1
|
Moonla C, Khan MI, Akgonullu S, Saha T, Wang J. Touch-based uric acid sweat biosensor towards personal health and nutrition. Biosens Bioelectron 2025; 277:117289. [PMID: 39993347 DOI: 10.1016/j.bios.2025.117289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Monitoring uric acid (UA) levels is critical since elevated UA levels are associated with diverse conditions, such as gout, kidney disorders, kidney stones, hypertension, cardiovascular diseases, and metabolic syndrome. Maintaining balanced UA levels demands reliable and regular monitoring. Traditionally, such frequent UA measurements rely on blood-based UA self-testing strips. Developing sensitive and reliable noninvasive sweat-based UA sensors presents challenges, including the low UA sweat concentrations and interpersonal variations. We present here an attractive on-site UA self-testing approach utilizing a touch-enabled fingertip sweat UA electrochemical biosensor based on a uricase-enzyme electrode and sweat wicking hydrogel. This noninvasive method is rapid, simple, convenient, and painless, leveraging the high sweat rate on the fingertip at rest without any sweat stimulation. The touch-based protocol exhibits a wide linear range of UA concentrations from 10 to 1000 μM, covering normal and elevated UA sweat levels with high selectivity, reproducibility (RSD = 4.94%), good storage stability (1 week), and significant tolerance to temperature and humidity variations. The performance of the UA-touch sweat biosensor was evaluated and validated by parallel blood meter measurements by monitoring dynamically-changing sweat UA levels in healthy subjects after consuming purine-rich meals. The distinct sweat UA temporal profiles among individuals highlight the potential of the touch-based UA biosensor for personal health and nutrition. The speed and simplicity of this sweat UA assay thus encourage frequent self-testing and enhance user's compliance towards dietary interventions and lifestyle changes in connection to diverse healthcare and nutrition applications.
Collapse
Affiliation(s)
- Chochanon Moonla
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Muhammad Inam Khan
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Semra Akgonullu
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tamoghna Saha
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Joseph Wang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Chen L, Tan T, Wu Q, Cui F, Chen Y, Chen H, Zhao Y, Xiang X, Shan Z, Tang Y, Deng Q. Dietary polyunsaturated fatty acid and risk of gout: a cohort study integrating genetic predisposition and metabolomics. Eur J Epidemiol 2025:10.1007/s10654-025-01242-9. [PMID: 40426003 DOI: 10.1007/s10654-025-01242-9] [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: 02/13/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025]
Abstract
OBJECTIVES Gout is the most common inflammatory arthritis and affects quality of life. Dietary polyunsaturated fatty acids (PUFAs) have protective effects against various diseases, but its role in gout remains uncertain. Our study aims to assess the association between PUFAs intake and gout risk, the role of genetic factors, and the possible impact of metabolites. METHODS This study included 198,033 participants who were free of gout at baseline and completed at least one reliable dietary assessment in the UK Biobank. Cox proportional hazard models were used to estimate the associations between PUFAs intake and gout risk, and the modified effects of genetic predisposition. Mediation analysis also explored the mediating role of metabolic signature in associations between specific PUFAs intake and gout. RESULTS Over a median follow-up of 9.47 years, 1,708 incident cases of gout were recorded. Gout risk was significantly associated with the second quartile of linoleic acid (LA) (0.86 [0.75, 1.00]) intake and the highest quartiles of alpha-linolenic acid (ALA) (0.72 [0.62, 0.84]), total PUFA (HR: 0.84 [95% CI:0.71, 0.99]), n-6 PUFA (0.84 [0.71, 0.99]), n-3 PUFA (0.83 [0.71, 0.98]), and eicosapentaenoic acid (EPA) (0.79 [0.68, 0.91]), compared to the lowest quartiles. We observed joint effects of PUFAs intake and genetic susceptibility on gout risk. Mediation analysis showed that high-density lipoprotein (HDL) and triglycerides mediated the associations of ALA and LA with gout risk. CONCLUSION Our findings suggested the potential benefits of PUFAs in reducing gout risk, particularly vegetable sources, with HDL and triglycerides as key mediators.
Collapse
Affiliation(s)
- Li Chen
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, Hubei, China
| | - Tianqi Tan
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430015, China
| | - Qi Wu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feipeng Cui
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yashu Chen
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, Hubei, China
| | - Huimin Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Zhao
- School of Public Health, Kunming Medical University, Kunming, 650500, China
| | - Xia Xiang
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, Hubei, China
| | - Zhilei Shan
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health and MOE Key Lab of Environment and Health, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, State Key Laboratory of Environment Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Qianchun Deng
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Key Laboratory of Oilseeds Processing, Ministry of Agriculture, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan, 430062, Hubei, China.
| |
Collapse
|
3
|
Lu X, Kou H, Li C, Zhan R, Guo R, Liu S, Shen P, Shen M, Du T, Lu J, Shen X. Development and validation of an interpretable machine learning model for predicting hyperuricemia risk: Based on environmental chemical exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118392. [PMID: 40403686 DOI: 10.1016/j.ecoenv.2025.118392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/30/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
Hyperuricemia is a global health concern, with environmental chemicals as risk factors. This study used data of multiple environmental chemical exposures from the 2011-2012 cycle of the National Health and Nutrition Examination Survey (NHANES) to develop an interpretable machine learning model for hyperuricemia risk prediction. The least absolute shrinkage and selection operator (LASSO) regression method was employed to select relevant variables. The dataset was split into training (80 %) and test (20 %) sets and six machine learning models were constructed, including Random Forest (RF), Gaussian Naive Bayes (GNB), Light Gradient Boosting (LGB), Extreme Gradient Boosting (XGB), Adaptive Boosting Classifier (AB), and Support Vector Machine (SVM). Our study identified a hyperuricemia prevalence of 20.58 % in the 2011-2012 NHANES cycle, which was consistent with previous studies. The XGB model exhibited optimal performance, achieving the highest AUC (0.806, 95 % CI: 0.768-0.845), balanced accuracy (0.762; 95 % CI: 0.721-0.802), F1 value (0585; 95 % CI: 0.535-0.635), as well as the lowest Brier score (0.133; 95 % CI:0.122-0.144). Estimated glomerular filtration rate (eGFR), body mass index (BMI), cobalt (Co), mono-(2-ethyl)-hexyl phthalate (MEHP), mono-(3-carboxypropyl) phthalate (MCPP), mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), 2-hydroxynaphthalene (OHNa2) were identified as the key factors contributing to the predictive model. The results of Shapley additive explanations and partial dependence plots indicated that hyperuricemia was positively associated with MCPP, MEHHP, and OHNa2, while negatively associated with Co and MEHP. This study is the first to predict the risk of hyperuricemia based on multiple environmental chemical exposures using a machine learning model.
Collapse
Affiliation(s)
- Xiaochuan Lu
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Huawei Kou
- Medical Affairs Department of Cancer Hospital, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| | - Cong Li
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | | | - Rongrong Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Shengnan Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Peixuan Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Meiyue Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Tingwei Du
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Jiaqi Lu
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Xiaoli Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
4
|
Zhang Z, Xie Y, Bu Z, Xiang Y, Sheng W, Cao Y, Lian L, Zhang L, Qian W, Ji G. Genetically proxied glucokinase activation and risk of diabetic complications: Insights from phenome-wide and multi-omics mendelian randomization. Diabetes Res Clin Pract 2025; 225:112246. [PMID: 40374125 DOI: 10.1016/j.diabres.2025.112246] [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: 11/10/2024] [Revised: 03/31/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
AIMS This study aims to assess the benefits and adverse effects of long-term glucokinase (GK) activation from a genetic perspective. METHODS We identified genetic variants in the GCK gene associated with glycated hemoglobin (HbA1c) levels from a genome-wide association study (GWAS) involving 146,806 individuals, which served as proxies for glucokinase activation. To assess the effects and potential pathways of GK activation on a range of diabetic complications and safety outcomes, we integrated drug-target Mendelian randomization (MR), lipidome-wide and proteome-wide MR, phenome-wide MR, and colocalization analyses. RESULTS Genetically proxied GK activation was associated with reduced risks of several predefined diabetic complications, including cardiovascular diseases, stroke and diabetic retinopathy. No kidney-related benefits were observed. Safety analysis revealed a relationship between GK activation and elevated AST levels, while impaired interaction between GK and glucokinase regulatory protein (GKRP) was associated with dyslipidemia, increased liver fat content, AST, systolic blood pressure, and uric acid. Phenome-wide MR suggested that GK activation may have potential benefits for lung function and fluid intelligence score. CONCLUSIONS Our genetic evidence supports GK as a promising target for reducing the risk of specific diabetic complications. These findings require further validation through cohort studies and randomized controlled trials in patients with diabetes.
Collapse
Affiliation(s)
- Ziqi Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanxiao Xie
- Department of Respiratory Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong, China; The Ninth Clinical Medical College, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
| | - Zhenlin Bu
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China; The Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yingying Xiang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Sheng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Cao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - LeShen Lian
- Department of Respiratory Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong, China; The Ninth Clinical Medical College, Guangzhou University of Chinese Medicine, Dongguan, Guangdong, China
| | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, Shanghai, China
| | - Wei Qian
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Integration and Innovation of Classical Formula and Modern Chinese Medicine, Shanghai, China.
| |
Collapse
|
5
|
Zhang H, Liao W, Wang F, Jiang F, Ahmad F, Liu X, Hou J, Li Y, Mao Z, Zheng Z, Wang C. Trajectory of body shape in early and middle life and hyperuricemia: an observational study integrating mendelian randomization analysis. Nutr Metab Cardiovasc Dis 2025:104107. [PMID: 40425405 DOI: 10.1016/j.numecd.2025.104107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/06/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND AND AIM Although it is recognized that obesity is linked to hyperuricemia, the research on how obesity at different stages of life affects hyperuricemia is still unclear. METHODS AND RESULTS Body shape trajectory of over the first 50 years of life in Henan Rural Cohort Study was accessed by using a group-based trajectory modeling approach. Multivariate logistic regression was utilized to estimate odd ratio (OR) for hyperuricemia. Causation was further assessed using mendelian randomization (MR). Five distinct trajectories were identified and a total of 22,655 participants were enrolled for final analysis. Compared to lean-stable participants, medium-moderate increase, heavy-stable, and lean-marked increase showed significantly higher OR and 95 % confidence interval (CI) for hyperuricemia, with 1.27 (1.07, 1.50), 1.81 (1.48, 2.21) and 1.84 (1.51, 2.25) for women, 1.25 (1.01, 1.54), 1.35 (1.02, 1.77) and 1.91 (1.50, 2.43) for men. This positive association was weakened in women with high healthy lifestyle score, but the weakening effect was not significant in men. Genetically predicted birth weight, childhood body mass index (BMI), and adult BMI were significantly associated with serum uric acid (SUA), with regression coefficient (β) and 95 % CI was -0.09 (-0.14, -0.04), 0.10 (0.04, 0.16), 0.20 (0.16, 0.24), respectively. CONCLUSION Body shape trajectory is closely associated with hyperuricemia, with MR analysis suggesting potential causal links. lifelong weight management and maintaining healthy lifestyles can reduce the adverse effects of weight gain on hyperuricemia.
Collapse
Affiliation(s)
- Huanxiang Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Wei Liao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Fengling Wang
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, PR China
| | - Feng Jiang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Fayaz Ahmad
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Xiaotian Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Jian Hou
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yuqian Li
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Zhengxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Zhaohui Zheng
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China.
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China.
| |
Collapse
|
6
|
Yu J, Xu C, Ma D, Li Y, Yang L. Serum uric acid/creatinine ratio and osteoporosis in the elderly: a NHANES study. Front Med (Lausanne) 2025; 12:1530116. [PMID: 40351460 PMCID: PMC12062183 DOI: 10.3389/fmed.2025.1530116] [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: 11/28/2024] [Accepted: 04/04/2025] [Indexed: 05/14/2025] Open
Abstract
Background Osteoporosis (OP) is a metabolic bone disorder that is of significant concern to the elderly. However, few studies have investigated the correlation between the serum uric acid to creatinine ratio (UA/Cr) and OP in elderly individuals. This research seeks to examine the connection between UA/Cr levels and OP in older adults. Methods Data on participant information for the study was obtained from four cycles of the NHANES database. Multivariable logistic regression was employed to examine the correlation between UA/Cr and OP, adjusting for potential confounders such as age, gender, and race. The diagnostic efficacy of UA/Cr for OP was evaluated utilizing ROC curves. Results Multivariable logistic regression analysis showed that serum UA/Cr levels were significantly lower in individuals with OP than in those without OP. (OR = 0.83 [0.76, 0.91], P < 0.001). Subgroup analyses indicated a stronger association in men (OR = 0.77 [0.64, 0.94], P = 0.009) and women (OR = 0.85 [0.76, 0.95], P < 0.003). Furthermore, multivariable logistic regression analyses by ethnicity revealed that this association was significant solely among Non-Hispanic Whites (OR = 0.78 [0.68, 0.90], P < 0.001). The area under the ROC curve (AUC) for UA/Cr in predicting OP was higher than that for SUA alone, indicating superior predictive value. Conclusion A higher UA/Cr level within the normal range is associated with a lower risk of OP, providing insights for its diagnosis and risk assessment.
Collapse
Affiliation(s)
| | | | | | | | - Lili Yang
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Lv W, Wang X, Feng Z, Sun C, Wu H, Zeng M, Gao T, Cao K, Xu J, Zou X, Yang T, Li H, Chen L, Liu J, Dong S, Feng Z. Allantoin Serves as a Novel Risk Factor for the Progression of MASLD. Antioxidants (Basel) 2025; 14:500. [PMID: 40427382 PMCID: PMC12108491 DOI: 10.3390/antiox14050500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Uric acid (UA), traditionally recognized as an extracellular antioxidant, exhibits paradoxical associations with metabolic disorders such as metabolic dysfunction-associated steatotic liver disease (MASLD), though its mechanistic contributions remain elusive. Here, we integrate multi-modal evidence to explore the role of UA and its oxidative metabolite, allantoin, in MASLD progression. Analysis of UK Biobank data revealed a strong association between elevated UA levels and increased risks of MASLD and type 2 diabetes (T2D). However, Mendelian randomization analysis of over 2 million samples demonstrated causal effects of urate solely on serum triglycerides and T2D risk. Targeted metabolomics in an elderly Chinese cohort identified allantoin, an oxidative by-product of UA, significantly elevated in individuals with dyslipidemia or T2D, with serum allantoin levels positively correlated with fasting glucose, triglycerides, and cholesterol. Animal studies indicated that allantoin exacerbates hepatic lipid accumulation and glucose intolerance in high-fat diet mice, driven by increased hepatic lipid biogenesis and reduced bile acid production. Notably, further research revealed a strong binding affinity of allantoin for PPARα, leading to the suppression of PPARα activity, which promotes the progression of MASLD. These findings underscore the critical role of allantoin, rather than UA, as a critical driver of MASLD development, offering valuable insights for the prediction and management of hepatic metabolic disorders.
Collapse
Affiliation(s)
- Weiqiang Lv
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (W.L.); (Z.F.); (K.C.); (J.X.); (H.L.); (J.L.)
- Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (C.S.); (H.W.)
| | - Xueqiang Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266100, China; (X.W.); (M.Z.); (L.C.)
| | - Zhaode Feng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (W.L.); (Z.F.); (K.C.); (J.X.); (H.L.); (J.L.)
| | - Cunxiao Sun
- Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (C.S.); (H.W.)
| | - Hansen Wu
- Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (C.S.); (H.W.)
| | - Mengqi Zeng
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266100, China; (X.W.); (M.Z.); (L.C.)
| | - Tianlin Gao
- School of Public Health, Qingdao University, Qingdao 266071, China;
| | - Ke Cao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (W.L.); (Z.F.); (K.C.); (J.X.); (H.L.); (J.L.)
| | - Jie Xu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (W.L.); (Z.F.); (K.C.); (J.X.); (H.L.); (J.L.)
| | - Xuan Zou
- Department of Geriatrics Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- Precision Medical Institute, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Tielin Yang
- Biomedical Informatics & Genomics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (T.Y.)
| | - Hao Li
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (W.L.); (Z.F.); (K.C.); (J.X.); (H.L.); (J.L.)
| | - Lei Chen
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266100, China; (X.W.); (M.Z.); (L.C.)
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (W.L.); (Z.F.); (K.C.); (J.X.); (H.L.); (J.L.)
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266100, China; (X.W.); (M.Z.); (L.C.)
| | - Shanshan Dong
- Biomedical Informatics & Genomics Center, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (T.Y.)
| | - Zhihui Feng
- Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (C.S.); (H.W.)
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266100, China; (X.W.); (M.Z.); (L.C.)
- Interdisciplinary Research Center of Frontier Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
8
|
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: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
9
|
Fan J, Xie W, Ke H, Zhang J, Wang J, Wang H, Guo N, Bai Y, Lei X. Structural Basis for Inhibition of Urate Reabsorption in URAT1. JACS AU 2025; 5:1308-1319. [PMID: 40151250 PMCID: PMC11937972 DOI: 10.1021/jacsau.4c01188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/29/2025]
Abstract
The urate transporter 1 (URAT1) is the primary urate transporter in the kidney responsible for urate reabsorption and, therefore, is crucial for urate homeostasis. Hyperuricemia causes the common human disease gout and other pathological consequences. Inhibition of urate reabsorption through URAT1 has been shown as a promising strategy in alleviating hyperuricemia, and clinical and preclinical drug candidates targeting URAT1 are emerging. However, how small molecules inhibit URAT1 remains undefined, and the lack of accurate URAT1 complex structures hinders the development of better therapeutics. Here, we present cryoelectron microscopy structures of a humanized rat URAT1 bound with benzbromarone, lingdolinurad, and verinurad, elucidating the structural basis for drug recognition and inhibition. The three small molecules reside in the URAT1 central cavity with different binding modes, locking URAT1 in an inward-facing conformation. This study provides mechanistic insights into the drug modulation of URAT1 and sheds light on the rational design of potential URAT1-specific therapeutics for treating hyperuricemia.
Collapse
Affiliation(s)
- Junping Fan
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Wenjun Xie
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
| | - Han Ke
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jing Zhang
- Jiangsu
JITRI Molecular Engineering Inst. Co., Ltd., Jiangsu 215500, China
| | - Jin Wang
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Haijun Wang
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Nianxin Guo
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yingjie Bai
- Institute
for Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Xiaoguang Lei
- Beijing National
Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry
and Molecular Engineering of Ministry of Education, Institute of Organic
Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking-Tsinghua
Center for Life Sciences, Peking University, Beijing 100871, China
- Institute
for Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| |
Collapse
|
10
|
Ismail Umlai UK, Toor SM, Al-Sarraj YA, Mohammed S, Al Hail MSH, Ullah E, Kunji K, El-Menyar A, Gomaa M, Jayyousi A, Saad M, Qureshi N, Al Suwaidi JM, Albagha OME. A multi-ancestry genome-wide association study and evaluation of polygenic scores of LDL-C levels. J Lipid Res 2025; 66:100752. [PMID: 39909172 PMCID: PMC11927683 DOI: 10.1016/j.jlr.2025.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/12/2024] [Accepted: 02/02/2025] [Indexed: 02/07/2025] Open
Abstract
The genetic determinants of low-density lipoprotein cholesterol (LDL-C) levels in blood have been predominantly explored in European populations and remain poorly understood in Middle Eastern populations. We investigated the genetic architecture of LDL-C variation in Qatar by conducting a genome-wide association study (GWAS) on serum LDL-C levels using whole genome sequencing data of 13,701 individuals (discovery; n = 5,939, replication; n = 7,762) from the population-based Qatar Biobank (QBB) cohort. We replicated 168 previously reported loci from the largest LDL-C GWAS by the Global Lipids Genetics Consortium (GLGC), with high correlation in allele frequencies (R2 = 0.77) and moderate correlation in effect sizes (Beta; R2 = 0.53). We also performed a multi-ancestry meta-analysis with the GLGC study using MR-MEGA (Meta-Regression of Multi-Ethnic Genetic Association) and identified one novel LDL-C-associated locus; rs10939663 (SLC2A9; genomic control-corrected P = 1.25 × 10-8). Lastly, we developed Qatari-specific polygenic score (PGS) panels and tested their performance against PGS derived from other ancestries. The multi-ancestry-derived PGS (PGS000888) performed best at predicting LDL-C levels, whilst the Qatari-derived PGS showed comparable performance. Overall, we report a novel gene associated with LDL-C levels, which may be explored further to decipher its potential role in the etiopathogenesis of cardiovascular diseases. Our findings also highlight the importance of population-based genetics in developing PGS for clinical utilization.
Collapse
Affiliation(s)
- Umm-Kulthum Ismail Umlai
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Yasser A Al-Sarraj
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar; Qatar Genome Program (QGP), Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Shaban Mohammed
- Department of Pharmacy, Hamad Medical Corporation, Doha, Qatar
| | | | - Ehsan Ullah
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Khalid Kunji
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ayman El-Menyar
- Trauma and Vascular Surgery, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Mohammed Gomaa
- Adult Cardiology, Heart Hospital, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Amin Jayyousi
- Department of Diabetes, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Mohamad Saad
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Nadeem Qureshi
- Primary Care Stratified Medicine Research Group, Centre for Academic Primary Care, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Jassim M Al Suwaidi
- Adult Cardiology, Heart Hospital, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Omar M E Albagha
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar; Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
11
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [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] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
12
|
Liu H, Abedini A, Ha E, Ma Z, Sheng X, Dumoulin B, Qiu C, Aranyi T, Li S, Dittrich N, Chen HC, Tao R, Tarng DC, Hsieh FJ, Chen SA, Yang SF, Lee MY, Kwok PY, Wu JY, Chen CH, Khan A, Limdi NA, Wei WQ, Walunas TL, Karlson EW, Kenny EE, Luo Y, Kottyan L, Connolly JJ, Jarvik GP, Weng C, Shang N, Cole JB, Mercader JM, Mandla R, Majarian TD, Florez JC, Haas ME, Lotta LA, Regeneron Genetics Center, GHS-RGC DiscovEHR Collaboration, Drivas TG, Penn Medicine BioBank, Vy HMT, Nadkarni GN, Wiley LK, Wilson MP, Gignoux CR, Rasheed H, Thomas LF, Åsvold BO, Brumpton BM, Hallan SI, Hveem K, Zheng J, Hellwege JN, Zawistowski M, Zöllner S, Franceschini N, Hu H, Zhou J, Kiryluk K, Ritchie MD, Palmer M, Edwards TL, Voight BF, Hung AM, Susztak K. Kidney multiome-based genetic scorecard reveals convergent coding and regulatory variants. Science 2025; 387:eadp4753. [PMID: 39913582 PMCID: PMC12013656 DOI: 10.1126/science.adp4753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/20/2024] [Indexed: 02/17/2025]
Abstract
Kidney dysfunction is a major cause of mortality, but its genetic architecture remains elusive. In this study, we conducted a multiancestry genome-wide association study in 2.2 million individuals and identified 1026 (97 previously unknown) independent loci. Ancestry-specific analysis indicated an attenuation of newly identified signals on common variants in European ancestry populations and the power of population diversity for further discoveries. We defined genotype effects on allele-specific gene expression and regulatory circuitries in more than 700 human kidneys and 237,000 cells. We found 1363 coding variants disrupting 782 genes, with 601 genes also targeted by regulatory variants and convergence in 161 genes. Integrating 32 types of genetic information, we present the "Kidney Disease Genetic Scorecard" for prioritizing potentially causal genes, cell types, and druggable targets for kidney disease.
Collapse
Affiliation(s)
- Hongbo Liu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Kidney Innovation Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Amin Abedini
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Eunji Ha
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ziyuan Ma
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Sheng
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, Zhejiang, China
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Bernhard Dumoulin
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengxiang Qiu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Tamas Aranyi
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Molecular Life Sciences, HUN-REN Research Center for Natural Sciences, Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Shen Li
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicole Dittrich
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Der-Cherng Tarng
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Feng-Jen Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Shih-Ann Chen
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- National Chung Hsing University, Taichung, Taiwan, ROC
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Internal Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Mei-Yueh Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Internal Medicine, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan, ROC
| | - Pui-Yan Kwok
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Nita A. Limdi
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Theresa L. Walunas
- Department of Medicine, Division of General Internal Medicine and Center for Health Information Partnerships, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Eimear E. Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Genomic Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leah Kottyan
- The Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - John J. Connolly
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gail P. Jarvik
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington, Seattle, WA, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ning Shang
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Joanne B. Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Josep M. Mercader
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ravi Mandla
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine and Cardiovascular Research Institute, Cardiology Division, University of California, San Francisco, CA, USA
- Graduate Program in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy D. Majarian
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Jose C. Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary E. Haas
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Luca A. Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | - Theodore G. Drivas
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ha My T. Vy
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N. Nadkarni
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute of Digital Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura K. Wiley
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Melissa P. Wilson
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christopher R. Gignoux
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Humaira Rasheed
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Laurent F. Thomas
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn Olav Åsvold
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ben M. Brumpton
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Clinic of Thoracic and Occupational Medicine, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Stein I. Hallan
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nephrology, St. Olav’s Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Kristian Hveem
- KGJebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jie Zheng
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jacklyn N. Hellwege
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Hailong Hu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianfu Zhou
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Palmer
- Pathology and Laboratory Medicine at the Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Todd L. Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin F. Voight
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Adriana M. Hung
- Division of Nephrology and Hypertension, Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Clinical Sciences Research and Development, Nashville, TN, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Kidney Innovation Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
Collaborators
Aris Baras, Gonçalo Abecasis, Adolfo Ferrando, Giovanni Coppola, Andrew Deubler, Aris Economides, Luca A Lotta, John D Overton, Jeffrey G Reid, Alan Shuldiner, Katherine Siminovitch, Jason Portnoy, Marcus B Jones, Lyndon Mitnaul, Alison Fenney, Jonathan Marchini, Manuel Allen Revez Ferreira, Maya Ghoussaini, Mona Nafde, William Salerno, John D Overton, Christina Beechert, Erin Fuller, Laura M Cremona, Eugene Kalyuskin, Hang Du, Caitlin Forsythe, Zhenhua Gu, Kristy Guevara, Michael Lattari, Alexander Lopez, Kia Manoochehri, Prathyusha Challa, Manasi Pradhan, Raymond Reynoso, Ricardo Schiavo, Maria Sotiropoulos Padilla, Chenggu Wang, Sarah E Wolf, Hang Du, Kristy Guevara, Amelia Averitt, Nilanjana Banerjee, Dadong Li, Sameer Malhotra, Justin Mower, Mudasar Sarwar, Deepika Sharma, Sean Yu, Aaron Zhang, Muhammad Aqeel, Jeffrey G Reid, Mona Nafde, Manan Goyal, George Mitra, Sanjay Sreeram, Rouel Lanche, Vrushali Mahajan, Sai Lakshmi Vasireddy, Gisu Eom, Krishna Pawan Punuru, Sujit Gokhale, Benjamin Sultan, Pooja Mule, Eliot Austin, Xiaodong Bai, Lance Zhang, Sean O'Keeffe, Razvan Panea, Evan Edelstein, Ayesha Rasool, William Salerno, Evan K Maxwell, Boris Boutkov, Alexander Gorovits, Ju Guan, Lukas Habegger, Alicia Hawes, Olga Krasheninina, Samantha Zarate, Adam J Mansfield, Lukas Habegger, Gonçalo Abecasis, Joshua Backman, Kathy Burch, Adrian Campos, Liron Ganel, Sheila Gaynor, Benjamin Geraghty, Arkopravo Ghosh, Salvador Romero Martinez, Christopher Gillies, Lauren Gurski, Joseph Herman, Eric Jorgenson, Tyler Joseph, Michael Kessler, Jack Kosmicki, Adam Locke, Priyanka Nakka, Jonathan Marchini, Karl Landheer, Olivier Delaneau, Maya Ghoussaini, Anthony Marcketta, Joelle Mbatchou, Arden Moscati, Aditeya Pandey, Anita Pandit, Jonathan Ross, Carlo Sidore, Eli Stahl, Timothy Thornton, Sailaja Vedantam, Rujin Wang, Kuan-Han Wu, Bin Ye, Blair Zhang, Andrey Ziyatdinov, Yuxin Zou, Jingning Zhang, Kyoko Watanabe, Mira Tang, Frank Wendt, Suganthi Balasubramanian, Suying Bao, Kathie Sun, Chuanyi Zhang, Adolfo Ferrando, Giovanni Coppola, Luca A Lotta, Alan Shuldiner, Katherine Siminovitch, Brian Hobbs, Jon Silver, William Palmer, Rita Guerreiro, Amit Joshi, Antoine Baldassari, Cristen Willer, Sarah Graham, Ernst Mayerhofer, Erola Pairo Castineira, Mary Haas, Niek Verweij, George Hindy, Jonas Bovijn, Tanima De, Parsa Akbari, Luanluan Sun, Olukayode Sosina, Arthur Gilly, Peter Dornbos, Juan Rodriguez-Flores, Moeen Riaz, Manav Kapoor, Gannie Tzoneva, Momodou W Jallow, Anna Alkelai, Ariane Ayer, Veera Rajagopal, Sahar Gelfman, Vijay Kumar, Jacqueline Otto, Neelroop Parikshak, Aysegul Guvenek, Jose Bras, Silvia Alvarez, Jessie Brown, Jing He, Hossein Khiabanian, Joana Revez, Kimberly Skead, Valentina Zavala, Jae Soon Sul, Lei Chen, Sam Choi, Amy Damask, Nan Lin, Charles Paulding, Marcus B Jones, Esteban Chen, Michelle G LeBlanc, Jason Mighty, Jennifer Rico-Varela, Nirupama Nishtala, Nadia Rana, Jaimee Hernandez, Alison Fenney, Randi Schwartz, Jody Hankins, Anna Han, Samuel Hart, Ann Perez-Beals, Gina Solari, Johannie Rivera-Picart, Michelle Pagan, Sunilbe Siceron, Adam Buchanan, David J Carey, Christa L Martin, Michelle Meyer, Kyle Retterer, David Rolston, Daniel J Rader, Marylyn D Ritchie, JoEllen Weaver, Nawar Naseer, Giorgio Sirugo, Afiya Poindexter, Yi-An Ko, Kyle P Nerz, Meghan Livingstone, Fred Vadivieso, Stephanie DerOhannessian, Teo Tran, Julia Stephanowski, Salma Santos, Ned Haubein, Joseph Dunn, Anurag Verma, Colleen Morse Kripke, Marjorie Risman, Renae Judy, Colin Wollack, Shefali S Verma, Scott M Damrauer, Yuki Bradford, Scott M Dudek, Theodore G Drivas,
Collapse
|
13
|
Yao X, Cai X, Zhang S, Yang Y, Yang X, Ma W, Jiang Z. Mendelian randomization study of serum uric acid levels and urate-lowering drugs on pulmonary arterial hypertension outcomes. Sci Rep 2025; 15:4460. [PMID: 39915571 PMCID: PMC11802783 DOI: 10.1038/s41598-025-88887-4] [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] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
This study aims to explore the causal relationships between serum uric acid level and pulmonary arterial hypertension (PAH) using the Mendelian randomization (MR) approach, and to assess the therapeutic impacts of urate-lowering drugs on PAH. Utilizing published genome-wide association study (GWAS) data, we applied MR and colocalization analysis to assess the link between serum uric acid levesl and PAH across four GWAS datasets from two distinct European populations. The validity and reliability of these findings were confirmed through multiple statistical methods, along with an MR analysis of urate-lowering drug targets to investigate their potential effects on PAH treatment. MR analysis revealed a significant positive correlation between serum uric acid levels and PAH (odds ratio (OR) 1.106, 95% confidence intervals (CI) 1.021-1.200, P = 0.014), corroborated by a replication MR analysis (OR 1.859, 95% CI 1.130-3.057, P = 0.015). No significant heterogeneity or horizontal pleiotropy was found in the sensitivity analyses. However, urate-lowering drugs did not demonstrate a significant direct therapeutic effect on PAH. This study establishes a genetic basis for a causal link between serum uric acid levels and PAH. However, urate-lowering drugs do not appear to have a direct causal effect on improving PAH. These findings provide a novel reference point for developing future therapeutic strategies for PAH.
Collapse
Affiliation(s)
- Xiaoling Yao
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Xin Cai
- Department of Rheumatology and Immunology, The First People's Hospital of Guiyang, Guiyang, 550001, China
| | - Shaoqin Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Guiyang, Guiyang, 550001, China
| | - Yuzheng Yang
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Xiangyan Yang
- Department of Rheumatology and Immunology, The First People's Hospital of Guiyang, Guiyang, 550001, China
| | - Wukai Ma
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
- Institute of the Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China.
| | - Zong Jiang
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
- Institute of the Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China.
| |
Collapse
|
14
|
Chen C, Li Y, Gu Y, Zhai Q, Guo S, Xiang J, Xie Y, An M, Li C, Qin N, Shi Y, Yang L, Zhou J, Xu X, Xu Z, Wang K, Zhu M, Jiang Y, He Y, Xu J, Yin R, Chen L, Xu L, Dai J, Jin G, Hu Z, Wang C, Ma H, Shen H. Massively parallel variant-to-function mapping determines functional regulatory variants of non-small cell lung cancer. Nat Commun 2025; 16:1391. [PMID: 39910069 PMCID: PMC11799298 DOI: 10.1038/s41467-025-56725-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: 05/01/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
Genome-wide association studies have identified thousands of genetic variants associated with non-small cell lung cancer (NSCLC), however, it is still challenging to determine the causal variants and to improve disease risk prediction. Here, we applied massively parallel reporter assays to perform NSCLC variant-to-function mapping at scale. A total of 1249 candidate variants were evaluated, and 30 potential causal variants within 12 loci were identified. Accordingly, we proposed three genetic architectures underlying NSCLC susceptibility: multiple causal variants in a single haplotype block (e.g. 4q22.1), multiple causal variants in multiple haplotype blocks (e.g. 5p15.33), and a single causal variant (e.g. 20q11.23). We developed a modified polygenic risk score using the potential causal variants from Chinese populations, improving the performance of risk prediction in 450,821 Europeans from the UK Biobank. Our findings not only augment the understanding of the genetic architecture underlying NSCLC susceptibility but also provide strategy to advance NSCLC risk stratification.
Collapse
Affiliation(s)
- Congcong Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China
| | - Yang Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yayun Gu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Qiqi Zhai
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211116, Jiangsu, China
| | - Songwei Guo
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211116, Jiangsu, China
| | - Jun Xiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yuan Xie
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211116, Jiangsu, China
| | - Mingxing An
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Chenmeijie Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Na Qin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yanan Shi
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211116, Jiangsu, China
| | - Liu Yang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jun Zhou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xianfeng Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ziye Xu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211116, Jiangsu, China
| | - Kai Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yue Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yuanlin He
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Rong Yin
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Department of Thoracic Surgery Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210029, Jiangsu, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Lin Xu
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Department of Thoracic Surgery Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210029, Jiangsu, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- State Key Laboratory of Reproductive Medicine (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, Jiangsu, China
| | - Cheng Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
15
|
Chang H, Tao Q, Wei L, Wang Y, Tu C. Spatiotemporal landscape of kidney in a mouse model of hyperuricemia at single-cell level. FASEB J 2025; 39:e70292. [PMID: 39817712 PMCID: PMC11737292 DOI: 10.1096/fj.202401801rr] [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: 08/04/2024] [Revised: 11/16/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025]
Abstract
Serum uric acid is an end-product of purine metabolism. Uric acid concentrations in excess of the physiological range may lead to diseases such as gout, cardiovascular disease, and kidney injury. The kidney includes a variety of cell types with specialized functions such as fluid and electrolyte homeostasis, detoxification, and endocrine functions. Two-thirds of uric acid is excreted through kidney, however, the exploration of markers and new therapeutic targets in renal tissue of hyperuricemia is still lacking. Single-cell and spatial omics techniques represent major milestones in life sciences. The combined measurement of the physical structure and molecular characteristics of tissues facilitates the exploration of the pathophysiological processes underlying disease development and the discovery of possible therapeutic targets. Here, the spatiotemporal atlas of hyperuricemic nephropathy was investigated using single-cell RNA sequencing, spatial transcriptomics, spatial proteomics, and spatial metabolomics in a urate oxidase knockout mouse model. Several emerging targets and pathways especially ribosome and metabolism related to uric acid excretion were discovered and will be investigated further in studies on lowering uric acid.
Collapse
Affiliation(s)
- Haining Chang
- Department of Internal MedicineThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
| | - Qianru Tao
- Department of Internal MedicineThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of NephrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
| | - Lan Wei
- Department of Internal MedicineThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
| | - Yangyang Wang
- Department of Clinical LaboratoryThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
| | - Chao Tu
- Department of Internal MedicineThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
| |
Collapse
|
16
|
Liu M, Yin N, Zhu Y, Du A, Cai C, Leng P. Associations between lipid-lowering drugs and urate and gout outcomes: a Mendelian randomization study. Front Endocrinol (Lausanne) 2025; 15:1398023. [PMID: 39926389 PMCID: PMC11802419 DOI: 10.3389/fendo.2024.1398023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 12/23/2024] [Indexed: 02/11/2025] Open
Abstract
Objective Gout is a common inflammatory arthritis and lipid metabolism plays a crucial role in urate and gout. Potential associations between urate and gout and lipid-lowering drugs have been revealed in observational studies. However, the effects of lipid-lowering drugs on urate and gout remain controversial. The aim of this study was to investigate the genetic association between lipid-lowering drugs and urate and gout. Methods In this study, two genetic proxies were employed to approximate lipid-lowering drug exposure: expression quantitative trait loci (eQTL) associated with drug-target genes and genetic variations proximal to or within genes targeted by these drugs, which are linked to low-density lipoprotein cholesterol (LDL-C) levels. The study's exposures encompassed genetic variants within drug target genes (HMGCR, PCSK9, NPC1L1), each representing distinct lipid-lowering mechanisms. Causal effects were estimated using the inverse variance weighting (IVW) method, while the Summary Data-based Mendelian Randomization (SMR) method, leveraging pooled data, was applied to compute effect estimates. These estimates were further refined through various approaches including MR-Egger, the weighted median method, simple and weighted models, and leave-one-out analyses to conduct sensitivity analyses. Result The analytical outcomes utilizing the IVW method indicated that inhibitors of HMGCR were correlated with an elevated risk of developing gout (IVW: OR [95%CI] = 1.25 [1.03, 1.46], p=0.0436), while PCSK9 inhibitors were linked to heightened levels of urate (IVW: OR [95%CI] = 1.06 [1.01,1.10], p=0.0167). Conversely, no significant correlation between NPC1L1 inhibitors and the levels of urate or the risk of gout was established. Furthermore, the SMR analysis failed to identify significant associations between the expression levels of the HMGCR, PCSK9, and NPC1L1 genes and the risk of gout or elevated urate levels (SMR method: all P values >0.05). Sensitivity analyses further confirmed the robustness of these results, with no significant heterogeneity or pleiotropy found. Conclusion This study furnishes novel causal evidence supporting the potential genetic correlation between the use of lipid-lowering drugs and the incidence of gout as well as urate levels. The findings indicate that inhibitors targeting HMGCR may elevate the risk associated with the development of gout, while inhibitors targeting PCSK9 are likely to increase urate concentrations.
Collapse
Affiliation(s)
- Min Liu
- Department of Orthopaedics, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Na Yin
- College of Nursing, Hangzhou Normal University, Hangzhou, China
| | - Yuhang Zhu
- Department of Orthopaedics, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Aili Du
- Department of Orthopaedics, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunyuan Cai
- Department of Orthopaedics, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pengyuan Leng
- Department of Orthopaedics, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Fu C, Liu B, Chen W, Qiu Y, Zheng C, Mao Y, Yin Z, Ye D. Association between serum iron status and gout: results from the NHANES and Mendelian randomization study. Food Funct 2025; 16:707-719. [PMID: 39745203 DOI: 10.1039/d4fo00294f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Aims. Previous observational studies have provided inconsistent evidence for the association between serum iron status and the risk of gout. Moreover, it remains uncertain whether the observed association is causal or due to confounding or reverse causality. This research aimed to investigate the association of serum iron status indicators with the risk of gout and to further examine the causal relationship by the Mendelian randomization (MR) method. Methods. We first conducted a cross-sectional study from the National Health and Nutrition Examination Survey 2017-2018, including a total of 4635 participants. The association of serum iron status indicators with gout risk was evaluated using a multivariable logistic regression model. Furthermore, a two-sample MR study using genetic data from large-scale genome-wide association studies of serum iron status indicators (246 139 individuals) and gout (discovery: 13 179 cases and 75 0634 controls; replication: 5292 cases and 368 788 controls; 2115 cases and 67 259 controls) was conducted to infer causality. Inverse-variance-weighting (IVW) was applied as the main method of MR analysis. A series of sensitivity analyses were used to evaluate the robustness of their relationship. Results. In the cross-sectional study, there was no significant relationship between serum iron status indicators and gout risk. However, IVW results showed that genetically predicted serum iron and transferrin saturation (TSAT) were significantly associated with the increased risk of gout in the discovery analysis [odds ratio (OR): 1.21; 95% confidence interval (CI): 1.10-1.32; P = 9.80 × 10-5 for serum iron and OR: 1.16; 95% CI: 1.08-1.25; P = 7.14 × 10-5 for TSAT]. The replication analysis provided similar results compared with the discovery analysis. Conclusion. Our study provides support for potential causal associations between serum iron and the altered risk of gout. Further investigations are warranted to elucidate the biological processes through which iron influences susceptibility to gout.
Collapse
Affiliation(s)
- Canya Fu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China.
- Department of Immunity, Quzhou Center for Disease Control and Prevention, Quzhou, China.
| | - Bin Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Weiwei Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yu Qiu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Canjie Zheng
- Department of Immunity, Quzhou Center for Disease Control and Prevention, Quzhou, China.
| | - Yingying Mao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Zhiying Yin
- Department of Immunity, Quzhou Center for Disease Control and Prevention, Quzhou, China.
| | - Ding Ye
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
18
|
Kim H, Do H, Son CN, Jang JW, Choi SS, Moon KW. Effects of Genetic Risk and Lifestyle Habits on Gout: A Korean Cohort Study. J Korean Med Sci 2025; 40:e1. [PMID: 39807002 PMCID: PMC11729237 DOI: 10.3346/jkms.2025.40.e1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/19/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Gout is a type of inflammatory arthritis caused by monosodium urate crystal deposits, and the prevalence of this condition has been increasing. This study aimed to determine the combined effects of genetic risk factors and lifestyle habits on gout, using data from a Korean cohort study. Identifying high-risk individuals in advance can help prevent gout and its associated disorders. METHODS We analyzed data from the Korean Genome and Epidemiology Study-Urban Health Examinees cohort (KoGES-HEXA). Genetic information of the participants was collected at baseline, and gout cases were identified based on patient statements. The polygenic risk score (PRS) was calculated using nine independent genome-wide association study datasets, and lifestyle factors and metabolic syndrome status were measured for each participant using the KoGES. Logistic regression models were used to estimate the odds ratios (ORs) for gout in relation to genetic risk, lifestyle habits, and metabolic health status, after adjusting for age and sex. RESULTS Among 44,605 participants, 617 were diagnosed with gout. Gout was associated with older age, higher body mass index, and higher prevalence of hypertension, diabetes, and hypertriglyceridemia. High PRS, unfavorable lifestyle habits, and poor metabolic profiles were significantly associated with an increased risk of gout. Compared with that in the low-genetic-risk and healthy lifestyle group or ideal metabolic profile group, the risk of gout was increased in the high-genetic-risk plus unfavorable lifestyle (OR, 3.64; 95% confidence interval [CI], 2.32-6.03) or poor metabolic profile (OR, 7.78; 95% CI, 4.61-13.40) group. Conversely, adherence to favorable lifestyle habits significantly reduced gout risk, especially in high-genetic-risk groups. CONCLUSION Genetic predisposition and unhealthy lifestyle habits significantly increase the risk of gout. Promoting healthy lifestyle habits is crucial to prevent the development of gout, particularly in individuals with high genetic susceptibility.
Collapse
Affiliation(s)
- Hyunjung Kim
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Korea
| | - Hyunsue Do
- Division of Rheumatology, Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Chang-Nam Son
- Eulji Rheumatology Research Institute, Eulji University School of Medicine, Uijeongbu, Korea
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University School of Medicine, Chuncheon, Korea
- Department of Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
| | - Sun Shim Choi
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Korea.
| | - Ki Won Moon
- Division of Rheumatology, Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
- Department of Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea.
| |
Collapse
|
19
|
Ahn EY, So MW. The pathogenesis of gout. JOURNAL OF RHEUMATIC DISEASES 2025; 32:8-16. [PMID: 39712248 PMCID: PMC11659655 DOI: 10.4078/jrd.2024.0054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/19/2024] [Accepted: 10/24/2024] [Indexed: 12/24/2024]
Abstract
Gout is the most common inflammatory arthritis in adults, associated with hyperuricemia and the chronic deposition of monosodium urate (MSU) crystals. Hyperuricemia results from increased production of uric acid and decreased excretion by the kidneys and intestines. Urate excretion is regulated by a group of urate transporters, and decreased renal or intestinal excretion is the primary mechanism of hyperuricemia in most people. Genetic variability in these urate transporters is strongly related to variances in serum urate levels. Not all individuals with hyperuricemia show deposition of MSU crystals or develop gout. The initiation of the inflammatory response to MSU crystals is mainly mediated by the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing protein 3 (NLRP3) inflammasome. The activated NLRP3 inflammasome complex cleaves pro-interleukin-1β (IL-1β) into its active form, IL-1β, which is a key mediator of the inflammatory response in gout. IL-1β leads to the upregulation of cytokines and chemokines, resulting in the recruitment of neutrophils and other immune cells. Neutrophils recruited to the site of inflammation also play a role in resolving inflammation. Aggregated neutrophil extracellular traps (NETs) trap and degrade cytokines and chemokines through NET-bound proteases, promoting the resolution of inflammation. Advanced gout is characterized by tophi, chronic inflammatory responses, and structural joint damage. Tophi are chronic foreign body granuloma-like structures containing collections of MSU crystals encased by inflammatory cells and connective tissue. Tophi are closely related to chronic inflammation and structural damage.
Collapse
Affiliation(s)
- Eun Young Ahn
- Division of Rheumatology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Min Wook So
- Division of Rheumatology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
20
|
Ji A, Sui Y, Xue X, Ji X, Shi W, Shi Y, Terkeltaub R, Dalbeth N, Takei R, Yan F, Sun M, Li M, Lu J, Cui L, Liu Z, Wang C, Li X, Han L, Fang Z, Sun W, Liang Y, He Y, Zheng G, Wang X, Wang J, Zhang H, Pang L, Qi H, Li Y, Cheng Z, Li Z, Xiao J, Zeng C, Merriman TR, Qu H, Fang X, Li C. Novel Genetic Loci in Early-Onset Gout Derived From Whole-Genome Sequencing of an Adolescent Gout Cohort. Arthritis Rheumatol 2025; 77:107-115. [PMID: 39118347 DOI: 10.1002/art.42969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Mechanisms underlying the adolescent-onset and early-onset gout are unclear. This study aimed to discover variants associated with early-onset gout. METHODS We conducted whole-genome sequencing in a discovery adolescent-onset gout cohort of 905 individuals (gout onset 12 to 19 years) to discover common and low-frequency single-nucleotide variants (SNVs) associated with gout. Candidate common SNVs were genotyped in an early-onset gout cohort of 2,834 individuals (gout onset ≤30 years old), and meta-analysis was performed with the discovery and replication cohorts to identify loci associated with early-onset gout. Transcriptome and epigenomic analyses, quantitative real-time polymerase chain reaction and RNA sequencing in human peripheral blood leukocytes, and knock-down experiments in human THP-1 macrophage cells investigated the regulation and function of candidate gene RCOR1. RESULTS In addition to ABCG2, a urate transporter previously linked to pediatric-onset and early-onset gout, we identified two novel loci (Pmeta < 5.0 × 10-8): rs12887440 (RCOR1) and rs35213808 (FSTL5-MIR4454). Additionally, we found associations at ABCG2 and SLC22A12 that were driven by low-frequency SNVs. SNVs in RCOR1 were linked to elevated blood leukocyte messenger RNA levels. THP-1 macrophage culture studies revealed the potential of decreased RCOR1 to suppress gouty inflammation. CONCLUSION This is the first comprehensive genetic characterization of adolescent-onset gout. The identified risk loci of early-onset gout mediate inflammatory responsiveness to crystals that could mediate gouty arthritis. This study will contribute to risk prediction and therapeutic interventions to prevent adolescent-onset gout.
Collapse
Affiliation(s)
- Aichang Ji
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Sui
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomei Xue
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiapeng Ji
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenrui Shi
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Yongyong Shi
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | | | | | - Riku Takei
- Asia Pacific Gout Consortium and University of Alabama at Birmingham
| | - Fei Yan
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingshu Sun
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Maichao Li
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jie Lu
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingling Cui
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhen Liu
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Can Wang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinde Li
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Han
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhanjie Fang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Wenyan Sun
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue Liang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Yuwei He
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guangmin Zheng
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Wang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiayi Wang
- Development Center for Medical Science & Technology, National Health Commission of the People's Republic of China, Beijing, China
| | - Hui Zhang
- Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Lei Pang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Han Qi
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yushuang Li
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zan Cheng
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhiqiang Li
- The Biomedical Sciences Institute and The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Jingfa Xiao
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Changqing Zeng
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Tony R Merriman
- Asia Pacific Gout Consortium, University of Alabama at Birmingham, Institute of Metabolic Diseases, Qingdao University, Qingdao, China, and University of Otago, Dunedin, New Zealand
| | - Hongzhu Qu
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, and Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Xiangdong Fang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, and Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Changgui Li
- The Affiliated Hospital of Qingdao University, Qingdao, China, Asia Pacific Gout Consortium, and Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| |
Collapse
|
21
|
Stiburkova B, Lukesova M, Zeman J. Pediatrics hyperuricemia in clinical practice: A retrospective analysis in 1753 children and adolescents with hyperuricemia. Joint Bone Spine 2025; 92:105796. [PMID: 39490564 DOI: 10.1016/j.jbspin.2024.105796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVES Serum levels of uric acid (S-UA) are influenced by the interaction of genetic and environmental factors; detailed studies of hyperuricemia in children are rare. This retrospective study aimed to analyze the causes, risk factors, and therapeutic approaches associated with the development of hyperuricemia in childhood. METHODS In a single-center study, serum uric acid levels were analyzed in 33,900 samples from 13,890 children and adolescents<19 years (6760 girls and 7130 boys) obtained between 2013 and 2023. Hyperuricemia was defined as S-UA>370μmol/L (6.22mg/dL) in girls and>420μmol/L (7.06mg/dL) in boys; mild hyperuricemia was defined as 370-420μmol/L in boys<13 years. RESULTS In the analyzed group, hyperuricemia was found in 1753 patients (12.6%), including 586 girls and 864 boys; mild hyperuricemia was found in 303 boys<13 years. The most common associated conditions were obesity with body mass index>95th percentile (27.8% of girls, 26.3% of boys) and chronic kidney disease (18.6% of boys, 11.4% of girls). Hyperuricemia was also relatively common in children with connective tissue disorders (10.6%) or different inherited metabolic disorders (10.7%). Transitory hyperuricemia was found in 19.1% of girls and 10.1% of boys with acute gastroenteritis. Urate-lowering therapy was used in 73 children and adolescents with severe hyperuricemia (S-UA 556±107μmol/L, fraction excretion of UA 3.27±1.98%). Eight treated children had chronic kidney disease, nine were extremely obese, one had combined antiepileptic therapy, and 55 had inherited metabolic diseases, including 26 children with disorders of purine metabolism. The initial daily dose of allopurinol (50-100mg) normalized the S-UA (350±80μmol/L) in a majority of children, except for extremely obese adolescents (weight 98-149kg) where the dose had to be increased to 200-300mg. CONCLUSIONS Asymptomatic hyperuricemia is a relatively common biochemical finding in pediatric clinical practice. The etiology of hyperuricemia should be carefully analyzed, and the value of individualized hyperuricemia management and the eventual benefits of urate-lowering therapy in children must be carefully considered.
Collapse
Affiliation(s)
- Blanka Stiburkova
- Institute of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Marketa Lukesova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jiri Zeman
- Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
22
|
Flanagan J, Liu X, Ortega-Reyes D, Tomizuka K, Matoba N, Akiyama M, Koido M, Ishigaki K, Ashikawa K, Takata S, Shi M, Aoi T, Momozawa Y, Ito K, Murakami Y, Matsuda K, Kamatani Y, Morris AP, Horikoshi M, Terao C. Population-specific reference panel improves imputation quality for genome-wide association studies conducted on the Japanese population. Commun Biol 2024; 7:1665. [PMID: 39702642 DOI: 10.1038/s42003-024-07338-4] [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: 07/22/2023] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
To improve imputation quality for genome-wide association studies (GWAS) conducted on the Japanese population, we developed and evaluated four Japanese population-specific reference panels. These panels were constructed through the augmentation of the 1000 Genomes Project (1KG) panel using Japanese whole genome sequencing (WGS) data, with sample sizes ranging from 1 K to 7 K individuals enrolled through the Biobank Japan (BBJ) project, and sequencing depths ranging from 3× to 30×. Among these panels, an augmented reference panel comprising 7472 WGS samples of mixed depth (1KG+7K) exhibit the greatest improvement in imputation quality relative to the Trans-Omics for Precision Medicine (TOPMed) reference panel. Notably, we observe these improvements primarily for rare variants with a minor allele frequency (MAF) <5%. To demonstrate the benefits of improved imputation quality in association analyses of complex traits, we conducted GWAS for serum uric acid and total cholesterol levels following imputation up to the 1KG+7K panel. The analysis reveals several loci reaching genome-wide significance (P < 5 × 10-8) in the 1KG+7K imputation output yet remaining undetected when the same sample set is imputed up to the TOPMed reference panel. In summary, the 1KG+7K panel demonstrates significant advantages in the discovery of trait-associated loci, particularly those influenced by low-frequency association signals.
Collapse
Affiliation(s)
- Jack Flanagan
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Graduate School of Data Science, Seoul National University, Seoul, Republic of Korea
| | - Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - David Ortega-Reyes
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for DNA Data Analysis, National Institute of Genetics, Shizuoka, Japan
- Department of Genetics, School of Life Science, The Graduate University for Advanced Studies, SOKENDAI, Kanagawa, Japan
| | - Kohei Tomizuka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nana Matoba
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Genetics, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Masato Akiyama
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Koido
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kyota Ashikawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Sadaaki Takata
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - MingYang Shi
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomomi Aoi
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, UK
| | - Momoko Horikoshi
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan.
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
23
|
Feng GJ, Xu Q, Zhao QG, Han BX, Yan SS, Zhu J, Pei YF. The genetic architecture of age at menarche and its causal effects on other traits. J Hum Genet 2024; 69:645-653. [PMID: 39147824 DOI: 10.1038/s10038-024-01287-w] [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: 12/21/2023] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 08/17/2024]
Abstract
Age at menarche (AAM) is a sign of puberty of females. It is a heritable trait associated with various adult diseases. However, the genetic mechanism that determines AAM and links it to disease risk is poorly understood. Aiming to uncover the genetic basis for AAM, we conducted a joint association study in up to 438,089 women from 3 genome-wide association studies of European and East Asian ancestries. A series of bioinformatical analyses and causal inference were then followed to explore in-depth annotations at the associated loci and infer the causal relationship between AAM and other complex traits/diseases. This largest meta-analysis identified a total of 21 novel AAM associated loci at the genome wide significance level (P < 5.0 × 10-8), 4 of which were European ancestry-specific loci. Functional annotations prioritized 33 candidate genes at newly identified loci. Significant genetic correlations were observed between AAM and 67 complex traits. Further causal inference demonstrated the effects of AAM on 13 traits, including forced vital capacity (FVC), high blood pressure, age at first live birth, etc, indicating that earlier AAM causes lower FVC, worse lung function, hypertension and earlier age at first (last) live birth. Enrichment analysis identified 5 enriched tissues, including the hypothalamus middle, hypothalamo hypophyseal system, neurosecretory systems, hypothalamus and retina. Our findings may provide useful insights that elucidate the mechanisms determining AAM and the genetic interplay between AAM and some traits of women.
Collapse
Affiliation(s)
- Gui-Juan Feng
- The First People's Hospital of Lianyungang, Jiangsu, PR China
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Qian Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Qi-Gang Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Bai-Xue Han
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Shan-Shan Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Jie Zhu
- Department of Gynaecology and Obstetrics, Suzhou Ninth Hospital Affiliated to Soochow University, 2666 Lu‑dang Rd., Wujiang District, Suzhou, 215200, Jiangsu, China.
| | - Yu-Fang Pei
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China.
| |
Collapse
|
24
|
McCormick N, Joshi AD, Yokose C, Yu B, Tin A, Terkeltaub R, Merriman TR, Zeleznik O, Eliassen AH, Curhan GC, Ea HK, Nayor M, Raffield LM, Choi HK. Prediagnostic Amino Acid Metabolites and Risk of Gout, Accounting for Serum Urate: Prospective Cohort Study and Mendelian Randomization. Arthritis Care Res (Hoboken) 2024; 76:1666-1674. [PMID: 39169570 PMCID: PMC11711019 DOI: 10.1002/acr.25420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/05/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVE Our objective was to prospectively investigate prediagnostic population-based metabolome for risk of hospitalized gout (ie, most accurate, severe, and costly cases), accounting for serum urate. METHODS We conducted prediagnostic metabolome-wide analyses among 249,677 UK Biobank participants with nuclear magnetic resonance metabolomic profiling (N = 168 metabolites, including eight amino acids) from baseline blood samples (2006-2010) without a history of gout. We calculated multivariable hazard ratios (HRs) for hospitalized incident gout, before and after adjusting for serum urate levels; we included patients with nonhospitalized incident gout in a sensitivity analysis. Potential causal effects were evaluated with two-sample Mendelian randomization. RESULTS Correcting for multiple testing, 107 metabolites were associated with incidence of hospitalized gout (n = 2,735) before urate adjustment, including glycine and glutamine (glutamine HR 0.64, 95% confidence interval [CI] 0.54-0.75, P = 8.3 × 10-8; glycine HR 0.69, 95% CI 0.61-0.78, P = 3.3 × 10-9 between extreme quintiles), and glycoprotein acetyls (HR 2.48, 95% CI 2.15-2.87, P = 1.96 × 10-34). Associations remained significant and directionally consistent following urate adjustment (HR 0.83, 95% CI 0.70-0.98; HR 0.86, 95% CI 0.76-0.98; HR 1.41, 95% CI 1.21-1.63 between extreme quintiles), respectively; corresponding HRs per SD were 0.91 (95% CI 0.86-0.97), 0.94 (95% CI 0.91-0.98), and 1.10 (95% CI 1.06-1.14). Findings persisted when including patients with nonhospitalized incident gout. Mendelian randomization corroborated their potential causal role on hyperuricemia or gout risk; with change in urate levels of -0.05 mg/dL (95% CI -0.08 to -0.01) and -0.12 mg/dL (95% CI -0.22 to -0.03) per SD of glycine and glutamine, respectively, and odds ratios of 0.94 (95% CI 0.88-1.00) and 0.81 (95% CI 0.67-0.97) for gout. CONCLUSION These prospective findings with causal implications could lead to biomarker-based risk prediction and potential supplementation-based interventions with glycine or glutamine.
Collapse
Affiliation(s)
- Natalie McCormick
- Massachusetts General Hospital, Boston MA USA
- Harvard Medical School, Boston MA USA
- Arthritis Research Canada, Vancouver BC Canada
| | | | - Chio Yokose
- Massachusetts General Hospital, Boston MA USA
- Harvard Medical School, Boston MA USA
| | - Bing Yu
- The University of Texas Health Science Center at Houston, Houston TX USA
| | - Adrienne Tin
- University of Mississippi Medical Center, Jackson MS USA
| | | | - Tony R. Merriman
- University of Alabama at Birmingham, Birmingham AL USA
- University of Otago, Dunedin, New Zealand
| | - Oana Zeleznik
- Harvard Medical School, Boston MA USA
- Brigham and Women’s Hospital, Boston MA USA
| | - A. Heather Eliassen
- Brigham and Women’s Hospital, Boston MA USA
- Harvard TH Chan School of Public Health, Boston MA USA
| | - Gary C. Curhan
- Harvard Medical School, Boston MA USA
- Brigham and Women’s Hospital, Boston MA USA
| | | | | | | | - Hyon K. Choi
- Massachusetts General Hospital, Boston MA USA
- Harvard Medical School, Boston MA USA
- Arthritis Research Canada, Vancouver BC Canada
| |
Collapse
|
25
|
Shine BK, Choi JE, Park YJ, Hong KW. The Genetic Variants Influencing Hypertension Prevalence Based on the Risk of Insulin Resistance as Assessed Using the Metabolic Score for Insulin Resistance (METS-IR). Int J Mol Sci 2024; 25:12690. [PMID: 39684400 DOI: 10.3390/ijms252312690] [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: 10/21/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Insulin resistance is a major indicator of cardiovascular diseases, including hypertension. The Metabolic Score for Insulin Resistance (METS-IR) offers a simplified and cost-effective way to evaluate insulin resistance. This study aimed to identify genetic variants associated with the prevalence of hypertension stratified by METS-IR score levels. Data from the Korean Genome and Epidemiology Study (KoGES) were analyzed. The METS-IR was calculated using the following formula: ln [(2 × fasting blood glucose (FBG) + triglycerides (TG)) × body mass index (BMI)]/ ln [high-density lipoprotein cholesterol (HDL-C)]. The participants were divided into tertiles 1 (T1) and 3 (T3) based on their METS-IR scores. Genome-wide association studies (GWAS) were performed for hypertensive cases and non-hypertensive controls within these tertile groups using logistic regression adjusted for age, sex, and lifestyle factors. Among the METS-IR tertile groups, 3517 of the 19,774 participants (17.8%) at T1 had hypertension, whereas 8653 of the 20,374 participants (42.5%) at T3 had hypertension. A total of 113 single-nucleotide polymorphisms (SNPs) reached the GWAS significance threshold (p < 5 × 10-8) in at least one tertile group, mapping to six distinct genetic loci. Notably, four loci, rs11899121 (chr2p24), rs7556898 (chr2q24.3), rs17249754 (ATP2B1), and rs1980854 (chr20p12.2), were significantly associated with hypertension in the high-METS-score group (T3). rs10857147 (FGF5) was significant in both the T1 and T3 groups, whereas rs671 (ALDH2) was significant only in the T1 group. The GWASs identified six genetic loci significantly associated with hypertension, with distinct patterns across METS-IR tertiles, highlighting the role of metabolic context in genetic susceptibility. These findings underscore critical genetic factors influencing hypertension prevalence and provide insights into the metabolic-genetic interplay underlying this condition.
Collapse
Affiliation(s)
- Bo-Kyung Shine
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Ja-Eun Choi
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| | - Young-Jin Park
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Kyung-Won Hong
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| |
Collapse
|
26
|
Nakayama A, Kawamura Y, Nakatochi M, Toyoda Y, Nakajima M, Maehara K, Kirihara M, Shimizu S, Matsuo K, Matsuo H. Strong genetic effect on gout revealed by genetic risk score from meta-analysis of two genome-wide association studies. Hum Cell 2024; 38:16. [PMID: 39527284 PMCID: PMC11554751 DOI: 10.1007/s13577-024-01138-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/14/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Mayuko Nakajima
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Kazuki Maehara
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Mana Kirihara
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan.
- Department of Biomedical Information Management, National Defense Medical College Research Institute, National Defense Medical College, Tokorozawa, Japan.
| |
Collapse
|
27
|
Tan Y, Chen Y, Wang T, Li J. Serum uric acid and pulmonary arterial hypertension: A two-sample Mendelian randomization study. Heart Lung 2024; 68:337-341. [PMID: 39236651 DOI: 10.1016/j.hrtlng.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Observational studies have suggested a correlation between hyperuricemia and pulmonary arterial hypertension (PAH), yet the causal relationship remains uncertain. We aimed to establish this link using Mendelian Randomization (MR) methods. OBJECTIVES Based on publicly accessible data, our study employs MR to determine the causal relationship between uric acid (UA) and PAH. METHOD MR analysis was conducted among individuals of European descent. Genetic instruments linked to UA (p-value < 5 × 10-8) were extracted from the Chronic Kidney Disease Genetic Consortium and genome-wide association study databases. PAH risk genetic associations were sourced separately. We employed four MR methods (MR-Egger, weighted median, inverse variance weighted, and weighted mode) with selected instrumental variables to assess the causal association between UA and PAH. MR-PRESSO was used to evaluate pleiotropy and outlier Single Nucleotide Polymorphisms (SNPs), while Cochran's Q test and funnel plot assessed SNP heterogeneity. Leave-one-out analysis examined SNP impacts on causal assessment. RESULT Two-sample MR analysis revealed a positive, causal relationship between UA levels and PAH. CONCLUSION Our MR analysis provides robust evidence of a causal link between serum UA and PAH, suggesting UA's potential as a biomarker and therapeutic target for PAH.
Collapse
Affiliation(s)
- Yingjie Tan
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yusi Chen
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tianyu Wang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiang Li
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
28
|
Major TJ, Takei R, Matsuo H, Leask MP, Sumpter NA, Topless RK, Shirai Y, Wang W, Cadzow MJ, Phipps-Green AJ, Li Z, Ji A, Merriman ME, Morice E, Kelley EE, Wei WH, McCormick SPA, Bixley MJ, Reynolds RJ, Saag KG, Fadason T, Golovina E, O'Sullivan JM, Stamp LK, Dalbeth N, Abhishek A, Doherty M, Roddy E, Jacobsson LTH, Kapetanovic MC, Melander O, Andrés M, Pérez-Ruiz F, Torres RJ, Radstake T, Jansen TL, Janssen M, Joosten LAB, Liu R, Gaal OI, Crişan TO, Rednic S, Kurreeman F, Huizinga TWJ, Toes R, Lioté F, Richette P, Bardin T, Ea HK, Pascart T, McCarthy GM, Helbert L, Stibůrková B, Tausche AK, Uhlig T, Vitart V, Boutin TS, Hayward C, Riches PL, Ralston SH, Campbell A, MacDonald TM, Nakayama A, Takada T, Nakatochi M, Shimizu S, Kawamura Y, Toyoda Y, Nakaoka H, Yamamoto K, Matsuo K, Shinomiya N, Ichida K, Lee C, Bradbury LA, Brown MA, Robinson PC, Buchanan RRC, Hill CL, Lester S, Smith MD, Rischmueller M, Choi HK, Stahl EA, Miner JN, Solomon DH, Cui J, Giacomini KM, Brackman DJ, Jorgenson EM, Liu H, Susztak K, Shringarpure S, So A, Okada Y, Li C, Shi Y, Merriman TR. A genome-wide association analysis reveals new pathogenic pathways in gout. Nat Genet 2024; 56:2392-2406. [PMID: 39406924 DOI: 10.1038/s41588-024-01921-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/21/2024] [Indexed: 10/18/2024]
Abstract
Gout is a chronic disease that is caused by an innate immune response to deposited monosodium urate crystals in the setting of hyperuricemia. Here, we provide insights into the molecular mechanism of the poorly understood inflammatory component of gout from a genome-wide association study (GWAS) of 2.6 million people, including 120,295 people with prevalent gout. We detected 377 loci and 410 genetically independent signals (149 previously unreported loci in urate and gout). An additional 65 loci with signals in urate (from a GWAS of 630,117 individuals) but not gout were identified. A prioritization scheme identified candidate genes in the inflammatory process of gout, including genes involved in epigenetic remodeling, cell osmolarity and regulation of NOD-like receptor protein 3 (NLRP3) inflammasome activity. Mendelian randomization analysis provided evidence for a causal role of clonal hematopoiesis of indeterminate potential in gout. Our study identifies candidate genes and molecular processes in the inflammatory pathogenesis of gout suitable for follow-up studies.
Collapse
Affiliation(s)
- Tanya J Major
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Riku Takei
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
- Department of Biomedical Information Management, National Defense Medical College Research Institute, National Defense Medical College, Saitama, Japan
| | - Megan P Leask
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicholas A Sumpter
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ruth K Topless
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Wei Wang
- Genomics R&D, 23andMe, Inc, Sunnyvale, CA, USA
| | - Murray J Cadzow
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | - Zhiqiang Li
- The Biomedical Sciences Institute and The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Aichang Ji
- Shandong Provincial Key Laboratory of Metabolic Diseases, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China
| | - Marilyn E Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily Morice
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Wen-Hua Wei
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | | | - Matthew J Bixley
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Richard J Reynolds
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth G Saag
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tayaza Fadason
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Evgenia Golovina
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Justin M O'Sullivan
- Liggins Institute, University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Abhishek Abhishek
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Michael Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Edward Roddy
- School of Medicine, Keele University, Keele, Staffordshire, United Kingdom
- Haywood Academic Rheumatology Centre, Midlands Partnership University NHS Foundation Trust, Stoke-on-Trent, UK
| | - Lennart T H Jacobsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Meliha C Kapetanovic
- Department of Clinical Sciences Lund, Section of Rheumatology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Mariano Andrés
- Rheumatology Department, Dr Balmis General University Hospital-ISABIAL, Alicante, Spain
- Department of Clinical Medicine, Miguel Hernandez University, Alicante, Spain
| | - Fernando Pérez-Ruiz
- Osakidetza, OSI-EE-Cruces, BIOBizkaia Health Research Institute and Medicine Department of Medicine and Nursery School, University of the Basque Country, Biskay, Spain
| | - Rosa J Torres
- Department of Biochemistry, Hospital La Paz Institute for Health Research (IdiPaz), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Timothy Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center, Utrecht, The Netherlands
| | - Timothy L Jansen
- Department of Rheumatology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Matthijs Janssen
- Department of Rheumatology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute of Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ruiqi Liu
- Department of Internal Medicine and Radboud Institute of Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Orsolya I Gaal
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tania O Crişan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Rednic
- Department of Rheumatology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
| | - Fina Kurreeman
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - René Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frédéric Lioté
- Rheumatology Department, Feel'Gout, GH Paris Saint Joseph, Paris, France
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Pascal Richette
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Thomas Bardin
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Hang Korng Ea
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Tristan Pascart
- Department of Rheumatology, Hopital Saint-Philibert, Lille Catholic University, Lille, France
| | - Geraldine M McCarthy
- Department of Rheumatology, Mater Misericordiae University Hospital and School of Medicine, University College, Dublin, Ireland
| | - Laura Helbert
- Department of Rheumatology, Mater Misericordiae University Hospital and School of Medicine, University College, Dublin, Ireland
| | - Blanka Stibůrková
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Institute of Rheumatology, Prague, Czech Republic
| | - Anne-K Tausche
- Department of Rheumatology, University Clinic 'Carl Gustav Carus' at the Technical University, Dresden, Germany
| | - Till Uhlig
- Center for Treatment of Rheumatic and Musculoskeletal Diseases, Diakonhjemmet Hospital, Oslo, Norway
| | - Véronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Thibaud S Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Philip L Riches
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Stuart H Ralston
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Thomas M MacDonald
- MEMO Research, Division of Molecular and Clinical Medicine, University of Dundee Medical School, Ninewells Hospital, Dundee, United Kingdom
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Yusuke Kawamura
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Fukuoka, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology & Prevention, Aichi Cancer Center, Aichi, Japan
- Division of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Aichi, Japan
- The Japan Multi-Institutional Collaborative Cohort (J-MICC) Study, Tokyo, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Kimiyoshi Ichida
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Chaeyoung Lee
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, South Korea
| | - Linda A Bradbury
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Matthew A Brown
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Philip C Robinson
- School of Clinical Medicine, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | | | - Catherine L Hill
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Susan Lester
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | | | - Maureen Rischmueller
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Hyon K Choi
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eli A Stahl
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeff N Miner
- Viscient Biosciences, 5752 Oberlin Dr., Suite 111, San Diego, CA, 92121, USA
| | - Daniel H Solomon
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing Cui
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Deanna J Brackman
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Eric M Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hongbo Liu
- Penn / The Children's Hospital of Pennsylvania Kidney Innovation Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
| | - Katalin Susztak
- Penn / The Children's Hospital of Pennsylvania Kidney Innovation Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
| | | | - Alexander So
- Service of Rheumatology, Center Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Diseases, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China
| | - Yongyong Shi
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China.
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
29
|
Chen HL, Chiang HY, Chang DR, Cheng CF, Wang CCN, Lu TP, Lee CY, Chattopadhyay A, Lin YT, Lin CC, Yu PT, Huang CF, Lin CH, Yeh HC, Ting IW, Tsai HK, Chuang EY, Tin A, Tsai FJ, Kuo CC. Discovery and prioritization of genetic determinants of kidney function in 297,355 individuals from Taiwan and Japan. Nat Commun 2024; 15:9317. [PMID: 39472450 PMCID: PMC11522641 DOI: 10.1038/s41467-024-53516-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/12/2024] [Indexed: 11/02/2024] Open
Abstract
Current genome-wide association studies (GWAS) for kidney function lack ancestral diversity, limiting the applicability to broader populations. The East-Asian population is especially under-represented, despite having the highest global burden of end-stage kidney disease. We conducted a meta-analysis of multiple GWASs (n = 244,952) on estimated glomerular filtration rate and a replication dataset (n = 27,058) from Taiwan and Japan. This study identified 111 lead SNPs in 97 genomic risk loci. Functional enrichment analyses revealed that variants associated with F12 gene and a missense mutation in ABCG2 may contribute to chronic kidney disease (CKD) through influencing inflammation, coagulation, and urate metabolism pathways. In independent cohorts from Taiwan (n = 25,345) and the United Kingdom (n = 260,245), polygenic risk scores (PRSs) for CKD significantly stratified the risk of CKD (p < 0.0001). Further research is required to evaluate the clinical effectiveness of PRSCKD in the early prevention of kidney disease.
Collapse
Affiliation(s)
- Hung-Lin Chen
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biomedical Informatics, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsiu-Yin Chiang
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biomedical Informatics, College of Medicine, China Medical University, Taichung, Taiwan
| | - David Ray Chang
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Chi-Fung Cheng
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Charles C N Wang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Tzu-Pin Lu
- Institute of Health Data Analytics and Statistics, Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chien-Yueh Lee
- Master Program in Artificial Intelligence, Innovation Frontier Institute of Research for Science and Technology, National Taipei University of Technology, Taipei, Taiwan
- Department of Electrical Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Amrita Chattopadhyay
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Institute of Epidemiology and Preventive Medicine, Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Lin
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biomedical Informatics, College of Medicine, China Medical University, Taichung, Taiwan
| | - Che-Chen Lin
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Pei-Tzu Yu
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chien-Fong Huang
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chieh-Hua Lin
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Hung-Chieh Yeh
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - I-Wen Ting
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Huai-Kuang Tsai
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Eric Y Chuang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
- Department of Electrical Engineering, College of Electrical Engineering and Computer Science, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical Electronics and Bioinformatics, College of Electrical Engineering and Computer Science, National Taiwan University, Taipei, Taiwan
| | - Adrienne Tin
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Division of Medical Genetics, China Medical University Children's Hospital, Taichung, Taiwan.
- Department of Medical Laboratory Science & Biotechnology, Asia University, Taichung, Taiwan.
| | - Chin-Chi Kuo
- Big Data Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Department of Biomedical Informatics, College of Medicine, China Medical University, Taichung, Taiwan.
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- College of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
30
|
Chi J, Chen Y, Li C, Liu S, Che K, Kong Z, Guo Z, Chu Y, Huang Y, Yang L, Sun C, Wang Y, Lv W, Zhang Q, Guo H, Zhao H, Yang Z, Xu L, Wang P, Dong B, Hu J, Liu S, Wang F, Zhao Y, Qi M, Xin Y, Nan H, Zhao X, Zhang W, Xiao M, Si K, Wang Y, Cao Y. NUMB dysfunction defines a novel mechanism underlying hyperuricemia and gout. Cell Discov 2024; 10:106. [PMID: 39433541 PMCID: PMC11494200 DOI: 10.1038/s41421-024-00708-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 07/03/2024] [Indexed: 10/23/2024] Open
Abstract
Defective renal excretion and increased production of uric acid engender hyperuricemia that predisposes to gout. However, molecular mechanisms underlying defective uric acid excretion remain largely unknown. Here, we report a rare genetic variant of gout-unprecedented NUMB gene within a hereditary human gout family, which was identified by an unbiased genome-wide sequencing approach. This dysfunctional missense variant within the conserved region of the NUMB gene (NUMBR630H) underwent intracellular redistribution and degradation through an autophagy-dependent mechanism. Mechanistically, we identified the uric acid transporter, ATP Binding Cassette Subfamily G Member 2 (ABCG2), as a novel NUMB-binding protein through its intracellular YxNxxF motif. In polarized renal tubular epithelial cells (RTECs), NUMB promoted ABCG2 trafficking towards the apical plasma membrane. Genetic loss-of-function of NUMB resulted in redistribution of ABCG2 in the basolateral domain and ultimately defective excretion of uric acid. To recapitulate the clinical situation in human gout patients, we generated a NUMBR630H knock-in mouse strain, which showed marked increases of serum urate and decreased uric acid excretion. The NUMBR630H knock-in mice exhibited clinically relevant hyperuricemia. In summary, we have uncovered a novel NUMB-mediated mechanism of uric acid excretion and a functional missense variant of NUMB in humans, which causes hyperuricemia and gout.
Collapse
Affiliation(s)
- Jingwei Chi
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Ying Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Changgui Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China
| | - Shiguo Liu
- Department of Medical Genetics, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Prenatal Diagnosis Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Kui Che
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zili Kong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ziheng Guo
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanchen Chu
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yajing Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Libo Yang
- Department of Endocrinology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Cunwei Sun
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenshan Lv
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qing Zhang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hui Guo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Han Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhitao Yang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lili Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ping Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Bingzi Dong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jianxia Hu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shihai Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fei Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanyun Zhao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mengmeng Qi
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Xin
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Huiqi Nan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiangzhong Zhao
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wei Zhang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Min Xiao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ke Si
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yangang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
31
|
Tang Z, Chen G, Chen S, Yao J, You L, Chen CYC. Modal-nexus auto-encoder for multi-modality cellular data integration and imputation. Nat Commun 2024; 15:9021. [PMID: 39424861 PMCID: PMC11489673 DOI: 10.1038/s41467-024-53355-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
Heterogeneous feature spaces and technical noise hinder the cellular data integration and imputation. The high cost of obtaining matched data across modalities further restricts analysis. Thus, there's a critical need for deep learning approaches to effectively integrate and impute unpaired multi-modality single-cell data, enabling deeper insights into cellular behaviors. To address these issues, we introduce the Modal-Nexus Auto-Encoder (Monae). Leveraging regulatory relationships between modalities and employing contrastive learning within modality-specific auto-encoders, Monae enhances cell representations in the unified space. The integration capability of Monae furnishes it with modality-complementary cellular representations, enabling the generation of precise intra-modal and cross-modal imputation counts for extensive and complex downstream tasks. In addition, we develop Monae-E (Monae-Extension), a variant of Monae that can converge rapidly and support biological discoveries. Evaluations on various datasets have validated Monae and Monae-E's accuracy and robustness in multi-modality cellular data integration and imputation.
Collapse
Affiliation(s)
- Zhenchao Tang
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- AI for Science (AI4S)-Preferred Program, School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Guanxing Chen
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- AI for Science (AI4S)-Preferred Program, School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Shouzhi Chen
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- AI for Science (AI4S)-Preferred Program, School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | | | - Linlin You
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Calvin Yu-Chian Chen
- AI for Science (AI4S)-Preferred Program, School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan.
- Guangdong L-Med Biotechnology Co., Ltd., Meizhou, 514699, China.
| |
Collapse
|
32
|
Wu H, Li H, Dai X, Dai Y, Liu H, Xu S, Huang J, Chi H, Wang S. A Mendelian randomization study of the association between serum uric acid and osteoporosis risk. Front Endocrinol (Lausanne) 2024; 15:1434602. [PMID: 39464184 PMCID: PMC11502378 DOI: 10.3389/fendo.2024.1434602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND The relationship between serum uric acid (SUA) and osteoporosis (OP) has yielded conflicting results in observational studies. This Mendelian randomization (MR) study aims to elucidate the causal association between SUA and OP. METHODS A two-sample MR analysis was conducted using summary-level data from genome-wide association studies (GWAS). Two sets of polygenic instruments strongly associated (p < 5 × 10-8) with SUA were extracted from the CKDGen consortium and UK biobank. Polygenic instruments associated with OP (p < 5 × 10-8) were derived from FinnGen biobank and UK biobank. Inverse variance weighting (IVW) was employed as the primary analysis method. Additionally, we utilized MR-Egger, weighted median, the simple mode method, and the weighted mode as complementary analyses. Cochran's Q statistics were used to assess heterogeneity, with MR-Egger intercept testing and MR pleiotropy residual sum and outlier (MR-PRESSO) to examine horizontal pleiotropy. Sensitivity analysis was performed using the leave-one-out method. RESULTS The IVW analysis conducted across four groups confirms no significant causal relationship between SUA concentration and OP: UKB-UKB (OR: 1.001, 95% CI: 0.999-1.003, p=0.464), CKD-UKB (OR: 1.001, 95% CI: 0.999-1.003, p=0.349), UKB-Fin (OR: 0.934, 95% CI: 0.747-1.168, p=0.549), CKD-Fin (OR: 1.041, 95%CI: 0.934-1.161, p=0.470). Furthermore, additional four MR analyses corroborated these findings. Upon excluding all outliers identified by the MR-PRESSO test, no significant directional pleiotropy was observed, except for some data heterogeneity noted in the UKB-UKB group (Q=50.65, P=0.002). Additionally, a leave-one-out analysis indicated that no single SNP exerted undue influence on the results. CONCLUSION This MR analysis provides convincing genetic evidence that there is no causal association between SUA and OP, SUA is unlikely to increase or reduce the risk of OP.
Collapse
Affiliation(s)
- Heng Wu
- Department of Orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hairui Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xiao Dai
- Department of Orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Dai
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Liu
- Department of Orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shuang Xu
- Department of Orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinbang Huang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Song Wang
- Department of Orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
33
|
Xiao W, Wang Q, Liu Y, Zhang H, Zou H. Association of visceral adipose tissue with gout: Observational and Mendelian randomization analyses. Chin Med J (Engl) 2024; 137:2351-2357. [PMID: 37882086 PMCID: PMC11441863 DOI: 10.1097/cm9.0000000000002908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND The causal relationship between visceral adipose tissue (VAT) and gout is still unclear. We aimed to examine the potential association between them using observational and Mendelian randomization (MR) analyses. METHODS In the observational analyses, a total of 11,967 participants (aged 39.5 ± 11.5 years) were included from the National Health and Nutrition Examination Survey. Logistic regression models were used to investigate the association between VAT mass and the risk of gout. In two-sample MR analyses, 211 VAT mass-related independent genetic variants (derived from genome-wide association studies in 325,153 UK biobank participants) were used as instrumental variables. The random-effects inverse-variance weighted (IVW) method was used as the primary analysis. Additional sensitivity analyses were also performed to validate our results. RESULTS Observational analyses found that an increase in VAT mass (per standard deviation) was associated with a higher risk of gout after controlling for confounding factors (odds ratio [OR] = 1.27, 95% confidence intervals [CI] = 1.11-1.45). The two-sample MR analyses demonstrated a causal relationship between increased VAT mass and the risk of gout in primary analyses (OR = 1.78, 95% CI = 1.57-2.03). Sensitivity analyses also showed similar findings, including MR-Egger, weighted median, simple mode, weighted mode, and leave-one-out analyses. CONCLUSIONS Observational analyses showed a robust association of VAT mass with the risk of gout. Meanwhile, MR analyses also provided evidence of a causal relationship between them. In summary, our findings suggested that targeted interventions for VAT mass may be beneficial to prevent gout.
Collapse
Affiliation(s)
- Wenze Xiao
- Department of Rheumatology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Qi Wang
- Department of Nephrology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Yining Liu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Centre, Fudan University, Shanghai 201203, China
| | - Hui Zhang
- Human Phenome Institute, Zhangjiang Fudan International Innovation Centre, Fudan University, Shanghai 201203, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai 200000, China
| |
Collapse
|
34
|
Lee S, Shin D. A combination of red and processed meat intake and polygenic risk score influences the incidence of hyperuricemia in middle-aged Korean adults. Nutr Res Pract 2024; 18:721-745. [PMID: 39398885 PMCID: PMC11464275 DOI: 10.4162/nrp.2024.18.5.721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/03/2024] [Accepted: 08/22/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND/OBJECTIVES The high consumption of purine-rich meat is associated with hyperuricemia. However, there is limited evidence linking the consumption of red and processed meat to the genetic risk of hyperuricemia. We investigated the relationship between various combinations of red and processed meat consumption and the polygenic risk scores (PRSs) and the incidence of hyperuricemia in middle-aged Koreans. SUBJECTS/METHODS We analyzed the data from 44,053 participants aged ≥40 years sourced from the Health Examinees (HEXA) cohort of the Korean Genome and Epidemiology Study (KoGES). Information regarding red and processed meat intake was obtained using a semiquantitative food frequency questionnaire (SQ-FFQ). We identified 69 independent single-nucleotide polymorphisms (SNPs) at uric acid-related loci using genome-wide association studies (GWASs) and clumping analyses. The individual PRS, which is the weighted sum of the effect size of each allele at the SNP, was calculated. We used multivariable Cox proportional hazards models adjusted for covariates to determine the relationship between red and processed meat intake and the PRS in the incidence of hyperuricemia. RESULTS During an average follow-up period of 5 years, 2,556 patients with hyperuricemia were identified. For both men and women, the group with the highest red and processed meat intake and the highest PRS was positively associated with the development of hyperuricemia when compared with the group with the lowest red and processed meat intake and the lowest PRS (hazard ratio [HR], 2.72; 95% confidence interval [CI], 2.10-3.53; P < 0.0001; HR, 3.28; 95% CI, 2.45-4.40; P < 0.0001). CONCLUSION Individuals at a high genetic risk for uric acid levels should moderate their consumption of red and processed meat to prevent hyperuricemia.
Collapse
Affiliation(s)
- Suyeon Lee
- Department of Food and Nutrition, Inha University, Incheon 22212, Korea
| | - Dayeon Shin
- Department of Food and Nutrition, Inha University, Incheon 22212, Korea
| |
Collapse
|
35
|
Borghi C, Fogacci F, Piani F. Causal relationship between serum uric acid and cardiovascular disease: A Mendelian randomization study. IJC HEART & VASCULATURE 2024; 54:101503. [PMID: 39411144 PMCID: PMC11474365 DOI: 10.1016/j.ijcha.2024.101503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Claudio Borghi
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Federica Fogacci
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Federica Piani
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
36
|
Li J, Yang G, Liu J, Li G, Zhou H, He Y, Fei X, Zhao D. Integrating transcriptomics, eQTL, and Mendelian randomization to dissect monocyte roles in severe COVID-19 and gout flare. Front Genet 2024; 15:1385316. [PMID: 39385934 PMCID: PMC11461236 DOI: 10.3389/fgene.2024.1385316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction There are considerable similarities between the pathophysiology of gout flare and the dysregulated inflammatory response in severe COVID-19 infection. Monocytes are the key immune cells involved in the pathogenesis of both diseases. Therefore, it is critical to elucidate the molecular basis of the function of monocytes in gout and COVID-19 in order to develop more effective therapeutic approaches. Methods The single-cell RNA sequencing (scRNA-seq), large-scale genome-wide association studies (GWAS), and expression quantitative trait loci (eQTL) data of gout and severe COVID-19 were comprehensively analyzed. Cellular heterogeneity and intercellular communication were identified using the scRNA-seq datasets, and the monocyte-specific differentially expressed genes (DEGs) between COVID-19, gout and normal subjects were screened. In addition, the correlation of the DEGs with severe COVID-19 and gout flare was analyzed through GWAS statistics and eQTL data. Results The scRNA-seq analysis exhibited that the proportion of classical monocytes was increased in both severe COVID-19 and gout patient groups compared to healthy controls. Differential expression analysis and MR analysis showed that NLRP3 was positively associated with the risk of severe COVID-19 and involved 11 SNPs, of which rs4925547 was not significantly co-localized. In contrast, IER3 was positively associated with the risk of gout and involved 9 SNPs, of which rs1264372 was significantly co-localized. Discussion Monocytes have a complex role in gout flare and severe COVID-19, which underscores the potential mechanisms and clinical significance of the interaction between the two diseases.
Collapse
Affiliation(s)
- Jiajia Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guixian Yang
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Junnan Liu
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guofeng Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Huiling Zhou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yuan He
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xinru Fei
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Dongkai Zhao
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
37
|
Sun J, Shen T, Guan Y, Jiang Y, Xu X. The Causal Effect of Urate Level on Female Infertility: A Mendelian Randomization Study. Metabolites 2024; 14:516. [PMID: 39452897 PMCID: PMC11509567 DOI: 10.3390/metabo14100516] [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: 08/19/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVE This study aimed to investigate the causal relationship between urate level and female infertility using Mendelian randomization (MR) analysis. METHODS To identify instrumental variables, we selected independent genetic loci associated with serum urate levels in individuals of European ancestry, utilizing data from large-scale genome-wide association studies (GWAS). The GWAS dataset included information on serum urate levels from 288,649 CKDGen participants. Female infertility data, including different etiologic classifications, consisted of 13,142 female infertility patients and 107,564 controls. We employed four MR methods, namely inverse variance weighted (IVW), MR-Egger, weighted median, and weighted model, to investigate the causal relationship between urate levels and female infertility. The Cochran Q-test was used to assess heterogeneity among single nucleotide polymorphisms (SNPs), and the MR-Egger intercept test was employed to evaluate the presence of horizontal pleiotropy. Additionally, a "leave-one-out" sensitivity analysis was conducted to examine the influence of individual SNPs on the MR study. RESULTS The IVW analysis demonstrated that elevated serum urate levels increased the risk of female infertility (odds ratio [OR] = 1.18, 95% confidence interval [CI]: 1.07-1.33). Furthermore, serum urate levels were found to be associated with infertility due to cervical, vaginal, or other unknown causes (OR = 1.16, 95% CI: 1.06-1.26), also confirmed by other methods. Heterogeneity among instrumental variables was assessed using Cochran's Q-test (p < 0.05), so a random-effects IVW approach was employed in the effects model. The MR-Egger intercept test indicated no presence of horizontal pleiotropy. A "leave-one-out" sensitivity analysis was conducted, demonstrating that no individual SNP had a substantial impact on the overall findings. CONCLUSIONS In the European population, the urate level is significantly and causally associated with an increased risk of female infertility.
Collapse
Affiliation(s)
- Jiawei Sun
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310052, China
- School of Medicine, Zhejiang University, No. 866 Yuhantang Road, Hangzhou 310058, China
| | - Ting Shen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310052, China
| | - Yining Guan
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310052, China
| | - Yixin Jiang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310052, China
| | - Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310052, China
| |
Collapse
|
38
|
Wang M, Tang Z. No causal relationship between serum urate and neurodegenerative diseases: A Mendelian randomization study. Exp Gerontol 2024; 194:112503. [PMID: 38955238 DOI: 10.1016/j.exger.2024.112503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
OBJECTIVE Observational studies have shown that increased serum urate is associated with a lower risk of neurodegenerative diseases (NDs), but the causality remains unclear. We employed a two-sample Mendelian randomization (MR) approach to assess the causal relationship between serum urate and four common subtypes of NDs, including Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). METHODS Serum urate data came from the CKDGen Consortium. GWAS data for PD, AD, ALS, and MS were obtained from four databases in the primary analysis and then acquired statistics from the FinnGen consortium for replication and meta-analysis. Inverse variance weighted (IVW), weighted median (WM), and MR-Egger regression methods were applied in the MR analyses. Pleiotropic effects, heterogeneity, and leave-one-out analyses were evaluated to validate the results. RESULTS There was no evidence for the effect of serum urate on PD (OR: 1.00, 95 % CI: 0.90-1.11, P = 0.97), AD (OR: 1.02, 95 % CI: 1.00-1.04, P = 0.06), ALS (OR: 1.05, 95 % CI: 0.97-1.13, P = 0.22), and MS (OR: 1.01, 95 % CI: 0.89-1.14, P = 0.90) risk when combined with the FinnGen consortium, neither was any evidence of pleiotropy detected between the instrumental variables (IVs). CONCLUSION The MR analysis suggested that serum urate may not be causally associated with a risk of PD, AD, ALS, and MS.
Collapse
Affiliation(s)
- Min Wang
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Zhiquan Tang
- People's Hospital of Yushan District, Ma'anshan, Anhui 243000, China.
| |
Collapse
|
39
|
Zeng L, Ma P, Li Z, Liang S, Wu C, Hong C, Li Y, Cui H, Li R, Wang J, He J, Li W, Xiao L, Liu L. Multimodal Machine Learning-Based Marker Enables Early Detection and Prognosis Prediction for Hyperuricemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404047. [PMID: 38976552 PMCID: PMC11425915 DOI: 10.1002/advs.202404047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/19/2024] [Indexed: 07/10/2024]
Abstract
Hyperuricemia (HUA) has emerged as the second most prevalent metabolic disorder characterized by prolonged and asymptomatic period, triggering gout and metabolism-related outcomes. Early detection and prognosis prediction for HUA and gout are crucial for pre-emptive interventions. Integrating genetic and clinical data from 421287 UK Biobank and 8900 Nanfang Hospital participants, a stacked multimodal machine learning model is developed and validated to synthesize its probabilities as an in-silico quantitative marker for hyperuricemia (ISHUA). The model demonstrates satisfactory performance in detecting HUA, exhibiting area under the curves (AUCs) of 0.859, 0.836, and 0.779 within the train, internal, and external test sets, respectively. ISHUA is significantly associated with gout and metabolism-related outcomes, effectively classifying individuals into low- and high-risk groups for gout in the train (AUC, 0.815) and internal test (AUC, 0.814) sets. The high-risk group shows increased susceptibility to metabolism-related outcomes, and participants with intermediate or favorable lifestyle profiles have hazard ratios of 0.75 and 0.53 for gout compared with those with unfavorable lifestyles. Similar trends are observed for other metabolism-related outcomes. The multimodal machine learning-based ISHUA marker enables personalized risk stratification for gout and metabolism-related outcomes, and it is unveiled that lifestyle changes can ameliorate these outcomes within high-risk group, providing guidance for preventive interventions.
Collapse
Affiliation(s)
- Lin Zeng
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pengcheng Ma
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zeyang Li
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shengxing Liang
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chengkai Wu
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
| | - Chang Hong
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Cui
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ruining Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiaren Wang
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jingzhe He
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenyuan Li
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lushan Xiao
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Liu
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
40
|
Hua B, Dong Z, Yang Y, Liu W, Chen S, Chen Y, Sun X, Ye D, Li J, Mao Y. Dietary Carbohydrates, Genetic Susceptibility, and Gout Risk: A Prospective Cohort Study in the UK. Nutrients 2024; 16:2883. [PMID: 39275199 PMCID: PMC11397129 DOI: 10.3390/nu16172883] [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: 08/01/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
This study aimed to investigate the associations between carbohydrate intake and gout risk, along with interactions between genetic susceptibility and carbohydrates, and the mediating roles of biomarkers. We included 187,387 participants who were free of gout at baseline and completed at least one dietary assessment in the UK Biobank. Cox proportional hazard models were used to estimate the associations between carbohydrate intake and gout risk. Over a median follow-up of 11.69 years, 2548 incident cases of gout were recorded. Total carbohydrate intake was associated with a reduced gout risk (Q4 vs. Q1: HR 0.67, 95% CI 0.60-0.74), as were total sugars (0.89, 0.80-0.99), non-free sugars (0.70, 0.63-0.78), total starch (0.70, 0.63-0.78), refined grain starch (0.85, 0.76-0.95), wholegrain starch (0.73, 0.65-0.82), and fiber (0.72, 0.64-0.80), whereas free sugars (1.15, 1.04-1.28) were associated with an increased risk. Significant additive interactions were found between total carbohydrates and genetic risk, as well as between total starch and genetic risk. Serum urate was identified as a significant mediator in all associations between carbohydrate intake (total, different types, and sources) and gout risk. In conclusion, total carbohydrate and different types and sources of carbohydrate (excluding free sugars) intake were associated with a reduced risk of gout.
Collapse
Affiliation(s)
- Baojie Hua
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziwei Dong
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yudan Yang
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei Liu
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shuhui Chen
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ying Chen
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaohui Sun
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ding Ye
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiayu Li
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yingying Mao
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
41
|
Du L, Zong Y, Li H, Wang Q, Xie L, Yang B, Pang Y, Zhang C, Zhong Z, Gao J. Hyperuricemia and its related diseases: mechanisms and advances in therapy. Signal Transduct Target Ther 2024; 9:212. [PMID: 39191722 DOI: 10.1038/s41392-024-01916-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 08/29/2024] Open
Abstract
Hyperuricemia, characterized by elevated levels of serum uric acid (SUA), is linked to a spectrum of commodities such as gout, cardiovascular diseases, renal disorders, metabolic syndrome, and diabetes, etc. Significantly impairing the quality of life for those affected, the prevalence of hyperuricemia is an upward trend globally, especially in most developed countries. UA possesses a multifaceted role, such as antioxidant, pro-oxidative, pro-inflammatory, nitric oxide modulating, anti-aging, and immune effects, which are significant in both physiological and pathological contexts. The equilibrium of circulating urate levels hinges on the interplay between production and excretion, a delicate balance orchestrated by urate transporter functions across various epithelial tissues and cell types. While existing research has identified hyperuricemia involvement in numerous biological processes and signaling pathways, the precise mechanisms connecting elevated UA levels to disease etiology remain to be fully elucidated. In addition, the influence of genetic susceptibilities and environmental determinants on hyperuricemia calls for a detailed and nuanced examination. This review compiles data from global epidemiological studies and clinical practices, exploring the physiological processes and the genetic foundations of urate transporters in depth. Furthermore, we uncover the complex mechanisms by which the UA induced inflammation influences metabolic processes in individuals with hyperuricemia and the association with its relative disease, offering a foundation for innovative therapeutic approaches and advanced pharmacological strategies.
Collapse
Grants
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
- 82002339, 81820108020 National Natural Science Foundation of China (National Science Foundation of China)
Collapse
Affiliation(s)
- Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Haorui Li
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Qiyue Wang
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Lei Xie
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Bo Yang
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Zhigang Zhong
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China.
| | - Junjie Gao
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
- Institute of Sports Medicine, Shantou University Medical College, Shantou, 515041, China.
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
42
|
Hu H, Li W, Hao Y, Peng Z, Zou Z, Wei J, Zhou Y, Liang W, Cao Y. The SGLT2 inhibitor dapagliflozin ameliorates renal fibrosis in hyperuricemic nephropathy. Cell Rep Med 2024; 5:101690. [PMID: 39168099 PMCID: PMC11384938 DOI: 10.1016/j.xcrm.2024.101690] [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: 01/03/2024] [Revised: 07/01/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024]
Abstract
Hyperuricemic nephropathy (HN) is a global metabolic disorder characterized by uric acid (UA) metabolism dysfunction, resulting in hyperuricemia (HUA) and tubulointerstitial fibrosis (TIF). Sodium-dependent glucose transporter 2 inhibitor, dapagliflozin, has shown potential in reducing serum UA levels in patients with chronic kidney disease (CKD), though its protective effects against HN remain uncertain. This study investigates the functional, pathological, and molecular changes in HN through histological, biochemical, and transcriptomic analyses in patients, HN mice, and UA-stimulated HK-2 cells. Findings indicate UA-induced tubular dysfunction and fibrotic activation, which dapagliflozin significantly mitigates. Transcriptomic analysis identifies estrogen-related receptor α (ERRα), a downregulated transcription factor in HN. ERRα knockin mice and ERRα-overexpressed HK-2 cells demonstrate UA resistance, while ERRα inhibition exacerbates UA effects. Dapagliflozin targets ERRα, activating the ERRα-organic anion transporter 1 (OAT1) axis to enhance UA excretion and reduce TIF. Furthermore, dapagliflozin ameliorates renal fibrosis in non-HN CKD models, underscoring the therapeutic significance of the ERRα-OAT1 axis in HN and CKD.
Collapse
Affiliation(s)
- Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Road, Wuhan, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiwei Li
- Division of Nephrology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Avenue, Enshi, China
| | - Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhuan Peng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhengping Zou
- Division of Nephrology, Qianjiang Hospital Affiliated to Renmin Hospital of Wuhan University, Wuhan, China; Qianjiang Clinical Medical College, Health Science Center, Yangtze University, Jingzhou, China
| | - Jiali Wei
- Department of Nephrology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Ying Zhou
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, No. 45 Changchun St, Xicheng District, Beijing 100053, China.
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Key Clinical Research Center of Kidney Disease in Hubei, 238 Jiefang Road, Wuhan, China.
| | - Yun Cao
- Department of Nephrology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China.
| |
Collapse
|
43
|
Leask MP, Crișan TO, Ji A, Matsuo H, Köttgen A, Merriman TR. The pathogenesis of gout: molecular insights from genetic, epigenomic and transcriptomic studies. Nat Rev Rheumatol 2024; 20:510-523. [PMID: 38992217 DOI: 10.1038/s41584-024-01137-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/13/2024]
Abstract
The pathogenesis of gout involves a series of steps beginning with hyperuricaemia, followed by the deposition of monosodium urate crystal in articular structures and culminating in an innate immune response, mediated by the NLRP3 inflammasome, to the deposited crystals. Large genome-wide association studies (GWAS) of serum urate levels initially identified the genetic variants with the strongest effects, mapping mainly to genes that encode urate transporters in the kidney and gut. Other GWAS highlighted the importance of uncommon genetic variants. More recently, genetic and epigenetic genome-wide studies have revealed new pathways in the inflammatory process of gout, including genetic associations with epigenomic modifiers. Epigenome-wide association studies are also implicating epigenomic remodelling in gout, which perhaps regulates the responsiveness of the innate immune system to monosodium urate crystals. Notably, genes implicated in gout GWAS do not include those encoding components of the NLRP3 inflammasome itself, but instead include genes encoding molecules involved in its regulation. Knowledge of the molecular mechanisms underlying gout has advanced through the translation of genetic associations into specific molecular mechanisms. Notable examples include ABCG2, HNF4A, PDZK1, MAF and IL37. Current genetic studies are dominated by participants of European ancestry; however, studies focusing on other population groups are discovering informative population-specific variants associated with gout.
Collapse
Affiliation(s)
- Megan P Leask
- Department of Physiology, University of Otago, Dunedin, Aotearoa, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tania O Crișan
- Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Aichang Ji
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Microbiology and Immunology, University of Otago, Dunedin, Aotearoa, New Zealand.
| |
Collapse
|
44
|
Wang L, Mesa-Eguiagaray I, Campbell H, Wilson JF, Vitart V, Li X, Theodoratou E. A phenome-wide association and factorial Mendelian randomization study on the repurposing of uric acid-lowering drugs for cardiovascular outcomes. Eur J Epidemiol 2024; 39:869-880. [PMID: 38992218 PMCID: PMC11410910 DOI: 10.1007/s10654-024-01138-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024]
Abstract
Uric acid has been linked to various disease outcomes. However, it remains unclear whether uric acid-lowering therapy could be repurposed as a treatment for conditions other than gout. We first performed both observational phenome-wide association study (Obs-PheWAS) and polygenic risk score PheWAS (PRS-PheWAS) to identify associations of uric acid levels with a wide range of disease outcomes. Then, trajectory analysis was conducted to explore temporal progression patterns of the observed disease outcomes. Finally, we investigated whether uric acid-lowering drugs could be repurposed using a factorial Mendelian randomization (MR) study design. A total of 41 overlapping phenotypes associated with uric acid levels were identified by both Obs- and PRS- PheWASs, primarily cardiometabolic diseases. The trajectory analysis illustrated how elevated uric acid levels contribute to cardiometabolic diseases, and finally death. Meanwhile, we found that uric acid-lowering drugs exerted a protective role in reducing the risk of coronary atherosclerosis (OR = 0.96, 95%CI: 0.93, 1.00, P = 0.049), congestive heart failure (OR = 0.64, 95%CI: 0.42, 0.99, P = 0.043), occlusion of cerebral arteries (OR = 0.93, 95%CI: 0.87, 1.00, P = 0.044) and peripheral vascular disease (OR = 0.60, 95%CI: 0.38, 0.94, P = 0.025). Furthermore, the combination of uric acid-lowering therapy (e.g. xanthine oxidase inhibitors) with antihypertensive treatment (e.g. calcium channel blockers) exerted additive effects and was associated with a 6%, 8%, 8%, 10% reduction in risk of coronary atherosclerosis, heart failure, occlusion of cerebral arteries and peripheral vascular disease, respectively. Our findings support a role of elevated uric acid levels in advancing cardiovascular dysfunction and identify potential repurposing opportunities for uric acid-lowering drugs in cardiovascular treatment.
Collapse
Affiliation(s)
- Lijuan Wang
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Ines Mesa-Eguiagaray
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Harry Campbell
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - James F Wilson
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK.
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
45
|
Liu Z, Crișan TO, Qi C, Gupta MK, Liu X, Moorlag SJ, Koeken VA, de Bree LCJ, Mourits VP, Gao X, Baccarelli A, Schwartz J, Pessler F, Guzmán CA, Li Y, Netea MG, Joosten LA, Xu CJ. Sex-specific epigenetic signatures of circulating urate and its increase after BCG vaccination. RESEARCH SQUARE 2024:rs.3.rs-4498597. [PMID: 39108482 PMCID: PMC11302698 DOI: 10.21203/rs.3.rs-4498597/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Background Urate concentration and the physiological regulation of urate homeostasis exhibit clear sex differences. DNA methylation has been shown to explain a substantial proportion of serum urate variance, mediate the genetic effect on urate concentration, and co-regulate with cardiometabolic traits. However, whether urate concentration is associated with DNA methylation in a sex-dependent manner is unknown. Additionally, it is worth investigating if urate changes after perturbations, such as vaccination, are associated with DNA methylation in a sex-specific manner. Methods We investigated the association between DNA methylation and serum urate concentrations in a Dutch cohort of 325 healthy individuals. Urate concentration and DNA methylation were measured before and after Bacillus Calmette-Guérin (BCG) vaccination, used as a perturbation associated with increased gout flares. The association analysis included united, interaction, and sex-stratified analysis. Validation of the identified CpG sites was conducted using three independent cohorts. Results 215 CpG sites were associated with serum urate in males, while 5 CpG sites were associated with serum urate in females, indicating sex-specific associations. Circulating urate concentrations significantly increased after BCG vaccination, and baseline DNA methylation was associated with differences in urate concentration before and after vaccination in a sex-specific manner. The CpG sites associated with urate concentration in males were enriched in neuro-protection pathways, whereas in females, the urate change-associated CpG sites were related to lipid and glucose metabolism. Conclusion Our study enhances the understanding of how epigenetic factors contribute to regulating serum urate levels in a sex-specific manner. These insights have significant implications for the diagnosis, prevention, and treatment of various urate-related diseases and highlight the importance of personalized and sex-specific approaches in medicine.
Collapse
Affiliation(s)
- Zhaoli Liu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Tania O. Crișan
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Department of Medical Genetics, „Iuliu Hațieganu” University of Medicine and Pharmacy. Cluj-Napoca, Romania
| | - Cancan Qi
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Manoj Kumar Gupta
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Xuan Liu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Simone J.C.F.M. Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
| | - Valerie A.C.M. Koeken
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Research Centre Innovations in Care, Rotterdam University of Applied Sciences, Rotterdam, the Netherlands
| | - L. Charlotte J. de Bree
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
| | - Vera P. Mourits
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
| | - Xu Gao
- Department of Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Andrea Baccarelli
- Department of Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Frank Pessler
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Research Group Biomarkers for Infectious Diseases, TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Carlos A. Guzmán
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Department Vaccinology and Applied Microbiology, Helmholtz-Centre for Infection Research (HZI), Braunschweig, Germany
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Cluster of Excellence RESIST (EXC 2155), Hanover Medical School, Hannover, Germany
- Lower Saxony center for artificial intelligence and causal methods in medicine (CAIMed). Hannover, Germany
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES). University of Bonn. Bonn, Germany
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Department of Medical Genetics, „Iuliu Hațieganu” University of Medicine and Pharmacy. Cluj-Napoca, Romania
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| |
Collapse
|
46
|
Fukui S, Okada M, Shinozaki T, Asano T, Nakai T, Tamaki H, Kishimoto M, Hasegawa H, Matsuda T, Marrugo J, Tedeschi SK, Choi H, Solomon DH. Changes in alcohol intake and serum urate changes: longitudinal analyses of annual medical examination database. Ann Rheum Dis 2024; 83:1072-1081. [PMID: 38418204 PMCID: PMC11250628 DOI: 10.1136/ard-2023-225389] [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: 12/07/2023] [Accepted: 02/16/2024] [Indexed: 03/01/2024]
Abstract
INTRODUCTION Despite the established cross-sectional association between alcohol intake and serum urate (SU), its longitudinal association remains unknown. This study aimed to determine whether changes in alcohol intake have a clinically relevant association with SU change. METHOD We conducted retrospective analyses using systematically collected annual medical examination data from October 2012 to October 2022 in a Japanese preventive medicine centre. The exposure was changes in alcohol intake between two consecutive visits. The association of SU changes with alcohol intake changes was estimated by mixed-effect linear regression with adjustment for relevant covariates. RESULTS We analysed 63 486 participants (median age, 47.0 years; 55% women; 58.6% regular alcohol drinkers with a median of 1.4 drinks/day) with 370 572 visits. The median SU level was 5.3 mg/dL, and 506 (0.8%) participants had diagnoses of gout or hyperuricemia without medication use during the study period. Decreasing one daily alcohol intake had a clinically small association with SU changes (-0.019 (95% CI: -0.021 to -0.017) mg/dL). Beer had the largest association with SU (-0.036 (95% CI: -0.039 to -0.032) mg/dL for one beer decrease). Complete discontinuation of any alcohol from a mean of 0.8 drinks/day was associated with -0.056 mg/dL (95% CI: -0.068 to -0.043) decrease in SU; the association became larger in hyperuricemic participants (-0.110 mg/dL (95% CI: -0.154 to -0.066) for alcohol discontinuation from a mean of 1.0 drinks/day). CONCLUSIONS This study revealed changes in alcohol intake had small associations with SU change at the general Japanese population level. Complete discontinuation of alcohol in hyperuricemic participants had only modest improvement in SU.
Collapse
Affiliation(s)
- Sho Fukui
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Immuno-Rheumatology Center, St. Luke's international Hospital, Tokyo, Japan
- Department of Emergency and General Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Masato Okada
- Immuno-Rheumatology Center, St. Luke's international Hospital, Tokyo, Japan
| | - Tomohiro Shinozaki
- Department of Information and Computer Technology, Faculty of Engineering, Tokyo University of Science, Tokyo, Japan
| | - Takahiro Asano
- Immuno-Rheumatology Center, St. Luke's international Hospital, Tokyo, Japan
| | - Takehiro Nakai
- Immuno-Rheumatology Center, St. Luke's international Hospital, Tokyo, Japan
| | - Hiromichi Tamaki
- Immuno-Rheumatology Center, St. Luke's international Hospital, Tokyo, Japan
| | - Mitsumasa Kishimoto
- Immuno-Rheumatology Center, St. Luke's international Hospital, Tokyo, Japan
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan
| | - Hiroshi Hasegawa
- Department of Emergency and General Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Takeaki Matsuda
- Department of Emergency and General Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Javier Marrugo
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sara K Tedeschi
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hyon Choi
- Arthritis Research Canada, Richmond, Virginia, Canada
- Division of Rheumatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel H Solomon
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Monti R, Eick L, Hudjashov G, Läll K, Kanoni S, Wolford BN, Wingfield B, Pain O, Wharrie S, Jermy B, McMahon A, Hartonen T, Heyne H, Mars N, Lambert S, Hveem K, Inouye M, van Heel DA, Mägi R, Marttinen P, Ripatti S, Ganna A, Lippert C. Evaluation of polygenic scoring methods in five biobanks shows larger variation between biobanks than methods and finds benefits of ensemble learning. Am J Hum Genet 2024; 111:1431-1447. [PMID: 38908374 PMCID: PMC11267524 DOI: 10.1016/j.ajhg.2024.06.003] [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: 11/20/2023] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/24/2024] Open
Abstract
Methods of estimating polygenic scores (PGSs) from genome-wide association studies are increasingly utilized. However, independent method evaluation is lacking, and method comparisons are often limited. Here, we evaluate polygenic scores derived via seven methods in five biobank studies (totaling about 1.2 million participants) across 16 diseases and quantitative traits, building on a reference-standardized framework. We conducted meta-analyses to quantify the effects of method choice, hyperparameter tuning, method ensembling, and the target biobank on PGS performance. We found that no single method consistently outperformed all others. PGS effect sizes were more variable between biobanks than between methods within biobanks when methods were well tuned. Differences between methods were largest for the two investigated autoimmune diseases, seropositive rheumatoid arthritis and type 1 diabetes. For most methods, cross-validation was more reliable for tuning hyperparameters than automatic tuning (without the use of target data). For a given target phenotype, elastic net models combining PGS across methods (ensemble PGS) tuned in the UK Biobank provided consistent, high, and cross-biobank transferable performance, increasing PGS effect sizes (β coefficients) by a median of 5.0% relative to LDpred2 and MegaPRS (the two best-performing single methods when tuned with cross-validation). Our interactively browsable online-results and open-source workflow prspipe provide a rich resource and reference for the analysis of polygenic scoring methods across biobanks.
Collapse
Affiliation(s)
- Remo Monti
- Hasso Plattner Institute, University of Potsdam, Digital Engineering Faculty, Potsdam, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Lisa Eick
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Georgi Hudjashov
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kristi Läll
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Brooke N Wolford
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Benjamin Wingfield
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Oliver Pain
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience; Institute of Psychiatry, Psychology and Neuroscience; King's College London, London, UK
| | - Sophie Wharrie
- Aalto University, Department of Computer Science, Espoo, Finland
| | - Bradley Jermy
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Aoife McMahon
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Tuomo Hartonen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Henrike Heyne
- Hasso Plattner Institute, University of Potsdam, Digital Engineering Faculty, Potsdam, Germany
| | - Nina Mars
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuel Lambert
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK; British Heart Foundation Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; 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 Cambridge Centre of Research Excellence, School of Clinical Medicine, University of Cambridge, Cambridge, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | | | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Pekka Marttinen
- Aalto University, Department of Computer Science, Espoo, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Andrea Ganna
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Massachusetts General Hospital and Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christoph Lippert
- Hasso Plattner Institute, University of Potsdam, Digital Engineering Faculty, Potsdam, Germany; Windreich Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
48
|
Zhong Y, Yang C, Zhang B, Chen Y, Cai W, Wang G, Zhao C, Zhao W. Causal impact of human blood metabolites and metabolic pathways on serum uric acid and gout: a mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1378645. [PMID: 39027467 PMCID: PMC11256090 DOI: 10.3389/fendo.2024.1378645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
Objective Hyperuricaemia and gout are common metabolic disorders. However, the causal relationships between blood metabolites and serum urate levels, as well as gout, remain unclear. A systematic evaluation of the causal connections between blood metabolites, hyperuricemia, and gout could enhance early screening and prevention of hyperuricemia and gout in clinical settings, providing novel insights and approaches for clinical treatment. Methods In this study, we employed a bidirectional two-sample Mendelian randomization analysis utilizing data from a genome-wide association study involving 7,286 participants, encompassing 486 blood metabolites. Serum urate and gout data were sourced from the Chronic Kidney Disease Genetics consortium, including 288,649 participants for serum urate and 9,819 African American and 753,994 European individuals for gout. Initially, LDSC methodology was applied to identify blood metabolites with a genetic relationship to serum urate and gout. Subsequently, inverse-variance weighting was employed as the primary analysis method, with a series of sensitivity and pleiotropy analyses conducted to assess the robustness of the results. Results Following LDSC, 133 blood metabolites exhibited a potential genetic relationship with serum urate and gout. In the primary Mendelian randomization analysis using inverse-variance weighting, 19 blood metabolites were recognized as potentially influencing serum urate levels and gout. Subsequently, the IVW p-values of potential metabolites were corrected using the false discovery rate method. We find leucine (IVW P FDR = 0.00004), N-acetylornithine (IVW P FDR = 0.0295), N1-methyl-3-pyridone-4-carboxamide (IVW P FDR = 0.0295), and succinyl carnitine (IVW P FDR = 0.00004) were identified as significant risk factors for elevated serum urate levels. Additionally, 1-oleoylglycerol (IVW P FDR = 0.0007) may lead to a substantial increase in the risk of gout. Succinyl carnitine exhibited acceptable weak heterogeneity, and the results for other blood metabolites remained robust after sensitivity, heterogeneity, and pleiotropy testing. We conducted an enrichment analysis on potential blood metabolites, followed by a metabolic pathway analysis revealing four pathways associated with serum urate levels. Conclusion The identified causal relationships between these metabolites and serum urate and gout offer a novel perspective, providing new mechanistic insights into serum urate levels and gout.
Collapse
Affiliation(s)
- Yan Zhong
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - ChengAn Yang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - BingHua Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - YingWen Chen
- College of Integrated Chinese and Western Medicine, Tianjin University of Chinese Medicine, Tianjin, China
| | - WenJun Cai
- Department of Orthopaedic Center, The Third Clinical Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - GuoChen Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - ChangWei Zhao
- Department of Orthopedics Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - WenHai Zhao
- Department of Orthopedics Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
49
|
Liu W, Ye L, Hua B, Yang Y, Dong Z, Jiang Y, Li J, Sun X, Ye D, Wen C, Mao Y, He Z. Association between combined exposure to ambient air pollutants, genetic risk, and incident gout risk: A prospective cohort study in the UK Biobank. Semin Arthritis Rheum 2024; 66:152445. [PMID: 38579592 DOI: 10.1016/j.semarthrit.2024.152445] [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: 01/12/2024] [Revised: 03/02/2024] [Accepted: 03/17/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND Limited research has been conducted on the association between long-term exposure to air pollutants and the incidence of gout. OBJECTIVES This study aims to assess the individual and combined effects of prolonged exposure to five air pollutants (NO2, NOx, PM10, PMcoarse and PM2.52) on the incidence of gout among 458,884 initially gout-free participants enrolled in the UK Biobank. METHODS Employing a land use regression model, we utilized an estimation method to ascertain the annual concentrations of the five air pollutants. Subsequently, we devised a weighted air pollution score to facilitate a comprehensive evaluation of exposure. The Cox proportional hazards model was utilized to investigate the association between ambient air pollution and gout risk. Interaction and stratification analyses were conducted to evaluate age, sex, BMI, and genetic predisposition as potential effect modifiers in the air pollution-gout relationship. Furthermore, mediation analyses were conducted to explore the potential involvement of biomarkers in mediating the association between air pollution and gout. RESULTS Over a median follow-up time of 12.0 years, 7,927 cases of gout were diagnosed. Significant associations were observed between the risk of gout and a per IQR increase in NO2 (HR3: 1.05, 95 % CI4: 1.02-1.08, p = 0.003), NOx (HR: 1.04, 95 % CI: 1.01-1.06, p = 0.003), and PM2.5 (HR: 1.03, 95 % CI: 1.00-1.06, p = 0.030). Per IQR increase in the air pollution score was associated with an elevated risk of gout (p = 0.005). Stratified analysis revealed a significant correlation between the air pollution score and gout risk in participants ≥60 years (HR: 1.05, 95 % CI: 1.02-1.09, p = 0.005), but not in those <60 years (p = 0.793), indicating a significant interaction effect with age (p-interaction=0.009). Mediation analyses identified five serum biomarkers (SUA:15.87 %, VITD: 5.04 %, LDLD: 3.34 %, GGT: 1.90 %, AST: 1.56 %5) with potential mediation effects on this association. CONCLUSIONS Long-term exposure to air pollutants, particularly among the elderly population, is associated with an increased risk of gout. The underlying mechanisms of these associations may involve the participation of five serum biomarkers.
Collapse
Affiliation(s)
- Wei Liu
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Lihong Ye
- Department of Infection Prevention and Control, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, PR China
| | - Baojie Hua
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Yudan Yang
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Ziwei Dong
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Yuqing Jiang
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Jiayu Li
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Xiaohui Sun
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Ding Ye
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Chengping Wen
- Institute of Basic Research in Clinical Medicine, School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China
| | - Yingying Mao
- Department of Epidemiology, School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
| | - Zhixing He
- Institute of Basic Research in Clinical Medicine, School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, PR China.
| |
Collapse
|
50
|
Kasten A, Cascorbi I. Understanding the impact of ABCG2 polymorphisms on drug pharmacokinetics: focus on rosuvastatin and allopurinol. Expert Opin Drug Metab Toxicol 2024; 20:519-528. [PMID: 38809523 DOI: 10.1080/17425255.2024.2362184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/28/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION In addition to the well-established understanding of the pharmacogenetics of drug-metabolizing enzymes, there is growing data on the effects of genetic variation in drug transporters, particularly ATP-binding cassette (ABC) transporters. However, the evidence that these genetic variants can be used to predict drug effects and to adjust individual dosing to avoid adverse events is still limited. AREAS COVERED This review presents a summary of the current literature from the PubMed database as of February 2024 regarding the impact of genetic variants on ABCG2 function and their relevance to the clinical use of the HMG-CoA reductase inhibitor rosuvastatin and the xanthine oxidase inhibitor allopurinol. EXPERT OPINION Although there are pharmacogenetic guidelines for the ABCG2 missense variant Q141K, there is still some conflicting data regarding the clinical benefits of these recommendations. Some caution appears to be warranted in homozygous ABCG2 Q141K carriers when rosuvastatin is administered at higher doses and such information is already included in the drug label. The benefit of dose adaption to lower possible side effects needs to be evaluated in prospective clinical studies.
Collapse
Affiliation(s)
- Anne Kasten
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|