(s)
- Chunyan Liu
- Department of Audiology & Vestibular Medicine, Senior Department of Otolaryngology-Head & Neck Surgery, the Sixth Medical Center of People’s Liberation Army (PLA) General Hospital, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
- Nankai University, School of Medicine, Tianjin, China
| | - Xiaonan Wu
- Department of Audiology & Vestibular Medicine, Senior Department of Otolaryngology-Head & Neck Surgery, the Sixth Medical Center of People’s Liberation Army (PLA) General Hospital, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jin Li
- Department of Audiology & Vestibular Medicine, Senior Department of Otolaryngology-Head & Neck Surgery, the Sixth Medical Center of People’s Liberation Army (PLA) General Hospital, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Shan Song
- Nankai University, School of Medicine, Tianjin, China
| | - Jing Guan
- Department of Audiology & Vestibular Medicine, Senior Department of Otolaryngology-Head & Neck Surgery, the Sixth Medical Center of People’s Liberation Army (PLA) General Hospital, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Qiuju Wang
- Department of Audiology & Vestibular Medicine, Senior Department of Otolaryngology-Head & Neck Surgery, the Sixth Medical Center of People’s Liberation Army (PLA) General Hospital, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
- Nankai University, School of Medicine, Tianjin, China
| |
Collapse
|
9
|
Zhou G, Qie X, Zhao H. A Bayesian approach to correcting the attenuation bias of regression using polygenic risk score. Genetics 2025; 229:iyaf018. [PMID: 39891671 DOI: 10.1093/genetics/iyaf018] [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: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Polygenic risk score has become increasingly popular for predicting the value of complex traits. In many settings, polygenic risk score is used as a covariate in regression analysis to study the association between different phenotypes. However, measurement error in polygenic risk score causes attenuation bias in the estimation of regression coefficients. In this paper, we employ a Bayesian approach to accounting for the measurement error of polygenic risk score and correcting the attenuation bias in linear and logistic regression. Through simulation, we show that our approach is able to obtain approximately unbiased estimation of coefficients and credible intervals with correct coverage probability. We also empirically compare our Bayesian measurement error model with the conventional regression model by analyzing real traits in the UK Biobank. The results demonstrate the effectiveness of our approach as it significantly reduces the error in coefficient estimates.
Collapse
Affiliation(s)
- Geyu Zhou
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| | - Xinyue Qie
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
10
|
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 DOI: 10.1038/s41467-025-58684-8] [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: 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
|
11
|
Liu Y, Chang J, Zhao Y, Gao P, Tang Y. Frailty and social contact with dementia risk: A prospective cohort study. J Affect Disord 2025; 375:129-136. [PMID: 39862976 DOI: 10.1016/j.jad.2025.01.112] [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: 10/07/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Frailty and social contact are significant factors influencing dementia risk. While previous studies have separately examined these factors, their combined impact on dementia remains underexplored. METHODS This study included 338,567 UK biobank participants from 2006 to 2010, with follow-up until December 2022. Additionally, 30,408 participants with brain magnetic resonance imaging data were analyzed for hippocampal volume. Cox proportional hazards regression and linear regression models were used to assess associations. RESULTS The study followed 338,567 participants (mean [SD] age, 60.4 [5.2] years; 54.1 % men) for a median of 13.7 years, documenting 7362 cases of all-cause dementia. Both frailty and lower social contact independently increased the risk of all-cause dementia, Alzheimer's disease (AD), and vascular dementia (VaD). Compared to individuals with non-frailty and high social contact, those with lower social contact and higher frailty had a significantly increased risk of all-cause dementia, with the highest risk observed in individuals with frailty and low social contact (HR = 2.65, 95 % CI: 2.27-3.11). Similar patterns were found for AD and VaD. Furthermore, hippocampal volume was significantly reduced in individuals with frailty and low social contact (β = -0.24, 95 % CI: -0.43 to -0.06) compared to those with non-frailty and high social contact. LIMITATIONS The study predominantly included European descent individuals, with most frailty and social contact data based on baseline self-reports. CONCLUSIONS The combination of frailty and low social contact is associated with the highest risk of dementia. These findings suggest that both physiological and social factors should be simultaneously considered in dementia prevention strategies.
Collapse
Affiliation(s)
- Yufei Liu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Chang
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yiwei Zhao
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Peiyang Gao
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China; National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing, China.
| |
Collapse
|
12
|
Zhou Y, Wu Z, Zeng L, Chen R. Combining genetic and non-genetic factors to predict the risk of pancreatic cancer in patients with new-onset diabetes mellitus. BMC Med 2025; 23:224. [PMID: 40234846 DOI: 10.1186/s12916-025-04048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Recent research suggests that new-onset diabetes mellitus (NODM) often results from pancreatic cancer (PC) rather than causing it. Determining if NODM is type 2 diabetes mellitus (T2DM) or PC-related NODM (NODM-PC) aids in the early diagnosis of PC. We developed a NODM-PC risk prediction model to stratify PC risk in patients with NODM. METHODS This study utilized data from the UK Biobank, including 238 NODM-PC cases and 14,825 cancer-free T2DM controls. Polygenic risk scores (PRSs) for PC and T2DM were constructed using previously reported single nucleotide polymorphisms (SNPs) separately, while the NODM-PC PRS was developed by combining SNPs from both. Non-genetic factors were selected as candidate predictors based on prior NODM-PC prediction models. We developed three Cox models to estimate the risk of PC diagnosis within 3 years in the NODM population and evaluated them by internal-external cross-validation. RESULTS Elevated NODM-PC PRS and PC PRS scores positively correlated with NODM-PC risk, while T2DM PRS showed an inverse correlation. The NODM-PC PRS achieved the highest AUC at 0.719. Three Cox models were developed: Model 1 included age at T2DM diagnosis, smoking status, HbA1c, PC PRS, and T2DM PRS; Model 2 replaced PC and T2DM PRS with NODM-PC PRS; Model 3 included only non-genetic factors. Model 2 had the highest discrimination (Harrell's C-index 0.823 (95% CI: 0.806-0.840)), demonstrated the best clinical utility with good calibration, and showed significant classification improvement (continuous net reclassification index: 26.89% and 31.93% for cases, 28.51% and 30.90% for controls, compared to Models 1 and 3). The positive predictive value for the top 1% predicted risk in Model 2 was 13.25%. CONCLUSIONS This NODM-PC PRS enhances NODM-PC risk prediction, efficiently identifies individuals at high risk for PC screening, and improves PC screening efficiency at the population level among NODM individuals.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhuo Wu
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Liangtang Zeng
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
| | - Rufu Chen
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
| |
Collapse
|
13
|
He P, Li H, Liu M, Ye Z, Zhou C, Zhang Y, Yang S, Zhang Y, Qin X. Life's Essential 8 scores, socioeconomic deprivation, genetic susceptibility, and new-onset chronic kidney diseases. Chin Med J (Engl) 2025:00029330-990000000-01518. [PMID: 40223562 DOI: 10.1097/cm9.0000000000003491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND The American Heart Association recently released a new cardiovascular health (CVH) metric, Life's Essential 8 (LE8), for health promotion. However, the association between LE8 scores and the risk of chronic kidney disease (CKD) remains uncertain. We aimed to explore the association of LE8 scores with new-onset CKD and examine whether socioeconomic deprivation and genetic risk modify this association. METHODS A total of 286,908 participants from UK Biobank and without prior CKD were included between 2006 and 2010. CVH was categorized using LE8 scores: low (LE8 scores <50), moderate (LE8 scores ≥50 but <80), and high (LE8 scores ≥80). The study outcome was new-onset CKD, ascertained by data linkage with primary care, hospital inpatient, and death data. Cox proportional hazard regression models were used to investigate the association between CVH categories and new-onset CKD. RESULTS During a median follow-up of 12.5 years, 8857 (3.1%) participants developed new-onset CKD. Compared to the low CVH group, the moderate (adjusted hazards ratio [HR], 0.50; 95% confidence interval [CI]: 0.47-0.53) and high CVH (adjusted HR, 0.31; 95% CI: 0.27-0.34) groups had a significantly lower risk of developing new-onset CKD. The population-attributable risk associated with high vs. intermediate or low CVH scores was 40.3%. Participants who were least deprived (vs. most deprived; adjusted HR, 0.75; 95% CI: 0.71-0.79) and with low genetic risk of CKD (vs. high genetic risk; adjusted HR, 0.89; 95% CI: 0.85-0.94) had a significantly lower risk of developing new-onset CKD. However, socioeconomic deprivation and genetic risks of CKD did not significantly modify the relationship between LE8 scores and new-onset CKD (both P-interaction >0.05). CONCLUSION Achieving a higher LE8 score was associated with a lower risk of developing new-onset CKD, regardless of socioeconomic deprivation and genetic risks of CKD.
Collapse
Affiliation(s)
- Panpan He
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, Guangdong 510515, China
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bao J, Wen J, Chang C, Mu S, Chen J, Shivakumar M, Cui Y, Erus G, Yang Z, Yang S, Wen Z, Zhao Y, Kim D, Duong-Tran D, Saykin AJ, Zhao B, Davatzikos C, Long Q, Shen L. A genetically informed brain atlas for enhancing brain imaging genomics. Nat Commun 2025; 16:3524. [PMID: 40229250 DOI: 10.1038/s41467-025-57636-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025] Open
Abstract
Brain imaging genomics has manifested considerable potential in illuminating the genetic determinants of human brain structure and function. This has propelled us to develop the GIANT (Genetically Informed brAiN aTlas) that accounts for genetic and neuroanatomical variations simultaneously. Integrating voxel-wise heritability and spatial proximity, GIANT clusters brain voxels into genetically informed regions, while retaining fundamental anatomical knowledge. Compared to conventional (non-genetics) brain atlases, GIANT exhibits smaller intra-region variations and larger inter-region variations in terms of voxel-wise heritability. As a result, GIANT yields increased regional SNP heritability, enhanced polygenicity, and its polygenic risk score explains more brain volumetric variation than traditional neuroanatomical brain atlases. We provide extensive validation to GIANT and demonstrate its neuroanatomical validity, confirming its generalizability across populations with diverse genetic ancestries and various brain conditions. Furthermore, we present a comprehensive genetic architecture of the GIANT regions, covering their functional annotation at the molecular levels, their associations with other complex traits/diseases, and the genetic and phenotypic correlations among GIANT-defined imaging endophenotypes. In summary, GIANT constitutes a brain atlas that captures the complexity of genetic and neuroanatomical heterogeneity, thereby enhancing the discovery power and applicability of imaging genomics investigations in biomedical science.
Collapse
Affiliation(s)
- Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, New York, NY, USA
- New York Genome Center (NYGC), New York, NY, USA
| | - Changgee Chang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shizhuo Mu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jiong Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuhan Cui
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhijian Yang
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zixuan Wen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Yize Zhao
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Duy Duong-Tran
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Mathematics, United States Naval Academy, Annapolis, MD, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for AI and Data Science for Integrated Diagnostics (AI2D), Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Jung JY, Ahn Y, Park JW, Jung K, Kim S, Lim S, Jung SH, Kim H, Kim B, Hwang MY, Kim YJ, Park WY, Okbay A, O'Connell KS, Andreassen OA, Myung W, Won HH. Polygenic overlap between subjective well-being and psychiatric disorders and cross-ancestry validation. Nat Hum Behav 2025:10.1038/s41562-025-02155-z. [PMID: 40229577 DOI: 10.1038/s41562-025-02155-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/24/2025] [Indexed: 04/16/2025]
Abstract
Subjective well-being (SWB) is important for understanding human behaviour and health. Although the connection between SWB and psychiatric disorders has been studied, common genetic mechanisms remain unclear. This study aimed to explore the genetic relationship between SWB and psychiatric disorders. Bivariate causal mixture modelling (MiXeR), polygenic risk score (PRS) and Mendelian randomization (MR) analyses showed substantial polygenic overlap and associations between SWB and the psychiatric disorders. Subsequent replication studies in East Asian populations confirmed the polygenic overlap between schizophrenia and SWB. The conditional and conjunctional false discovery rate analyses identified additional or shared genetic loci associated with SWB or psychiatric disorders. Functional annotation revealed enrichment of specific brain tissues and genes associated with SWB. The identified genetic loci showed cross-ancestry transferability between the European and Korean populations. Our findings provide valuable insights into the common genetic mechanisms underlying SWB and psychiatric disorders.
Collapse
Affiliation(s)
- Jin Young Jung
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Psychiatry, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Yeeun Ahn
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Jung-Wook Park
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Kyeongmin Jung
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soyeon Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Soohyun Lim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Sang-Hyuk Jung
- Department of Medical Informatics, Kangwon National University College of Medicine, Chuncheon, South Korea
| | - Hyejin Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Beomsu Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Mi Yeong Hwang
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Chungcheongbuk-do, South Korea
| | - Young Jin Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Chungcheongbuk-do, South Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Aysu Okbay
- Department of Economics, School of Business and Economics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Kevin S O'Connell
- Norwegian Center for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Center for Mental Disorders Research (NORMENT), Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea.
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Chungcheongbuk-do, South Korea.
| |
Collapse
|
16
|
Xu L, Kun E, Pandey D, Wang JY, Brasil MF, Singh T, Narasimhan VM. The genetic architecture of and evolutionary constraints on the human pelvic form. Science 2025; 388:eadq1521. [PMID: 40208988 DOI: 10.1126/science.adq1521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/09/2025] [Indexed: 04/12/2025]
Abstract
Human pelvic evolution following the human-chimpanzee divergence is thought to result in an obstetrical dilemma, a mismatch between large infant brains and narrowed female birth canals, but empirical evidence has been equivocal. By using deep learning on 31,115 dual-energy x-ray absorptiometry scans from UK Biobank, we identified 180 loci associated with seven highly heritable pelvic phenotypes. Birth canal phenotypes showed sex-specific genetic architecture, aligning with reproductive function. Larger birth canals were linked to slower walking pace and reduced back pain but increased hip osteoarthritis risk, whereas narrower birth canals were associated with reduced pelvic floor disorder risk but increased obstructed labor risk. Lastly, genetic correlation between birth canal and head widths provides evidence of coevolution between the human pelvis and brain, partially mitigating the dilemma.
Collapse
Affiliation(s)
- Liaoyi Xu
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Eucharist Kun
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Devansh Pandey
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Joyce Y Wang
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Marianne F Brasil
- Department of Anthropology, Western Washington University, Bellingham, WA, USA
| | - Tarjinder Singh
- The Department of Psychiatry at Columbia University Irving Medical Center, New York, NY, USA
- The New York Genome Center, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute at Columbia University, New York, NY, USA
| | - Vagheesh M Narasimhan
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
- Department of Statistics and Data Science, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
17
|
Cao X, Jiang M, Guan Y, Li S, Duan C, Gong Y, Kong Y, Shao Z, Wu H, Yao X, Li B, Wang M, Xu H, Hao X. Trans-ancestry GWAS identifies 59 loci and improves risk prediction and fine-mapping for kidney stone disease. Nat Commun 2025; 16:3473. [PMID: 40216741 PMCID: PMC11992175 DOI: 10.1038/s41467-025-58782-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Kidney stone disease is a multifactorial disease with increasing incidence worldwide. Trans-ancestry GWAS has become a popular strategy to dissect genetic structure of complex traits. Here, we conduct a large trans-ancestry GWAS meta-analysis on kidney stone disease with 31,715 cases and 943,655 controls in European and East Asian populations. We identify 59 kidney stone disease susceptibility loci, including 13 novel loci and show similar effects across populations. Using fine-mapping, we detect 1612 variants at these loci, and pinpoint 25 causal signals with a posterior inclusion probability >0.5 among them. At a novel locus, we pinpoint TRIOBP gene and discuss its potential link to kidney stone disease. We show that a cross-population polygenic risk score, PRS-CSxEAS&EUR, exhibits superior predictive performance for kidney stone disease than other polygenic risk scores constructed in our study. Relative to individuals in the third quintile of PRS-CSxEAS&EUR, those in the lowest and highest quintiles exhibit distinct kidney stone disease risks with odds ratios of 0.57 (0.51-0.63) and 1.83 (1.68-1.98), respectively. Our results suggest that kidney stone disease patients with higher polygenic risk scores are younger at onset. In summary, our study advances the understanding of kidney stone disease genetic architecture and improves its genetic predictability.
Collapse
Affiliation(s)
- Xi Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Minghui Jiang
- Department of Neurology; Center of excellence for Omics Research (CORe), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yunlong Guan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chen Duan
- Department of Urology, Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Urology, Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yifan Kong
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhonghe Shao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongji Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiangyang Yao
- Department of Urology, Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bo Li
- Department of Urology, Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Miao Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hua Xu
- Department of Urology, Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Zhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier KR, Chittoor G, Josyula NS, Meyer M, Gupta S, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SLR, Kelly TN, Lange EM, LeNoir M, Li C, Le Marchand L, McDonald MLN, McHugh CP, Morrison AC, Naseri T, O'Connell J, O'Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao DC, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PWF, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Ida Chen YD, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, Kooperberg C, Minster RL, Mitchell BD, Nouraie M, Psaty BM, Raffield LM, Reiner AP, Rich SS, Rotter JI, Benjamin Shoemaker M, Smith NL, Taylor KD, Telen MJ, Weiss ST, Zhang Y, Heard-Costa N, Sun YV, Lin X, Cupples LA, Lange LA, Liu CT, Loos RJF, North KE, Justice AE. Whole genome sequencing analysis of body mass index identifies novel African ancestry-specific risk allele. Nat Commun 2025; 16:3470. [PMID: 40216759 PMCID: PMC11992084 DOI: 10.1038/s41467-025-58420-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Obesity is a major public health crisis associated with high mortality rates. Previous genome-wide association studies (GWAS) investigating body mass index (BMI) have largely relied on imputed data from European individuals. This study leveraged whole-genome sequencing (WGS) data from 88,873 participants from the Trans-Omics for Precision Medicine (TOPMed) Program, of which 51% were of non-European population groups. We discovered 18 BMI-associated signals (P < 5 × 10-9), including two secondary signals. Notably, we identified and replicated a novel low-frequency single nucleotide polymorphism (SNP) in MTMR3 that was common in individuals of African descent. Using a diverse study population, we further identified two novel secondary signals in known BMI loci and pinpointed two likely causal variants in the POC5 and DMD loci. Our work demonstrates the benefits of combining WGS and diverse cohorts in expanding current catalog of variants and genes confer risk for obesity, bringing us one step closer to personalized medicine.
Collapse
Affiliation(s)
- Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanfei Xu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kendra R Ferrier
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Mariah Meyer
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Shreyash Gupta
- Population Health Sciences, Geisinger, Danville, PA, USA
| | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zilin Li
- Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
- School of Mathematics and Statistics and KLAS, Northeast Normal University, Changchun, Jilin, China
| | - Matthew A Allison
- Department of Family Medicine, Division of Preventive Medicine, The University of California San Diego, La Jolla, CA, USA
| | - Diane M Becker
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jai G Broome
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Erin J Buth
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Kyong-Mi Chang
- The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sameer Chavan
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yen-Feng Chiu
- Institute of Population Health Sciences, National Health Research Institutes, Taipei, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, Division of Metabolism/Endocrinology, National Taiwan University Hospital, Taipei, Taiwan
| | - Matthew P Conomos
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Dawn L DeMeo
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ravindranath Duggirala
- Life Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
- Department of Health and Behavioral Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Celeste Eng
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Alison E Fohner
- Epidemiology, Institute of Public Health Genetics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Barry I Freedman
- Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiuqing Guo
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Chris Haiman
- Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin D Heavner
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - James E Hixson
- Department of Epidemiology, School of Public Health, UTHealth Houston, Houston, TX, USA
| | - Yuk-Lam Ho
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Brian D Hobbs
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Donglei Hu
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Chii-Min Hwu
- Department of Medicine, Division of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei, Taiwan, Taiwan
| | | | - Deepti Jain
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Rita R Kalyani
- Department of Medicine, Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Ethan M Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael LeNoir
- Department of Pediatrics, Bay Area Pediatrics, Oakland, CA, USA
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Merry-Lynn N McDonald
- Department of Medicine, Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caitlin P McHugh
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Take Naseri
- Naseri & Associates Public Health Consultancy Firm and Family Health Clinic, Apia, Samoa
- International Health Institute, Brown University, Providence, RI, USA
| | - Jeffrey O'Connell
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, MD, USA
| | - Christopher J O'Donnell
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James A Perry
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D C Rao
- Center for Biostatistics and Data Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Elizabeth A Regan
- Department of Medicine, Rheumatology, National Jewish Health, Denver, CO, USA
| | | | - Dan M Roden
- Medicine, Pharmacology, and Biomedical Informatics, Clinical Pharmacology and Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | | | - Zeyuan Wang
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Daniel E Weeks
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer Wessel
- Department of Epidemiology, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
- Diabaetes Translational Research Center, Indiana University, Indianapolis, IN, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Peter W F Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lisa R Yanek
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachary T Yoneda
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics, Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Donna K Arnett
- Department of Epidemiology, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Esteban G Burchard
- Bioengineering and Therapeutic Sciences and Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yii-Der Ida Chen
- Department of Medical Genetics, Genomic Outcomes, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Myriam Fornage
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Victor R Gordeuk
- Department of Medicine, School of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lifang Hou
- Northwestern University, Chicago, IL, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ryan L Minster
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland, Baltimore, MD, USA
| | - Mehdi Nouraie
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - M Benjamin Shoemaker
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas L Smith
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veterans Affairs, Seattle, WA, USA
| | - Kent D Taylor
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marilyn J Telen
- Department of Medicine, Division of Hematology, Duke University School of Medical, Durham, NC, USA
| | - Scott T Weiss
- Department of Medicine, Channing Division of Network Medicine, Harvard Medical School, Boston, MA, USA
| | - Yingze Zhang
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nancy Heard-Costa
- Framingham Heart Study, School of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
| | - L Adrienne Cupples
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne E Justice
- Population Health Sciences, Geisinger, Danville, PA, USA.
| |
Collapse
|
19
|
Weine E, Smith SP, Knowlton RK, Harpak A. Trade-offs in modeling context dependency in complex trait genetics. eLife 2025; 13:RP99210. [PMID: 40207770 PMCID: PMC11984953 DOI: 10.7554/elife.99210] [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] [Indexed: 04/11/2025] Open
Abstract
Genetic effects on complex traits may depend on context, such as age, sex, environmental exposures, or social settings. However, it remains often unclear if the extent of context dependency, or gene-by-environment interaction (GxE), merits more involved models than the additive model typically used to analyze data from genome-wide association studies (GWAS). Here, we suggest considering the utility of GxE models in GWAS as a trade-off between bias and variance parameters. In particular, we derive a decision rule for choosing between competing models for the estimation of allelic effects. The rule weighs the increased estimation noise when context is considered against the potential bias when context dependency is ignored. In the empirical example of GxSex in human physiology, the increased noise of context-specific estimation often outweighs the bias reduction, rendering GxE models less useful when variants are considered independently. However, for complex traits, we argue that the joint consideration of context dependency across many variants mitigates both noise and bias. As a result, polygenic GxE models can improve both estimation and trait prediction. Finally, we exemplify (using GxDiet effects on longevity in fruit flies) how analyses based on independently ascertained 'top hits' alone can be misleading, and that considering polygenic patterns of GxE can improve interpretation.
Collapse
Affiliation(s)
- Eric Weine
- Department of Integrative Biology, The University of Texas at AustinAustinUnited States
- Department of Population Health, The University of Texas at AustinAustinUnited States
- Department of Human Genetics, University of ChicagoChicagoUnited States
| | - Samuel Pattillo Smith
- Department of Integrative Biology, The University of Texas at AustinAustinUnited States
- Department of Population Health, The University of Texas at AustinAustinUnited States
| | - Rebecca Kathryn Knowlton
- Department of Statistics and Data Sciences, The University of Texas at AustinAustinUnited States
| | - Arbel Harpak
- Department of Integrative Biology, The University of Texas at AustinAustinUnited States
- Department of Population Health, The University of Texas at AustinAustinUnited States
| |
Collapse
|
20
|
Moen GH, Hwang LD, Brito Nunes C, Warrington NM, Evans DM. The genetics of low and high birthweight and their relationship with cardiometabolic disease. Diabetologia 2025:10.1007/s00125-025-06420-8. [PMID: 40210729 DOI: 10.1007/s00125-025-06420-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/11/2025] [Indexed: 04/12/2025]
Abstract
AIMS/HYPOTHESIS Low birthweight infants are at increased risk not only of mortality, but also of type 2 diabetes mellitus and CVD in later life. At the opposite end of the spectrum, high birthweight infants have increased risk of birth complications, such as shoulder dystocia, neonatal hypoglycaemia and obesity, and similarly increased risk of type 2 diabetes mellitus and CVD. However, previous genome-wide association studies (GWAS) of birthweight in the UK Biobank have primarily focused on individuals within the 'normal' range and have excluded individuals with high and low birthweight (<2.5 kg or >4.5 kg). The aim of this study was to investigate genetic variation associated within the tail ends of the birthweight distribution, to: (1) see whether the genetic factors operating in these regions were different from those that explained variation in birthweight within the normal range; (2) explore the genetic correlation between extremes of birthweight and cardiometabolic disease; and (3) investigate whether analysing the full distribution of birthweight values, including the extremes, improved the ability to detect genuine loci in GWAS. METHODS We performed case-control GWAS analysis of low (<2.5 kg) and high (>4.5 kg) birthweight in the UK Biobank using REGENIE software (Nlow=20,947; Nhigh=12,715; Ncontrols=207,506) and conducted three continuous GWAS of birthweight, one including the full range of birthweights, one involving a truncated GWAS including only individuals with birthweights between 2.5 and 4.5 kg and a third GWAS that winsorised birthweight values <2.5 kg and >4.5 kg. Additionally, we performed bivariate linkage disequilibrium (LD) score regression to estimate the genetic correlation between low/normal/high birthweight and cardiometabolic traits. RESULTS Bivariate LD score regression analyses suggested that high birthweight had a mostly similar genetic aetiology to birthweight within the normal range (genetic correlation coefficient [rG]=0.91, 95% CI 0.83, 0.99), whereas there was more evidence for a separate set of genes underlying low birthweight (rG=-0.74, 95% CI 0.66, 0.82). Low birthweight was also significantly positively genetically correlated with most cardiometabolic traits and diseases we examined, whereas high birthweight was mostly positively genetically correlated with adiposity and anthropometric-related traits. The winsorisation strategy performed best in terms of locus detection, with the number of independent genome-wide significant associations (p<5×10-8) increasing from 120 genetic variants at 94 loci in the truncated GWAS to 270 genetic variants at 178 loci, including 27 variants at 25 loci that had not been identified in previous birthweight GWAS. This included a novel low-frequency missense variant in the ABCC8 gene, a gene known to be involved in congenital hyperinsulinism, neonatal diabetes mellitus and MODY, that was estimated to be responsible for a 170 g increase in birthweight amongst carriers. CONCLUSIONS/INTERPRETATION Our results underscore the importance of genetic factors in the genesis of the phenotypic correlation between birthweight and cardiometabolic traits and diseases.
Collapse
Affiliation(s)
- Gunn-Helen Moen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- The Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia.
| | - Liang-Dar Hwang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Caroline Brito Nunes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Nicole M Warrington
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- The Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - David M Evans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
- The Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia.
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| |
Collapse
|
21
|
Yao XI, Sun S, Yang Q, Tong X, Shen C. Associations between multiple ambient air pollutants, genetic risk, and incident mental disorders: An interaction study in the UK population. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 973:179137. [PMID: 40120411 DOI: 10.1016/j.scitotenv.2025.179137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Mental disorders can be triggered by genetic and environmental risk factors. Limited studies have explored the effects of long-term exposure to air pollution on mental disorders, and most of the studies have focused on individual air pollutants. This study aimed to examine the relationship between long-term exposure to multiple air pollutants and incident mental disorders, including depression, anxiety, and schizophrenia, and whether the associations were affected by genetic susceptibility. Participants in the UK Biobank with no history of mental disorders were followed from baseline (2006 to 2010) to October 31st, 2022. Cox regression was applied to evaluate the correlation between PM2.5 absorbance, PM2.5, PM2.5-10, PM10, NO2, and NOx and any or specific mental disorders. Additive and multiplicative scales were used to measure the interaction between air pollution and schizophrenia polygenic risk score (PRS), depression PRS, or anxiety PRS on specific mental diseases. After a median of 13.36 years of follow-up on 252,376 participants, we observed per interquartile increase of PM2.5 absorbance (0.32 per meter), PM2.5 (1.28 μg/m3), NO2 (10.08 μg/m3), and NOx (16.78 μg/m3) were related to a 2-6 % higher risk of incident mental disorders. The HR (95 % CI) of incident mental disorder for the 2nd, 3rd, and 4th quartile of the air pollution score were 1.05 (1.01-1.18), 1.13 (1.09-1.18), and 1.14 (1.09-1.19), respectively, in comparison to the lowest level of the score. Per interquartile increase in the air pollution score was associated with a 6 %, 24 %, 4 %, and 6 % higher risk of incident mental disorders, schizophrenia, depression, and anxiety, respectively. No interaction between air pollution and genetic risk of schizophrenia, depression or anxiety on corresponding incident disorders was observed. These findings emphasize the importance of implementing air pollution control standards to decrease the burden of mental disorders.
Collapse
Affiliation(s)
- Xiaoxin I Yao
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China; Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, PR China
| | - Shengzhi Sun
- School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Qian Yang
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Xinning Tong
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China.
| | - Chen Shen
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK; National Institute for Health Research Health Protection Research Unit in Chemical and Radiation Threats and Hazards, Imperial College London, UK.
| |
Collapse
|
22
|
Yngvadottir B, Richman L, Andreou A, Woodley J, Luharia A, Lim D, Maher ER, Marciniak SJ. Inherited predisposition to pneumothorax: estimating the frequency of Birt-Hogg-Dubé syndrome from genomics and population cohorts. Thorax 2025:thorax-2024-221738. [PMID: 40210444 DOI: 10.1136/thorax-2024-221738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Birt-Hogg-Dubé syndrome (BHDS) is the most common monogenic cause of pneumothorax. Most affected families have pathogenic variants in the FLCN gene. Using large genomic registries (UK Biobank (UKB), 100,000 Genomes Project and East London Genes & Health) including >550 000 individuals, we demonstrate that the frequency of clinically validated loss-of-function FLCN variants is 1 in 2710 to 4190. While the lifetime risk of pneumothorax in FLCN mutation carriers in the UKB and a BHDS clinical cohort was substantial (28.4% and 37.3%, respectively, to age 65 years), the lifetime risk of renal cancer was significantly lower in UKB than in BHDS patients (1% vs 32.1%). These findings highlight the importance of clinical context in managing individuals with FLCN mutations.
Collapse
Affiliation(s)
- Bryndis Yngvadottir
- Department of Genomic Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Lucy Richman
- Department of Respiratory Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Avgi Andreou
- Department of Genomic Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Jessica Woodley
- Central and South Genomic Laboratory Hub, Birmingham Women's and Children's Hospitals NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Anita Luharia
- Central and South Genomic Laboratory Hub, Birmingham Women's and Children's Hospitals NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Derek Lim
- Department of Clinical Genetics, Birmingham Women's and Children's Hospitals NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Eamonn R Maher
- Department of Genomic Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Department of Clinical Genetics, Birmingham Women's and Children's Hospitals NHS Foundation Trust, Birmingham, Birmingham, UK
- Aston University, Birmingham, UK
| | - Stefan J Marciniak
- Department of Respiratory Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
McCaw ZR, Dey R, Somineni H, Amar D, Mukherjee S, Sandor K, Karaletsos T, Koller D, Aschard H, Smith GD, MacArthur D, O'Dushlaine C, Soare TW. Pitfalls in performing genome-wide association studies on ratio traits. HGG ADVANCES 2025; 6:100406. [PMID: 39818621 PMCID: PMC11808723 DOI: 10.1016/j.xhgg.2025.100406] [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: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Genome-wide association studies (GWASs) are often performed on ratios composed of a numerator trait divided by a denominator trait. Examples include body mass index (BMI) and the waist-to-hip ratio, among many others. Explicitly or implicitly, the goal of forming the ratio is typically to adjust for an association between the numerator and denominator. While forming ratios may be clinically expedient, there are several important issues with performing GWAS on ratios. Forming a ratio does not "adjust" for the denominator in the sense of conditioning on it, and it is unclear whether associations with ratios are attributable to the numerator, the denominator, or both. Here we demonstrate that associations arising in ratio GWAS can be entirely denominator driven, implying that at least some associations uncovered by ratio GWAS may be due solely to a putative adjustment variable. In a survey of 10 common ratio traits, we find that the ratio model disagrees with the adjusted model (performing GWAS on the numerator while conditioning on the denominator) at around 1/3 of loci. Using BMI as an example, we show that variants detected by only the ratio model are more strongly associated with the denominator (height), while variants detected by only the adjusted model are more strongly associated with the numerator (weight). Although the adjusted model provides effect sizes with a clearer interpretation, it is susceptible to collider bias. We propose and validate a simple method of correcting for the genetic component of collider bias via leave-one-chromosome-out polygenic scoring.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hugues Aschard
- Institut Pasteur, Université Paris Cité, Department of Computational Biology, Paris, France
| | | | - Daniel MacArthur
- Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia; Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | | | | |
Collapse
|
24
|
Xie H, Xi Z, Wen S, Zhang W, Liu Y, Zheng J, Feng H, Wu D, Li Y. Associations Between Chronotype, Genetic Susceptibility and Risk of Colorectal Cancer in UK Biobank. J Epidemiol Glob Health 2025; 15:57. [PMID: 40208451 PMCID: PMC11985712 DOI: 10.1007/s44197-025-00399-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Sleep problems are common in the general population, with evidence suggesting a link between circadian rhythm disruptions and various health outcomes. However, the role of chronotype in influencing colorectal cancer (CRC) risk, particularly in conjunction with genetic predisposition, remains unclear and warrants further investigation. METHODS We analyzed data from 295,729 UK Biobank participants, among whom 4305 developed colorectal cancer. Chronotype was self-reported as morning or evening type, and a polygenic risk score for chronotype was generated from 316 genome-wide significant SNPs using 23andMe effect sizes to reduce overlap bias. Colorectal cancer risk was estimated using Cox proportional hazards models adjusted for age, sex, smoking, alcohol consumption, and the Townsend index. RESULTS Late chronotype and high polygenic risk were independently associated with an increased risk of CRC. Compared to participants with an early chronotype, those with a late chronotype exhibited a 6.5% increased risk of CRC [HR 1.065, P = 0.046]. Similarly, individuals in the high genetic risk group had a 11.0% increased risk compared with those in the low genetic risk group [HR, 1.110, P = 0.032]. Stratified analyses revealed that individuals with an intermediate genetic risk who had a late chronotype showed a 17.6% higher risk of CRC [OR, 1.176, P = 0.004], whereas those with a high genetic risk had a 25.3% increase [OR, 1.253, P = 0.001]. Through analyzing the combined effects of chronotype and PRS, we found that among individuals with an early chronotype, those with intermediate PRS had a 15.4% increased risk of CRC [HR, 1.154, P = 0.005], and those with high PRS had a 14.7% increased risk [HR, 1.147, P = 0.027]. CONCLUSIONS Our findings highlight the importance of considering circadian rhythm patterns and genetic predispositions when assessing CRC risk, suggesting that chronotype may be associated with CRC risk, but further studies are needed to integrate objective circadian measurements.
Collapse
Affiliation(s)
- Huajie Xie
- Guangdong Medical University, Zhanjiang, 524000, China
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zhihui Xi
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Suqi Wen
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, 341000, China
| | - WenRunbei Zhang
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yongfeng Liu
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jiabin Zheng
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Huolun Feng
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Deqing Wu
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yong Li
- Guangdong Medical University, Zhanjiang, 524000, China.
- Department of Gastrointestinal Surgery, Department of Genral Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
25
|
Zhong X, Mitchell R, Billstrand C, Thompson EE, Sakabe NJ, Aneas I, Salamone IM, Gu J, Sperling AI, Schoettler N, Nóbrega MA, He X, Ober C. Integration of functional genomics and statistical fine-mapping systematically characterizes adult-onset and childhood-onset asthma genetic associations. Genome Med 2025; 17:35. [PMID: 40205616 PMCID: PMC11983851 DOI: 10.1186/s13073-025-01459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified hundreds of loci underlying adult-onset asthma (AOA) and childhood-onset asthma (COA). However, the causal variants, regulatory elements, and effector genes at these loci are largely unknown. METHODS We performed heritability enrichment analysis to determine relevant cell types for AOA and COA, respectively. Next, we fine-mapped putative causal variants at AOA and COA loci. To improve the resolution of fine-mapping, we integrated ATAC-seq data in blood and lung cell types to annotate variants in candidate cis-regulatory elements (CREs). We then computationally prioritized candidate CREs underlying asthma risk, experimentally assessed their enhancer activity by massively parallel reporter assay (MPRA) in bronchial epithelial cells (BECs) and further validated a subset by luciferase assays. Combining chromatin interaction data and expression quantitative trait loci, we nominated genes targeted by candidate CREs and prioritized effector genes for AOA and COA. RESULTS Heritability enrichment analysis suggested a shared role of immune cells in the development of both AOA and COA while highlighting the distinct contribution of lung structural cells in COA. Functional fine-mapping uncovered 21 and 67 credible sets for AOA and COA, respectively, with only 16% shared between the two. Notably, one-third of the loci contained multiple credible sets. Our CRE prioritization strategy nominated 62 and 169 candidate CREs for AOA and COA, respectively. Over 60% of these candidate CREs showed open chromatin in multiple cell lineages, suggesting their potential pleiotropic effects in different cell types. Furthermore, COA candidate CREs were enriched for enhancers experimentally validated by MPRA in BECs. The prioritized effector genes included many genes involved in immune and inflammatory responses. Notably, multiple genes, including TNFSF4, a drug target undergoing clinical trials, were supported by two independent GWAS signals, indicating widespread allelic heterogeneity. Four out of six selected candidate CREs demonstrated allele-specific regulatory properties in luciferase assays in BECs. CONCLUSIONS We present a comprehensive characterization of causal variants, regulatory elements, and effector genes underlying AOA and COA genetics. Our results supported a distinct genetic basis between AOA and COA and highlighted regulatory complexity at many GWAS loci marked by both extensive pleiotropy and allelic heterogeneity.
Collapse
Affiliation(s)
- Xiaoyuan Zhong
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Robert Mitchell
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Emma E Thompson
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Noboru J Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Isabella M Salamone
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Jing Gu
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Anne I Sperling
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nathan Schoettler
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Marcelo A Nóbrega
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
26
|
Middha P, Kachuri L, Nierenberg JL, Graff RE, Cavazos TB, Hoffmann TJ, Zhang J, Alexeeff S, Habel L, Corley DA, Van Den Eeden S, Kushi LH, Ziv E, Sakoda LC, Witte JS. Unraveling the genetic landscape of susceptibility to multiple primary cancers. HGG ADVANCES 2025; 6:100413. [PMID: 39910817 PMCID: PMC11910107 DOI: 10.1016/j.xhgg.2025.100413] [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: 10/31/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
With advances in cancer screening and treatment, there is a growing population of cancer survivors who may develop subsequent primary cancers. While hereditary cancer syndromes account for only a portion of multiple cancer cases, we sought to explore the role of common genetic variation in susceptibility to multiple primary tumors. We conducted a cross-ancestry genome-wide association study (GWAS) and transcriptome-wide association study (TWAS) of 10,983 individuals with multiple primary cancers, 84,475 individuals with single cancer, and 420,944 cancer-free controls from two large-scale studies. Our GWAS identified six lead variants across five genomic regions that were significantly associated (p < 5 × 10-8) with the risk of developing multiple primary tumors (overall and invasive) relative to cancer-free controls (at 3q26, 8q24, 10q24, 11q13.3, and 17p13). We also found one variant significantly associated with multiple cancers when compared with single cancer cases (at 22q13.1). Multi-tissue TWAS detected associations with genes involved in telomere maintenance in two of these regions (ACTRT3 in 3q26 and SLK and STN1 in 10q24) and the development of multiple cancers. Additionally, the TWAS also identified several novel genes associated with multiple cancers, including two immune-related genes, IRF4 and TNFRSF6B. Telomere maintenance and immune dysregulation emerge as central, common pathways influencing susceptibility to multiple cancers. These findings underscore the importance of exploring shared mechanisms in carcinogenesis, offering insights for targeted prevention and intervention strategies.
Collapse
Affiliation(s)
- Pooja Middha
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Jovia L Nierenberg
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca E Graff
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA; Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Taylor B Cavazos
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas J Hoffmann
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Jie Zhang
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Stacey Alexeeff
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Laurel Habel
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Douglas A Corley
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | | | - Lawrence H Kushi
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Elad Ziv
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Lori C Sakoda
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA; Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - John S Witte
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA; Stanford Cancer Institute, Stanford University, Stanford, CA, USA; Department of Biomedical Data Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Karp-Tatham E, O'Neill CR, Knight JC, Mentzer AJ, Chong AY. Lack of association between classical HLA genes and asymptomatic SARS-CoV-2 infection. HGG ADVANCES 2025; 6:100382. [PMID: 39491366 PMCID: PMC11937655 DOI: 10.1016/j.xhgg.2024.100382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Affiliation(s)
- Eleanor Karp-Tatham
- The Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Callum R O'Neill
- The Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Julian C Knight
- The Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford, UK
| | - Alexander J Mentzer
- The Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford, UK.
| | - Amanda Y Chong
- The Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
28
|
Li S, Mu A, Jing Z, Liu Z, Cao X, Guo J, Xi Y, Guo Q. Cross ethnic Mendelian randomization analysis reveals causal relationship between air pollution and risk of kidney stones. Sci Rep 2025; 15:12132. [PMID: 40204920 PMCID: PMC11982192 DOI: 10.1038/s41598-025-97436-y] [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: 08/25/2024] [Accepted: 04/04/2025] [Indexed: 04/11/2025] Open
Abstract
Accumulating evidence has indicated that exposures to air pollution increase the odds of kidney stones. However, the previous research methods were limited. To address this gap, we employed genome-wide association studies (GWAS) datasets and Mendelian randomization (MR) to verify the causation. Applying publicly accessible summary datasets from UK Biobank, FinnGen consortium and Biobank Japan, a two-sample MR, and further multivariate MR were carried out to calculate the causality between air pollution [particulate matter 2.5 (PM2.5), PM2.5 absorbance, PM2.5-10, PM10, nitrogen dioxide, and nitrogen oxides] and kidney stone risk in three different populations (European, East Asian, and South Asian). The inverse variance weighted (IVW) was utilized for its first-step assessment, supplemented with MR-Egger, weighted median, Cochran's Q test, MR-Egger intercept and leave-one-out analysis to ensure the robustness. Employing IVW, we discovered in the European population that PM2.5 absorbance was statistically correlated with kidney stone risk (odds ratio (OR) = 1.40; 95% confidence interval (CI), 1.01-1.94; P = 0.04), with no heterogeneity, pleiotropy, or sensitivity observed. Additionally, the MVMR result revealed the directly causative connection between a single PM2.5 absorbance and the increase in kidney stone risk (OR = 1.77, 95%CI: 1.06-2.98, p = 0.03). Our investigation proposed the correlation between PM2.5 absorbance and an increased risk of kidney stones in European populations. The control of air pollution, especially PM2.5, may have crucial implications for the prevention of kidney stones.
Collapse
Affiliation(s)
- Shuangping Li
- Department of Urology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, China
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China
| | - Aijia Mu
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China
| | - Zhinan Jing
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China
| | - Ziyi Liu
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China
| | - Xinfang Cao
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China
| | - Jincheng Guo
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China
| | - Yujia Xi
- Department of Urology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, China.
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China.
| | - Qiang Guo
- Department of Urology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, China.
- Male Reproductive Health Research Center, Shanxi Medical University, Shanxi Province, Jinzhong, China.
| |
Collapse
|
29
|
Lin C, Xia M, Dai Y, Huang Q, Sun Z, Zhang G, Luo R, Peng Q, Li J, Wang X, Lin H, Gao X, Tang H, Shen X, Wang S, Jin L, Hao X, Zheng Y. Cross-ancestry analyses of Chinese and European populations reveal insights into the genetic architecture and disease implication of metabolites. CELL GENOMICS 2025; 5:100810. [PMID: 40118068 DOI: 10.1016/j.xgen.2025.100810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025]
Abstract
Differential susceptibilities to various diseases and corresponding metabolite variations have been documented across diverse ethnic populations, but the genetic determinants of these disparities remain unclear. Here, we performed large-scale genome-wide association studies of 171 directly quantifiable metabolites from a nuclear magnetic resonance-based metabolomics platform in 10,792 Han Chinese individuals. We identified 15 variant-metabolite associations, eight of which were successfully replicated in an independent Chinese population (n = 4,480). By cross-ancestry meta-analysis integrating 213,397 European individuals from the UK Biobank, we identified 228 additional variant-metabolite associations and improved fine-mapping precision. Moreover, two-sample Mendelian randomization analyses revealed evidence that genetically predicted levels of triglycerides in high-density lipoprotein were associated with a higher risk of coronary artery disease and that of glycine with a lower risk of heart failure in both ancestries. These findings enhance our understanding of the shared and specific genetic architecture of metabolites as well as their roles in complex diseases across populations.
Collapse
Affiliation(s)
- Chenhao Lin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qingxia Huang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Guoqing Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; National Genomics Data Center& Bio-Med Big Data Center, University of Chinese Academy of Sciences, Chinese Academy of Science, Shanghai 200031, China
| | - Ruijin Luo
- Shanghai Southgene Technology Co., Ltd., Shanghai 201203, China
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinxi Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xiaofeng Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan University-the People's Hospital of Rugao Joint Research Institute of Longevity and Aging, Rugao, Jiangsu 226500, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xia Shen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, Guangdong 511400, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
30
|
Ogloblinsky MSC, Conrad DF, Baudot A, Tournier-Lasserve E, Génin E, Marenne G. Benchmark of computational methods to detect digenism in sequencing data. Eur J Hum Genet 2025:10.1038/s41431-025-01834-9. [PMID: 40204980 DOI: 10.1038/s41431-025-01834-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Digenic inheritance is characterized by the combined alteration of two different genes leading to a disease. It could explain the etiology of many currently undiagnosed rare diseases. With the advent of next-generation sequencing technologies, the identification of digenic inheritance patterns has become more technically feasible, yet still poses significant challenges without any gold standard method. Here, we present a comprehensive overview of the existing methods developed to detect digenic inheritance in sequencing data and provide a classification in cohort-based and individual-based methods. The latter category of methods appeared the most applicable to rare diseases, especially the ones not needing patient phenotypic description as input. We discuss the availability of the different methods, their output and scalability to inform potential users. Focusing on methods to detect digenic inheritance in the case of very rare or heterogeneous diseases, we propose a benchmark using different real-life scenarios involving known digenic and putative neutral pairs of genes. Among these different methods, DiGePred stood out as the one giving the least number of false positives, ARBOCK as giving the greatest number of true positives, and DIEP as having the best balance between both. By synthesizing the state-of-the-art techniques and providing insights into their practical utility, this benchmark serves as a valuable resource for researchers and clinicians in selecting suitable methodologies for detecting digenic inheritance in a wide range of disorders using sequencing data.
Collapse
Affiliation(s)
| | - Donald F Conrad
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Anaïs Baudot
- Aix Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Elisabeth Tournier-Lasserve
- Université Paris Cité, Inserm, NeuroDiderot, Unité Mixte de Recherche 1141, F-75019, Paris, France
- Assistance publique-Hôpitaux de Paris, Service de Génétique Moléculaire Neurovasculaire, Hôpital Saint-Louis, F-75010, Paris, France
| | - Emmanuelle Génin
- Univ Brest, Inserm, EFS, UMR 1078, GGB, Brest, France
- Assistance publique-Hôpitaux de Paris, Service de Génétique Moléculaire Neurovasculaire, Hôpital Saint-Louis, F-75010, Paris, France
| | | |
Collapse
|
31
|
Su L, Sun Q, Li Y, Alvarez JF, Tao B, Zhang G, Gu Y, Hanudel MR, Espinoza A, Zhang L, Pan C, Hilser JR, Hartiala JA, Li S, Pellegrini M, Allayee H, Lusis AJ, Deb A. Collagen V regulates renal function after kidney injury and can be pharmacologically targeted to enhance kidney repair in mice. Sci Transl Med 2025; 17:eads7714. [PMID: 40203084 DOI: 10.1126/scitranslmed.ads7714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/03/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
Kidney fibrosis determines clinical outcomes in individuals with chronic kidney disease (CKD). The stoichiometric ratio of collagens in renal scar differs from that of healthy kidney extracellular matrix (ECM), but the functional importance of altered collagen types in injured kidneys remains unclear. Using human population studies, we show that circulating protein and renal mRNA amounts of collagen V A1 (COL5A1) exhibited associations with kidney disease and incident CKD risk. We show that Col5a1 regulates the degree of postinjury fibrosis and renal function. Mice with conditionally knocked out Col5a1 (Col5a1 CKO) exhibited decreased renal function and greater renal fibrosis after dietary adenine- or ureteric obstruction-mediated kidney injury. Renal fibroblasts in Col5a1 CKO animals up-regulated the profibrotic αvβ3 integrin. Inhibition of αvβ3 signaling with a small molecule, cilengitide, rescued postinjury renal function in Col5a1 CKO animals. Using the hybrid mouse diversity panel that comprises 100 diverse inbred strains of mice, we observed that gene expression of Col5a1 after injury exhibited genetic variation across 100 strains. Strains with low Col5a1 expression after injury exhibited worse renal function compared with animals that had higher degrees of expression. We next measured Col5a1 expression in peripheral blood mononuclear cells in mice to identify nonresponder strains that did not have increased Col5a1 expression after kidney injury. We observed that administration of cilengitide in nonresponder strains significantly rescued postinjury renal fibrosis and function. These studies point to the feasibility of precision medicine approaches to target Col5a1 for enhancing renal repair.
Collapse
Affiliation(s)
- Lianjiu Su
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qihao Sun
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yusheng Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Juan Felipe Alvarez
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Tao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yiqian Gu
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark R Hanudel
- Department of Pediatric Nephrology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Espinoza
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linlin Zhang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - James R Hilser
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jaana A Hartiala
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hooman Allayee
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
32
|
Huang Y, Zhang Y, Zhang Y, Xiang H, Ye Z, Yang S, Gan X, Wu Y, Zhang Y, Qin X. Hearing impairment, psychological distress, and incident heart failure: a prospective cohort study. Heart 2025:heartjnl-2024-325394. [PMID: 40199582 DOI: 10.1136/heartjnl-2024-325394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/18/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND The relationship between objectively measured hearing ability and the risk of incident heart failure (HF) remains unclear. This study aimed to assess this association, explore potential modifying factors, and examine whether psychological factors mediate this relationship. METHODS We included 164 431 participants from the UK Biobank without HF at baseline. Speech-in-noise hearing ability was measured using the Digit Triplets Test and quantified by the speech-reception-threshold (SRT). Incident HF was identified through hospital admission and death records. Mediation analyses assessed the role of social isolation, psychological distress, and neuroticism. RESULTS Over a median follow-up of 11.7 years, 4449 (2.7%) participants developed incident HF. Higher SRT levels were associated with an increased risk of HF (adjusted HR per SD increment 1.05, 95% CI 1.02 to 1.08). Compared with those with normal hearing, participants with insufficient hearing, poor hearing, or hearing aid use had higher HF risks (adjusted HRs 1.15, 1.28, and 1.26, respectively). Psychological distress mediated 16.9% of the association between SRT levels and HF, while social isolation and neuroticism mediated 3.0% and 3.1%, respectively. The association was stronger in participants without coronary heart disease or stroke at baseline. CONCLUSIONS Poor hearing ability is associated with an increased risk of incident HF, with psychological distress playing a notable mediating role. These findings suggest that hearing health and psychological well-being should be considered in cardiovascular risk assessment and prevention strategies.
Collapse
Affiliation(s)
- Yu Huang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanjun Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanyuan Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao Xiang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziliang Ye
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sisi Yang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoqin Gan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiting Wu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiwei Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xianhui Qin
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Institute of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Sun J, Li X, Ma S, Lin H, Li Z, Jia J, Alizadeh Y, Wu Q, Hou Y, Wang H, Wang Q, Zhang G, Li X, Li W, Zhang C. Impact of anti-VZV IgG levels on Parkinson's disease risk and progression: a Mendelian randomization analysis. Sci Rep 2025; 15:11985. [PMID: 40199927 PMCID: PMC11978792 DOI: 10.1038/s41598-025-96382-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Research suggests a potential link between varicella zoster virus (VZV) and Parkinson's disease (PD), but the causal relationship between anti-VZV IgG levels and PD is not well understood. Using two-sample Mendelian Randomization (MR), we assessed the causal impact of anti-VZV IgG levels on PD risk and progression. Our study found a significant association between higher anti-VZV IgG levels and an increased risk of PD. For PD progression, higher anti-VZV IgG levels were linked to a greater risk of constipation, insomnia, and Restless Legs. These findings remained consistent after sensitivity analyses. In conclusion, our study suggests that elevated anti-VZV IgG levels may contribute to an increased risk and progression of PD, supporting a potential causal link that warrants further mechanistic investigation.
Collapse
Affiliation(s)
- Jinxing Sun
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China
| | - Xiangchen Li
- Department of Computer Science, The University of Manchester, Manchester, UK
| | - Shengmei Ma
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Haopeng Lin
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China
| | - Zhenke Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China
| | - Junheng Jia
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China
| | - Yasaman Alizadeh
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China
| | - Qianqian Wu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China
| | - Ying Hou
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Hong Wang
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, China
| | - Qi Wang
- Department of Gerontology, Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Guangjian Zhang
- Department of Neurology, Weifang People's Hospital, Weifang, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China
| | - Weiguo Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China.
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China.
| | - Chao Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Jinan, China.
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.
- National Medicine-Engineering Interdisciplinary Industry-Education Integration Innovation Platform, Beijing, China.
| |
Collapse
|
34
|
Zhang Y, Wang J, Zong H, Singla RK, Ullah A, Liu X, Wu R, Ren S, Shen B. The comprehensive clinical benefits of digital phenotyping: from broad adoption to full impact. NPJ Digit Med 2025; 8:196. [PMID: 40195396 PMCID: PMC11977243 DOI: 10.1038/s41746-025-01602-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Abstract
Digital phenotyping collects health data digitally, supporting early disease diagnosis and health management. This paper systematically reviews the diversity of research methods in digital phenotyping and its clinical benefits, while also focusing on its importance within the P4 medicine paradigm and its core role in advancing its application in biobanks. Furthermore, the paper envisions the continued clinical benefits of digital phenotyping, driven by technological innovation, global collaboration, and policy support.
Collapse
Affiliation(s)
- Yingbo Zhang
- Department of Urology, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences (CATAS), Haikou, China
| | - Jiao Wang
- Department of Urology, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Computer Science and Information Technology, University of A Coruña, A Coruña, Spain
| | - Hui Zong
- Department of Urology, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K Singla
- Department of Pharmacy, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Amin Ullah
- Department of Pharmacy, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xingyun Liu
- Department of Urology, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Department of Computer Science and Information Technology, University of A Coruña, A Coruña, Spain
| | - Rongrong Wu
- Department of Urology, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shumin Ren
- Department of Urology, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bairong Shen
- Department of Urology, Institutes for Systems Genetics, and Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
- West China Tianfu Hospital Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
35
|
Milani L, Alver M, Laur S, Reisberg S, Haller T, Aasmets O, Abner E, Alavere H, Allik A, Annilo T, Fischer K, Hofmeister R, Hudjashov G, Jõeloo M, Kals M, Karo-Astover L, Kasela S, Kolde A, Krebs K, Krigul KL, Kronberg J, Kruusmaa K, Kukuškina V, Kõiv K, Lehto K, Leitsalu L, Lind S, Luitva LB, Läll K, Lüll K, Metsalu K, Metspalu M, Mõttus R, Nelis M, Nikopensius T, Nurm M, Nõukas M, Oja M, Org E, Palover M, Palta P, Pankratov V, Pantiukh K, Pervjakova N, Pujol-Gualdo N, Reigo A, Reimann E, Smit S, Rogozina D, Särg D, Taba N, Talvik HA, Teder-Laving M, Tõnisson N, Vaht M, Vainik U, Võsa U, Yelmen B, Esko T, Kolde R, Mägi R, Vilo J, Laisk T, Metspalu A. The Estonian Biobank's journey from biobanking to personalized medicine. Nat Commun 2025; 16:3270. [PMID: 40188112 PMCID: PMC11972354 DOI: 10.1038/s41467-025-58465-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/04/2025] [Indexed: 04/07/2025] Open
Abstract
Large biobanks have set a new standard for research and innovation in human genomics and implementation of personalized medicine. The Estonian Biobank was founded a quarter of a century ago, and its biological specimens, clinical, health, omics, and lifestyle data have been included in over 800 publications to date. What makes the biobank unique internationally is its translational focus, with active efforts to conduct clinical studies based on genetic findings, and to explore the effects of return of results on participants. In this review, we provide an overview of the Estonian Biobank, highlight its strengths for studying the effects of genetic variation and quantitative phenotypes on health-related traits, development of methods and frameworks for bringing genomics into the clinic, and its role as a driving force for implementing personalized medicine on a national level and beyond.
Collapse
Affiliation(s)
- Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia.
| | - Maris Alver
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sven Laur
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Sulev Reisberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Toomas Haller
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Oliver Aasmets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Erik Abner
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Helene Alavere
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Annely Allik
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tarmo Annilo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Robin Hofmeister
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Georgi Hudjashov
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maarja Jõeloo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mart Kals
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Liis Karo-Astover
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Silva Kasela
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anastassia Kolde
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kertu Liis Krigul
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jaanika Kronberg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Karoliina Kruusmaa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Viktorija Kukuškina
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kadri Kõiv
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kelli Lehto
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Liis Leitsalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sirje Lind
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Laura Birgit Luitva
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Kristi Läll
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kreete Lüll
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kristjan Metsalu
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mait Metspalu
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - René Mõttus
- Institute of Psychology, University of Tartu, Tartu, Estonia
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Mari Nelis
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tiit Nikopensius
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Miriam Nurm
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Margit Nõukas
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Marek Oja
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Elin Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Marili Palover
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Priit Palta
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Vasili Pankratov
- Centre for Genomics, Evolution and Medicine, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kateryna Pantiukh
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Natalia Pervjakova
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Natàlia Pujol-Gualdo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anu Reigo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Ene Reimann
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Steven Smit
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Diana Rogozina
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Dage Särg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Nele Taba
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Harry-Anton Talvik
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Maris Teder-Laving
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Neeme Tõnisson
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mariliis Vaht
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Uku Vainik
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Psychology, University of Tartu, Tartu, Estonia
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Burak Yelmen
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Raivo Kolde
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
| |
Collapse
|
36
|
Wang LH, Shih MY, Lin YF, Kuo PH, Feng YCA. Polygenic dissection of treatment-resistant depression with proxy phenotypes in the UK Biobank. J Affect Disord 2025; 381:350-359. [PMID: 40187433 DOI: 10.1016/j.jad.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Treatment-resistant depression (TRD) affects one-third of major depressive disorder (MDD) patients. Previous pharmacogenetic studies suggest genetic variation may influence medication response but findings are heterogeneous. We conducted a comprehensive genetic investigation using proxy TRD phenotypes (TRDp) that mirror the treatment options of MDD from UK Biobank primary care records. METHODS Among 15,125 White British MDD patients, we identified TRDp with medication changes (switching or receiving multiple antidepressants [AD]); augmentation therapy (antipsychotics; mood stabilizers; valproate; lithium); or electroconvulsive therapy (ECT). Hospitalized TRDp patients (HOSP-TRDp) were also identified. We conducted genome-wide association analysis, estimated SNP-heritability (hg2), and assessed the genetic burden for nine psychiatric diseases using polygenic risk scores (PRS). RESULTS TRDp patients were more often female, unemployed, less educated, and had higher BMI, with hospitalization rates twice as high as non-TRDp. While no credible risk variants emerged, heritability analysis showed significant genetic influence on TRDp (liability hg2 21-24 %), particularly for HOSP-TRDp (28-31 %). TRDp classified by AD changes and augmentation carried an elevated yet varied polygenic burden for MDD, ADHD, BD, and SCZ. Higher BD PRS increased the likelihood of receiving ECT, lithium, and valproate by 1.27-1.80 fold. Patients in the top 10 % PRS relative to the average had a 12-36 % and 24-51 % higher risk of TRDp and HOSP-TRDp, respectively. CONCLUSIONS Our findings support a significant polygenic basis for TRD, highlighting genetic and phenotypic distinctions from non-TRD. We demonstrate that different TRDp endpoints are enriched with various spectra of psychiatric genetic liability, offering insights into pharmacogenomics and TRD's complex genetic architecture.
Collapse
Affiliation(s)
- Ling-Hua Wang
- Institute of Health Data Analytics and Statistics, College of Public Health, National Taiwan University, Taiwan
| | - Mu-Yi Shih
- Institute of Health Data Analytics and Statistics, College of Public Health, National Taiwan University, Taiwan; Department of Public Health, College of Public Health, National Taiwan University, Taiwan
| | - Yen-Feng Lin
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan; Department of Public Health & Medical Humanities, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Hsiu Kuo
- Department of Public Health, College of Public Health, National Taiwan University, Taiwan; Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Chen A Feng
- Institute of Health Data Analytics and Statistics, College of Public Health, National Taiwan University, Taiwan; Department of Public Health, College of Public Health, National Taiwan University, Taiwan; Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
37
|
Poriswanish N, Eales J, Xu X, Scannali D, Neumann R, Wetton JH, Tomaszewski M, Jobling MA, May CA. Multiple origins and phenotypic implications of an extended human pseudoautosomal region shown by analysis of the UK Biobank. Am J Hum Genet 2025; 112:927-939. [PMID: 39983723 DOI: 10.1016/j.ajhg.2025.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/23/2025] Open
Abstract
The 2.7-Mb major pseudoautosomal region (PAR1) on the short arms of the human X and Y chromosomes plays a critical role in meiotic sex chromosome segregation and male fertility and has been regarded as evolutionarily stable. However, some European Y chromosomes belonging to Y haplogroups (Y-Hgs) R1b and I2a carry an ∼115-kb extension (ePAR [extended PAR]) arising from X-Y non-allelic homologous recombination (NAHR). To investigate the diversity, history, and dynamics of ePAR formation, we screened for its presence, and that of the predicted reciprocal X chromosome deletion, among ∼218,300 46,XY males of the UK Biobank (UKB), a cohort associated with longitudinal clinical data. The UKB incidence of ePAR is ∼0.77%, and that of the deletion is ∼0.02%. We found that Y-Hg I2a sub-lineages accounted for nearly 90% of ePAR cases but, by Y haplotyping and breakpoint sequencing, determined that, in total, there have been at least 18 independent ePAR origins, associated with nine different Y-Hgs. We found examples of ePAR linked to Y-Hg K among men of self-declared Pakistani ancestry and Y-Hg E1, typical of men with African ancestry, showing that ePAR is not restricted to Europeans. ePAR formation is likely random, with high frequencies in some Y-Hgs arising through drift and male-mediated expansions. Sequencing recombination junction fragments identified likely reciprocal events, and the heterogeneity of ePAR and X-deletion junctions highlighted the recurrent nature of the NAHR events. A phenome-wide association study revealed an association between ePAR and elevated levels of circulating IGF-1 as well as musculoskeletal phenotypes.
Collapse
Affiliation(s)
- Nitikorn Poriswanish
- Department of Genetics, Genomics and Cancer Sciences, University of Leicester, Leicester, UK; Department of Forensic Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - James Eales
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David Scannali
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rita Neumann
- Department of Genetics, Genomics and Cancer Sciences, University of Leicester, Leicester, UK
| | - Jon H Wetton
- Department of Genetics, Genomics and Cancer Sciences, University of Leicester, Leicester, UK
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester, UK
| | - Mark A Jobling
- Department of Genetics, Genomics and Cancer Sciences, University of Leicester, Leicester, UK.
| | - Celia A May
- Department of Genetics, Genomics and Cancer Sciences, University of Leicester, Leicester, UK.
| |
Collapse
|
38
|
Tong X, Cui S, Shen H, Yao XI. Developing and validating a nomogram prediction model for osteoporosis risk in the UK biobank: a national prospective cohort. BMC Public Health 2025; 25:1263. [PMID: 40181326 PMCID: PMC11970002 DOI: 10.1186/s12889-025-22485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Osteoporosis is a prevalent bone disease that increases frailty. Developing a nomogram prediction model to predict osteoporosis risk at multiple time points using bone mineral densities, behavioral habits, and clinical risk factors would be essential to identify individual risk and guide prevention. METHODS The study population from the UK Biobank was followed from 2014 to December 31st, 2022. The study outcome was identified as the first occurrence of osteoporosis in the UK Biobank during the follow-up period. After rebalancing with the synthetic minority over-sampling technique, a nomogram prediction model was developed using a LASSO Cox regression. Model discrimination between different risk levels was visualised with Kaplan-Meier curves, and model performance was evaluated with integrated c-index, time-dependent AUC, calibration curves and decision curve analysis (DCA). RESULTS The model identified several risk factors for osteoporosis, including higher age, underweight, and various clinical risk factors (such as menopause, lower hand grip strength, lower bone mineral density, fracture history within 5 years, and a history of chronic disease including hypercholesterolemia, cardiovascular disease, bone disease, arthritis, and cancer). Kaplan-Meier curves showed that risk levels predicted by the nomogram model were significantly distinct. The c-indexes were 0.844 and 0.823 for training and validation datasets, respectively. Time-dependent AUC, calibration curves and DCA indicated good discrimination, model fit and clinical utility, respectively. CONCLUSIONS The nomogram model could properly quantify the five-year risk of osteoporosis and identify high-risk individuals. This might effectively reduce the burden of osteoporosis on the population.
Collapse
Affiliation(s)
- Xinning Tong
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025 Shennan Road, Shenzhen, 518033, China
| | - Shuangnan Cui
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025 Shennan Road, Shenzhen, 518033, China
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Huiyong Shen
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025 Shennan Road, Shenzhen, 518033, China
- Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaoxin Iris Yao
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, 3025 Shennan Road, Shenzhen, 518033, China.
- Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
39
|
Hernández CF, Villaman C, Leu C, Lal D, Mata I, Klein AD, Pérez-Palma E. Polygenic score analysis identifies distinct genetic risk profiles in Alzheimer's disease comorbidities. Sci Rep 2025; 15:11407. [PMID: 40181078 PMCID: PMC11968852 DOI: 10.1038/s41598-025-95755-8] [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: 11/20/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
Alzheimer's disease (AD) is usually accompanied by comorbidities such as type 2 diabetes (T2D), epilepsy, major depressive disorder (MDD), and migraine headaches (MH) that can significantly affect patient management and progression. As AD, these comorbidities have their own cumulative common genetic risk component that can be explored in a single individual through polygenic scores. Utilizing data from the UK Biobank, we investigated the correlation between polygenic scores (PGS) for these comorbidities and their actual presentation in AD patients. We show that individuals with higher PGS values showed an elevated risk of developing T2D (OR 2.1, p = 1.07 × 10-11) and epilepsy (OR 1.5, p = 0.0176). High T2D-PGS is also associated with an earlier AD onset in individuals at high genetic risk for AD (AD-PGS). In contrast, no significant genetic associations were found for MDD and MH. Our findings show distinct common genetic risk factors for T2D and epilepsy carried by AD patients that are associated with increased prevalence and earlier disease onset. These results highlight the contribution of common genetic variation to the broader clinical landscape of AD and will contribute to future tailored patient management strategies for individuals at high genetic risk.
Collapse
Affiliation(s)
- Carlos F Hernández
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile
| | - Camilo Villaman
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile
| | - Costin Leu
- Center for Neurogenetics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Dennis Lal
- Center for Neurogenetics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Cologne Center for Genomics (CCG), Medical Faculty of the University of Cologne, 50923, Köln, Germany
| | - Ignacio Mata
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrés D Klein
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile
| | - Eduardo Pérez-Palma
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile.
| |
Collapse
|
40
|
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
|
41
|
Yi F, Yuan J, Han F, Somekh J, Peleg M, Wu F, Jia Z, Zhu YC, Huang Z. Machine learning reveals connections between preclinical type 2 diabetes subtypes and brain health. Brain 2025; 148:1389-1404. [PMID: 39932872 DOI: 10.1093/brain/awaf057] [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: 07/22/2024] [Revised: 12/28/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
Previous research has established type 2 diabetes mellitus as a significant risk factor for various disorders, adversely impacting human health. While evidence increasingly links type 2 diabetes to cognitive impairment and brain disorders, understanding the causal effects of its preclinical stage on brain health is yet to be fully known. This knowledge gap hinders advancements in screening and preventing neurological and psychiatric diseases. To address this gap, we employed a robust machine learning algorithm (Subtype and Stage Inference, SuStaIn) with cross-sectional clinical data from the UK Biobank (20 277 preclinical type 2 diabetes participants and 20 277 controls) to identify underlying subtypes and stages for preclinical type 2 diabetes. Our analysis revealed one subtype distinguished by elevated circulating leptin levels and decreased leptin receptor levels, coupled with increased body mass index, diminished lipid metabolism, and heightened susceptibility to psychiatric conditions such as anxiety disorder, depression disorder, and bipolar disorder. Conversely, individuals in the second subtype manifested typical abnormalities in glucose metabolism, including rising glucose and haemoglobin A1c levels, with observed correlations with neurodegenerative disorders. A >10-year follow-up of these individuals revealed differential declines in brain health and significant clinical outcome disparities between subtypes. The first subtype exhibited faster progression and higher risk for psychiatric conditions, while the second subtype was associated with more severe progression of Alzheimer's disease and Parkinson's disease and faster progression to type 2 diabetes. Our findings highlight that monitoring and addressing the brain health needs of individuals in the preclinical stage of type 2 diabetes is imperative.
Collapse
Affiliation(s)
- Fan Yi
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310008, China
| | - Jing Yuan
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Judith Somekh
- Department of Information Systems, University of Haifa, Haifa 3303219, Israel
| | - Mor Peleg
- Department of Information Systems, University of Haifa, Haifa 3303219, Israel
| | - Fei Wu
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310008, China
| | - Zhilong Jia
- Medical Innovation Research Division of Chinese PLA General Hospital, Beijing 100853, China
| | - Yi-Cheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zhengxing Huang
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310008, China
| |
Collapse
|
42
|
Kharitonova EV, Sun Q, Ockerman F, Chen B, Zhou LY, Hysong MR, Tuftin B, Cao H, Mathias RA, Auer PL, Ober C, Raffield LM, Reiner AP, Cox NJ, Kelada SNP, Tao R, Li Y. EndoPRS: Incorporating endophenotype information to improve polygenic risk scores for clinical endpoints-A study in asthma. Am J Hum Genet 2025:S0002-9297(25)00107-7. [PMID: 40203832 DOI: 10.1016/j.ajhg.2025.03.008] [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: 12/05/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Polygenic risk score (PRS) prediction of complex diseases can be improved by leveraging related phenotypes. This has motivated the development of several multi-trait PRS methods that jointly model genetically correlated traits. However, these methods do not account for vertical pleiotropy, where one trait acts as a mediator for another. Here, we introduce endoPRS, a weighted lasso model that incorporates information from relevant endophenotypes to improve disease risk prediction without making assumptions about the genetic architecture underlying the endophenotype-disease relationship. Through extensive simulation analysis, we demonstrate the robustness of endoPRS in a variety of complex genetic frameworks. We also apply endoPRS to predict the risk of childhood-onset asthma in UK Biobank and All of Us by leveraging a paired genome-wide association study of eosinophil count, a relevant endophenotype. We find that endoPRS significantly improves prediction and transferability compared to many existing PRS methods, including multi-trait PRS methods MTAG and wMT-BLUP, which suggests advantages of endoPRS in real-life clinical settings.
Collapse
Affiliation(s)
- Elena V Kharitonova
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Computation and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Franklin Ockerman
- Department of Biostatistics, 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
| | - Laura Y Zhou
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Micah R Hysong
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bjoernar Tuftin
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongyuan Cao
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul L Auer
- Division of Biostatistics, Data Science Institute, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA 98105, USA
| | - Nancy J Cox
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Samir N P Kelada
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ran Tao
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, 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; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
43
|
Foguet C, Jiang X, Ritchie SC, Persyn E, Xu Y, Ben-Eghan C, Taylor HJ, Di Angelantonio E, Danesh J, Butterworth AS, Lambert SA, Inouye M. Metabolic reaction fluxes as amplifiers and buffers of risk alleles for coronary artery disease. Mol Syst Biol 2025:10.1038/s44320-025-00097-2. [PMID: 40175777 DOI: 10.1038/s44320-025-00097-2] [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: 09/04/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
Genome-wide association studies have identified thousands of variants associated with disease risk but the mechanism by which such variants contribute to disease remains largely unknown. Indeed, a major challenge is that variants do not act in isolation but rather in the framework of highly complex biological networks, such as the human metabolic network, which can amplify or buffer the effect of specific risk alleles on disease susceptibility. Here we use genetically predicted reaction fluxes to perform a systematic search for metabolic fluxes acting as buffers or amplifiers of coronary artery disease (CAD) risk alleles. Our analysis identifies 30 risk locus-reaction flux pairs with significant interaction on CAD susceptibility involving 18 individual reaction fluxes and 8 independent risk loci. Notably, many of these reactions are linked to processes with putative roles in the disease such as the metabolism of inflammatory mediators. In summary, this work establishes proof of concept that biochemical reaction fluxes can have non-additive effects with risk alleles and provides novel insights into the interplay between metabolism and genetic variation on disease susceptibility.
Collapse
Affiliation(s)
- Carles Foguet
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
| | - Xilin Jiang
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Scott C Ritchie
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Elodie Persyn
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Yu Xu
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Chief Ben-Eghan
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Henry J Taylor
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Science Research Centre, Fondazione Human Technopole, Milan, Italy
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Department of Human Genetics, the Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
| | - Samuel A Lambert
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
| |
Collapse
|
44
|
Huang Y, Syed MG, Chen R, Li C, Shang X, Wang W, Zhang X, Zhang X, Tang S, Liu J, Liu S, Srinivasan S, Hu Y, Mookiah MRK, Wang H, Trucco E, Yu H, Palmer C, Zhu Z, Doney ASF, He M. Genomic determinants of biological age estimated by deep learning applied to retinal images. GeroScience 2025; 47:2613-2629. [PMID: 39775603 PMCID: PMC11979078 DOI: 10.1007/s11357-024-01481-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
With the development of deep learning (DL) techniques, there has been a successful application of this approach to determine biological age from latent information contained in retinal images. Retinal age gap (RAG) defined as the difference between chronological age and predicted retinal age has been established previously to predict the age-related disease. In this study, we performed discovery genome-wide association analysis (GWAS) on the RAG using the 31,271 UK Biobank participants and replicated our findings in 8034 GoDARTS participants. The genetic correlation between RAGs predicted from the two cohorts was 0.67 (P = 0.021). After meta-analysis, we found 13 RAG loci which might be related to retinal vessel density and other aging processes. The SNP-wide heritability (h2) of RAG was 0.15. Meanwhile, by performing Mendelian randomization analysis, we found that glycated hemoglobin, inflammation hemocytes, and anemia might be associated with accelerated retinal aging. Our study explored the biological implications and molecular-level mechanism of RAG, which might enable causal inference of the aging process as well as provide potential pharmaceutical intervention targets for further treatment.
Collapse
Affiliation(s)
- Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Mohammad Ghouse Syed
- VAMPIRE Project, Computer Vision and Image Processing Group, School of Science and Engineering (Computing), University of Dundee, Dundee, DD1 9SY, UK
| | - Ruiye Chen
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia
| | - Cong Li
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xianwen Shang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xueli Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shulin Tang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jing Liu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shunming Liu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Sundar Srinivasan
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Yijun Hu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Muthu Rama Krishnan Mookiah
- VAMPIRE Project, Computer Vision and Image Processing Group, School of Science and Engineering (Computing), University of Dundee, Dundee, DD1 9SY, UK
| | - Huan Wang
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Emanuele Trucco
- VAMPIRE Project, Computer Vision and Image Processing Group, School of Science and Engineering (Computing), University of Dundee, Dundee, DD1 9SY, UK
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Colin Palmer
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Zhuoting Zhu
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia.
| | - Alexander S F Doney
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK.
| | - Mingguang He
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia.
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China.
| |
Collapse
|
45
|
Beydoun MA, Beydoun HA, Li Z, Hu YH, Noren Hooten N, Ding J, Hossain S, Maino Vieytes CA, Launer LJ, Evans MK, Zonderman AB. Alzheimer's Disease polygenic risk, the plasma proteome, and dementia incidence among UK older adults. GeroScience 2025; 47:2507-2523. [PMID: 39586964 PMCID: PMC11978584 DOI: 10.1007/s11357-024-01413-8] [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: 10/05/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex polygenic neurodegenerative disorder. Its genetic risk's relationship with all-cause dementia may be influenced by the plasma proteome. Up to 40,139 UK Biobank participants aged ≥ 50y at baseline assessment (2006-2010) were followed-up for ≤ 15 y for dementia incidence. Plasma proteomics were performed on a sub-sample of UK Biobank participants (k = 1,463 plasma proteins). AD polygenic risk scores (PRS) were used as the primary exposure and Cox proportional hazards models were conducted to examine the AD PRS-dementia relationship. A four-way decomposition model then partitioned the total effect (TE) of AD PRS on dementia into an effect due to mediation only, an effect due to interaction only, neither or both. The study found that AD PRS tertiles significantly increased the risk for all-cause dementia, particularly among women. The study specifically found that AD PRS was associated with a 79% higher risk for all-cause dementia for each unit increase (HR = 1.79, 95% CI: 1.70-1.87, P < 0.001). Eighty-six plasma proteins were significantly predicted by AD PRS, including a positive association with PLA2G7, BRK1, the glial acidic fibrillary protein (GFAP), neurofilament light chain (NfL), and negative with TREM2. Both GFAP and NfL significantly interacted synergistically with AD PRS to increase all-dementia risk (> 10% of TE is pure interaction), while GFAP was also an important consistent mediator in the AD PRS-dementia relationship. In summary, we detected significant interactions of NfL and GFAP with AD PRS, in relation to dementia incidence, suggesting potential for personalized dementia prevention and management.
Collapse
Affiliation(s)
- May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| | - Hind A Beydoun
- VA National Center On Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC, 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Jun Ding
- Translational Gerontology Branch, National Institute On Aging, NIA/NIH/IRP, Baltimore, MD, 21224, USA
| | - Sharmin Hossain
- Department of Human Services (DHS), State of Maryland, Baltimore, MD, 21202, USA
| | - Christian A Maino Vieytes
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, NIA/NIH/IRP, NIH Biomedical Research Center, National Institute On Aging Intramural Research Program, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| |
Collapse
|
46
|
Hou W, Liu Y, Hao X, Qi J, Jiang Y, Huang S, Zeng P. Relatively independent and complementary roles of family history and polygenic risk score in age at onset and incident cases of 12 common diseases. Soc Sci Med 2025; 371:117942. [PMID: 40073521 DOI: 10.1016/j.socscimed.2025.117942] [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: 10/11/2024] [Revised: 02/15/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Few studies have systematically compared the overlap and complementarity of family history (FH) and polygenic risk score (PRS) in terms of disease risk. We here investigated the impacts of FH and PRS on the risk of incident diseases or age at disease onset, as well as their clinical value in risk prediction. We analyzed 12 diseases in the prospective cohort study of UK Biobank (N = 461,220). First, restricted mean survival time analysis was performed to evaluate the influences of FH and PRS on age at onset. Then, Cox proportional hazards model was employed to estimate the effects of FH and PRS on the incident risk. Finally, prediction models were constructed to examine the clinical value of FH and PRS in the incident disease risk. Compared to negative FH, positive FH led to an earlier onset, with an average of 2.29 years earlier between the top and bottom 2.5% PRSs and high blood pressure showing the greatest difference of 6.01 years earlier. Both FH and PRS were related to higher incident risk; but they only exhibited weak interactions on high blood pressure and Alzheimer's disease/dementia, and provided relatively independent and partially complementary information on disease susceptibility, with PRS explaining 7.0% of the FH effect but FH accounting for only 1.1% of the PRS effect for incident cases. Further, FH and PRS showed additional predictive value in risk evaluation, with breast cancer showing the greatest improvement (31.3%). FH and PRS significantly affect a variety of diseases, and they are not interchangeable measures of genetic susceptibility, but instead offer largely independent and partially complementary information. Incorporating FH, PRS, and clinical risk factors simultaneously leads to the greatest predictive value for disease risk assessment.
Collapse
Affiliation(s)
- Wenyan Hou
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yuxin Liu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jike Qi
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yuchen Jiang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shuiping Huang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
47
|
Ling Y, Yuan S, Cheng H, Tan S, Huang X, Tang Y, Bai Z, Li R, Li L, Li S, Huang L, Xu A, Lyu J. Exploring the Link Between C-Reactive Protein Change and Stroke Risk: Insights From a Prospective Cohort Study and Genetic Evidence. J Am Heart Assoc 2025; 14:e038086. [PMID: 40135578 DOI: 10.1161/jaha.124.038086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 12/17/2024] [Indexed: 03/27/2025]
Abstract
BACKGROUND Previous research on how changes in CRP (C-reactive protein) levels predict stroke risk is limited. This study aimed to examine the association between CRP change and the risk of stroke and its subtypes. METHODS AND RESULTS Based on the UK Biobank data, we investigated the association between CRP change and the risk of stroke and its subtypes with Cox proportional hazards regression analysis. We further performed genetic analyses including genetic correlation, pairwise genome-wide association study, and polygenic risk score. Our study involved 14 754 participants with a median follow-up time of 10.4 years. After categorizing participants by CRP percentage change and making adjustments for potential confounders, it was observed that those with an elevated percentage of CRP change had a higher risk of any stroke (hazard ratio [HR], 1.44 [95% CI, 1.12-1.85]) and ischemic stroke (HR, 1.65 [95% CI, 1.24-2.18]). After categorization by CRP change types and adjustment for confounders, the group that became high level had a higher any-stroke risk (HR, 1.45 [95% CI, 1.04-2.02]), with the group that remained at a high level facing the greatest risk (HR, 1.74 [95% CI, 1.30-2.33]). Similar trends were observed for ischemic stroke. The group that remained at a high level also had a heightened hemorrhagic stroke risk (HR, 1.91 [95% CI, 1.07-3.44]). Genetic analysis showed a significant genetic correlation between CRP and stroke (rg, 0.257; rg_P=2.39E-07). Pairwise genome-wide association study analysis identified 5 shared genomic regions between CRP and stroke. Polygenic risk score analysis showed that participants with high stroke polygenic risk score and elevated or remaining high CRP levels have the highest risk of stroke. CONCLUSIONS Both any stroke and ischemic stroke are related to elevated and remaining high CRP levels, while hemorrhagic stroke is only related to remaining high CRP levels.
Collapse
Affiliation(s)
- Yitong Ling
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Shiqi Yuan
- Department of Neurology The Second People's Hospital of Guiyang City Guiyang Guizhou China
| | - Hongtao Cheng
- School of Nursing Jinan University Guangzhou Guangdong China
- School of Nursing Sun Yat-sen University Guangzhou China
| | - Shanyuan Tan
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Xiaxuan Huang
- Department of Anesthesiology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Yonglan Tang
- School of Nursing Jinan University Guangzhou Guangdong China
| | - Zihong Bai
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Rui Li
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Li Li
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Shuna Li
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Liying Huang
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Anding Xu
- Department of Neurology Jinan University First Affiliated Hospital Guangzhou Guangdong China
| | - Jun Lyu
- Department of Clinical Research Jinan University First Affiliated Hospital Guangzhou Guangdong China
- Key Laboratory of Regenerative Medicine of Ministry of Education Guangzhou Guangdong China
| |
Collapse
|
48
|
Kou M, Ma H, Wang X, Heianza Y, Qi L. Plasma proteomics-based brain aging signature and incident dementia risk. GeroScience 2025; 47:2335-2349. [PMID: 39532828 PMCID: PMC11978599 DOI: 10.1007/s11357-024-01407-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Investigating brain-enriched proteins with machine learning methods may enable a brain-specific understanding of brain aging and provide insights into the molecular mechanisms and pathological pathways of dementia. The study aims to analyze associations of brain-specific plasma proteomic aging signature with risks of incident dementia. In 45,429 dementia-free UK Biobank participants at baseline, we generated a brain-specific biological age using 63 brain-enriched plasma proteins with machine learning methods. The brain age gap was estimated, and Cox proportional hazards models were used to study the association with incident all-cause dementia, Alzheimer's disease (AD), and vascular dementia. Per-unit increment in the brain age gap z-score was associated with significantly higher risks of all-cause dementia (hazard ratio [95% confidence interval], 1.67 [1.56-1.79], P < 0.001), AD (1.85 [1.66-2.08], P < 0.001), and vascular dementia (1.86 [1.55-2.24], P < 0.001), respectively. Notably, 2.1% of the study population exhibited extreme old brain aging defined as brain age gap z-score > 2, correlating with over threefold increased risks of all-cause dementia and vascular dementia (3.42 [2.25-5.20], P < 0.001, and 3.41 [1.05-11.13], P = 0.042, respectively), and fourfold increased risk of AD (4.45 [2.32-8.54], P < 0.001). The associations were stronger among participants with healthier lifestyle factors (all P-interaction < 0.05). These findings were corroborated by magnetic resonance imaging assessments showing that a higher brain age gap aligns global pathophysiology of dementia, including global and regional atrophy in gray matter, and white matter lesions (P < 0.001). The brain-specific proteomic age gap is a powerful biomarker of brain aging, indicative of dementia risk and neurodegeneration.
Collapse
Affiliation(s)
- Minghao Kou
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Hao Ma
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Xuan Wang
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Yoriko Heianza
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lu Qi
- Department of Epidemiology, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
49
|
Kramer S, Su MH, Stephenson M, Rabinowitz J, Maher B, Roberson-Nay R, Castro-de-Arajuo LFS, Zhou Y, Neale MC, Gillespie N. Measuring the associations between brain morphometry and polygenic risk scores for substance use disorders in drug-naive adolescents. RESEARCH SQUARE 2025:rs.3.rs-6190536. [PMID: 40235481 PMCID: PMC11998789 DOI: 10.21203/rs.3.rs-6190536/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Substance use has been associated with differences in adult brain morphology; however, it is unclear whether these differences precede or are a result of substance use substance use. We investigated the impact of polygenic risk scores (PRSs) for cannabis use disorder (CUD) and general substance use and substance use disorder liability (SU/SUD) on brain morphology in drug-naïve adolescents. Baseline data were used from 1,874 European-descent participants (ages 9-11) comprising 222, 328 and 387 pairs of MZ twins, DZ twins, and Non-Twin Siblings, respectively, in the Adolescent Brain Cognitive Development Study. We fitted multivariate twin models to estimate the putative effects of CUD, SU/SUD, and brain region-specific PRSs. These models assessed their influence on six subcortical and two cortical phenotypes. PRS for CUD and SU/SUD were created based on GWAS conducted by Johnson et al. (2020) and Hatoum et al. (2023), respectively. When decomposing variance in each brain region of interest (ROI), we used the corresponding ROI-specific PRS. Brain morphometry in drug-naive subjects was unrelated to CUD PRS. The variance explained in each ROI by its corresponding PRS ranged from 0.8-4.4%. The SU/SUD PRS showed marginally significant effects (0.2-0.4%) on cortical surface area and nucleus accumbens volume, but overall effect sizes were small. Our findings indicate that differences in brain morphometry among baseline drug-naive individuals are not associated with the genetic risk for CUD but show a weak association with general addiction and substance use risk (SU/SUD), particularly in nucleus accumbens volume and total cortical surface area.
Collapse
|
50
|
Mekhael M, Bidaoui G, Falloon A, Pandey AC. Personalization of primary prevention: Exploring the role of coronary artery calcium and polygenic risk score in cardiovascular diseases. Trends Cardiovasc Med 2025; 35:154-163. [PMID: 39442739 DOI: 10.1016/j.tcm.2024.10.003] [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: 07/29/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Personalized healthcare is becoming increasingly popular given the vast heterogeneity in disease manifestation between individuals. Many commonly encountered diseases within cardiology are multifactorial in nature and disease progression and response is often variable due to environmental and genetic factors influencing disease states. This makes accurate early identification and primary prevention difficult in certain populations, especially young patients with limited Atherosclerotic Cardiovascular Disease (ASCVD) risk factors. Newer strategies, such as coronary artery calcium (CAC) scans and polygenic risk scores (PRS), are being implemented to aid in the detection of subclinical disease and heritable risk, respectively. Data surrounding CAC scans have shown promising results in their ability to detect subclinical atherosclerosis and predict the risk of future coronary events, especially at the extremes; however, predictive variability exists among different patient populations, limiting the test's specificity. Furthermore, relying only on CAC scores and ASCVD risk scores may fail to identify a large group of patients needing primary prevention who lack subclinical disease and traditional risk factors, but harbor genetic variabilities strongly associated with certain cardiovascular diseases. PRS can overcome these limitations. These scores can be measured in individuals as early as birth to identify genetic variants placing them at elevated risk for developing cardiovascular disease, irrespective of their current cardiovascular health status. By applying PRS alongside CAC scores, previously overlooked patient populations can be identified and begin primary prevention strategies early to achieve optimal outcomes. In this review, we expand on the current knowledge surrounding CAC scores and PRS and highlight the future possibilities of these technologies for preventive cardiology.
Collapse
Affiliation(s)
- Mario Mekhael
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ghassan Bidaoui
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Austin Falloon
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Amitabh C Pandey
- Section of Cardiology, Deming Dept of Medicine, Tulane University School of Medicine, New Orleans, LA, United States; Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States.
| |
Collapse
|