1
|
Zhong S, Xiao R, Lin Y, Xie B, Sun J. The impact of leisure sedentary behaviors on risk of chronic kidney disease, diabetes, and related complications: Mendelian randomization study. Ren Fail 2025; 47:2479177. [PMID: 40113344 PMCID: PMC11926908 DOI: 10.1080/0886022x.2025.2479177] [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: 10/31/2024] [Revised: 02/15/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The causal relationship between leisure sedentary behaviors (LSBs) and chronic kidney disease, diabetes and related complications is still equivocal. In this study, we performed two-sample Mendelian randomization for declaring the potential causal association between LSBs and these diseases and summarized the causal estimates. METHODS In this study, we used GWAS summary statistics from the public database for exposures (LSB: television watching, computer use, and driving) and outcomes (chronic kidney diseases, diabetes mellitus, and related complications). To ensure reliable results for this study, we applied several methods including IVW, MR-Egger, and weighted median for the regression process; MR-Egger intercept test, Cochran's Q test, 'leave-one-out' analysis and MR-PRESSO test were used to detect horizontal pleiotropy and heterogeneity for sensitivity analysis. RESULTS Television watching was harmful of CKD (OR = 1.26, 95%CI 1.09-1.44; p = 0.0011), T2D (OR = 1.82, 95%CI 1.48-2.24; p = 1.67e - 08) and DM (OR = 2.26, 95%CI 1.75-2.93; p = 6.44e - 10). No horizontal pleiotropy was detected in MR-Egger intercept test (p value > 0.05) and there were no influential SNPs based on 'leave-one-out' analysis. CONCLUSIONS Mendelian randomization estimates in our study genetically predicted the causal effect between television watching and CKD, T2D, and DM. However, we cannot get the definitive causal effect of television watching and other related complications, further studies need to be done to explore the mechanism of action of sedentary behavior on the complications of diabetes and chronic kidney disease.
Collapse
Affiliation(s)
- Shuo Zhong
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Rui Xiao
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Ying Lin
- Jinan Center for Disease Control and Prevention, Jinan, China
| | - Bo Xie
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Jing Sun
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
2
|
Qing J, Zhao Y, Wu J. The impact of rising peripheral blood naïve CD8 + T cell levels on chronic kidney disease onset: a Mendelian randomization study. Ren Fail 2025; 47:2486564. [PMID: 40230080 PMCID: PMC12001844 DOI: 10.1080/0886022x.2025.2486564] [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: 01/20/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The global incidence of chronic kidney disease (CKD) is rising rapidly. Immune cells play a crucial role in the onset and progression of CKD, however, the causal relationships and underlying immunological mechanisms remain incompletely elucidated. This deficiency hinders the development and application of early interventions and immunotherapies for CKD. METHODS In this study, we hypothesize that alterations in immune cell phenotypes (ICPs) in the blood may influence the onset of CKD. We collated Genome Wide Association Studies (GWAS) data for 731 ICPs, alongside summary data for CKD and estimated glomerular filtration rate (eGFR). Utilizing bidirectional mendelian randomization analysis (MR), we identified the impact of ICPs on the onset of CKD. RESULTS Preliminary MR analyses revealed three ICPs positively associated with CKD onset: the absolute number of CD45RA+ CD28- CD8+ T cells (p = 1.209 × 10-15, 95% CI: 1.0002-1.0003), the percentage of CD28+ CD45RA+ CD8+ T cells of total T cells (p = 5.831 × 10-6, 95% CI: 1.0028-1.0070), and the percentage of CD45RA- CD28- CD8+ T cells of total T cells (p = 4.292 × 10-5, 95% CI: 1.0005-1.0015). After conducting sensitivity and reverse MR analyses, only the percentage of CD28+ CD45RA+ CD8+ T cells (naïve CD8+ T Cells) was found to have a sufficiently robust causal impact on CKD. CONCLUSION We are the first to demonstrate a significant positive association between the percentage of naïve CD8+ T cells and CKD onset. This finding offers new insights for early prevention and treatment of CKD.
Collapse
Affiliation(s)
- Jianbo Qing
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiting Zhao
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junnan Wu
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Yang R, Xu S, Liu Q, Zhang X, He H, Xu Y, Chen L, Xing X, Yang J. Causal relationship between chronic kidney disease, renal function, and venous thromboembolism: a bidirectional Mendelian randomization study. Ren Fail 2025; 47:2496803. [PMID: 40321038 PMCID: PMC12054574 DOI: 10.1080/0886022x.2025.2496803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) and impaired renal function have been implicated in venous thromboembolism (VTE), but their causal relationships remain uncertain. This study employs Mendelian randomization (MR) to elucidate the potential bidirectional causal effects between CKD, renal function biomarkers, and VTE. METHODS We collated datasets from genome-wide association studies conducted among European individuals to perform MR analyses. The primary method utilized was the random-effect inverse variance-weighted (IVW) approach, with MR-Egger and the weighted median approaches employed as supplemental techniques. Several sensitivity studies were performed to assess the findings' robustness. RESULTS We identified a link between elevated serum creatinine levels and both VTE (OR: 1.14, 95% CI: 1.05-1.24, p = 0.001) and PE (OR: 1.20, 95% CI: 1.08-1.33, p = 0.001). After outlier removal and Bonferroni correction, the Cr-VTE association lost significance (p = 0.005). A suggestive causal relationship was found between eGFR and VTE (OR: 0.38, 95% CI: 0.20-0.73, p = 0.004), DVT (OR: 0.37, 95% CI: 0.16-0.87, p = 0.022), and PE (OR: 0.29, 95% CI: 0.12-0.66, p = 0.004). No causal effects of CKD or BUN on VTE or its subtypes were observed. Reverse causality inferences did not reveal any meaningful results. CONCLUSIONS This MR analysis provides evidence that elevated serum creatinine is associated with a higher risk of VTE and PE, while reduced eGFR may be a potential risk factor for VTE and its subtypes. These findings highlight the need for proactive monitoring and preventive strategies in individuals with impaired renal function. Further studies are warranted to confirm these associations and explore underlying mechanisms.
Collapse
Affiliation(s)
- Rongping Yang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shuanglan Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Qian Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xifeng Zhang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Huilin He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yue Xu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Linna Chen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiqian Xing
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Yunnan University, Kunming, China
- Key Laboratory of Respiratory Disease Research of Department of Education of Yunnan Province, Kunming, China
| | - Jiao Yang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
4
|
He J, Li L, Hu H. Causal associations between circulating metabolites and chronic kidney disease: a Mendelian randomization study. Ren Fail 2025; 47:2498090. [PMID: 40302304 PMCID: PMC12044913 DOI: 10.1080/0886022x.2025.2498090] [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: 12/08/2024] [Revised: 03/30/2025] [Accepted: 04/13/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Circulating metabolites have been associated with cross-sectional renal function in population-based research. Nevertheless, there is currently little proof to support the idea that metabolites either cause or prevent renal function. New treatment targets and ways to screen individuals with impaired renal function will be made possible via an in-depth analysis of the causal relationship between blood metabolites and renal function. METHODS We assessed the causal relationship between 452 serum metabolites and six renal phenotypes (CKD, rapid progression to CKD [CKDi25], rapid eGFR decline [CKD rapid3], dialysis, estimated glomerular filtration rate, and blood urea nitrogen) using univariate Mendelian randomization, primarily employing the inverse variance weighted method with robust sensitivity analyses. Heterogeneity and pleiotropy were examined via Cochrane's Q test and MR-Egger regression, and statistical significance was adjusted using Bonferroni correction. To assess potential adverse effects of metabolite modulation, we conducted a phenome-wide Mendelian randomization analysis, followed by multivariate Mendelian randomization to adjust for confounders. RESULTS We identified glycine and N-acetylornithine as potential causal mediators of CKD and renal dysfunction. Notably, lowering glycine levels may increase the risk of cholelithiasis and cholecystitis, while reducing N-acetylornithine could have unintended effects on tinnitus. CONCLUSION Glycine and N-acetylornithine represent promising therapeutic targets for CKD and renal function preservation, but their modulation requires careful risk-benefit assessment to avoid adverse effects.
Collapse
Affiliation(s)
- Jie He
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Lin Li
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
5
|
Wagner CA, Egli-Spichtig D, Rubio-Aliaga I. Updates on renal phosphate transport. Curr Opin Nephrol Hypertens 2025; 34:269-275. [PMID: 40357590 DOI: 10.1097/mnh.0000000000001090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW The kidneys control systemic phosphate balance by regulating phosphate transporters mediating the reabsorption of inorganic phosphate (Pi). At least three different Na + -driven Pi cotransporters are located in the brush border membrane (BBM) of proximal tubule cells, NaPi-IIa (SLC34A1), NaPi-IIc (SLC34A3) and PiT-2 (SLC20A2). This review will discuss novel aspects of their regulation, pharmacology, and genetics. RECENT FINDINGS Renal NaPi transporters are not only acutely regulated by the phosphaturic hormones parathyroid hormone (PTH) and Fibroblast Growth Factor 23 (FGF23) but possibly also by further mechanisms. A role of inositol hexakisphosphate (IP6) kinases has been found and their deletion from kidneys causes hypophosphatemia, hyperphosphaturia, and bone demineralization. Inhibitors of NaPis elicit phosphaturia and may reduce levels of PTH and FGF23 in chronic kidney disease (CKD) models. The relevance of renal NaPi transporters is highlighted by loss-of-function mutations in SLC34 transporters and analysis of patients provides new insights into diseases caused by variants. Major manifestations include nephrocalcinosis and -lithiasis, rickets, and variants may predispose to an accelerated decline in kidney function. SUMMARY Renal Pi transporters are regulated, may provide novel drug targets for prevention or treatment of hyperphosphatemia, and contribute to the genetic risk to develop kidney stones and CKD.
Collapse
|
6
|
Vosters TG, Stel VS, Jager KJ, Ferwerda B, Marsman RF, van Ittersum FJ, van den Born BJH, Galenkamp H, Vogt L, van Valkengoed IG. Performance of Current Chronic Kidney Disease Screening Criteria in Women and Men Across Ethnic Groups: The HELIUS Study. Mayo Clin Proc Innov Qual Outcomes 2025; 9:100613. [PMID: 40290568 PMCID: PMC12033983 DOI: 10.1016/j.mayocpiqo.2025.100613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Objective To investigate whether the currently recommended screening criteria in Kidney Disease: Improving Global Outcomes 2024 guidelines (hypertension, diabetes mellitus, and cardiovascular disease) equally detect women and men across ethnic groups and whether consideration of optional criteria (education level, occupation, obesity, and genetic risk factors) listed in the guideline improves performance. Patients and Methods We included 12,384 women and 9046 men of Dutch, South Asian and African Surinamese, Ghanaian, Turkish, and Moroccan origin from the baseline HELIUS Study (January 1, 2011, through December 31, 2015, Amsterdam, the Netherlands). Chronic kidney disease (CKD) was defined as estimated glomerular filtration rate of <60 mL/min/1.73 m2 or albumin-to-creatinine ratio of >3 mg/mmol. Poisson regression analyses estimated associations between CKD and optional criteria on top of current screening criteria. Model comparisons were made with likelihood ratio tests and Akaike information criterion estimations in women and men. Area under the curve (AUC), sensitivity, specificity, and positive and negative predictive values were calculated by sex and ethnicity. Results Chronic kidney disease prevalence ranged from 2.9% to 8.8% in women and 3.2% to 8.6% in men. Low educational level (women only) and obesity significantly improved the models with current criteria with CKD. High-risk occupations and polygenic risk score did not improve the model. However, these criteria did not improve predictive measures across ethnic groups. Overall, the AUCs for the current screening criteria were acceptable in men (AUC, 0.75; 95% CI, 0.73-0.77) and poor in women (AUC, 0.65; 95% CI, 0.63-0.67), and showed minimal change after adding the optional criteria. Conclusion Current screening criteria may not be equally detecting women and men across ethnic groups with CKD. Optional criteria had limited added value.
Collapse
Affiliation(s)
- Taryn G. Vosters
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Vianda S. Stel
- Department of Medical Informatics, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Public Health Research Institute, Quality of Care and Ageing Later Life, Amsterdam, Netherlands
| | - Kitty J. Jager
- Department of Medical Informatics, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Public Health Research Institute, Quality of Care and Ageing Later Life, Amsterdam, Netherlands
| | - Bart Ferwerda
- Department of Clinical Epidemiology, Biostatics and Bioinformatics, Amsterdam, Amsterdam UMC, University of Amsterdam, Netherlands
| | - Roos F. Marsman
- Section Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Amsterdam UMC, University of Amsterdam, Netherlands
| | - Frans J. van Ittersum
- Section Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University, Amsterdam, Netherlands
| | - Bert-Jan H. van den Born
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Internal and Vascular Medicine, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Henrike Galenkamp
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Liffert Vogt
- Section Nephrology, Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Amsterdam UMC, University of Amsterdam, Netherlands
| | - Irene G.M. van Valkengoed
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Þorsteinsson H, Baukmann HA, Sveinsdóttir HS, Halldórsdóttir DÞ, Grzymala B, Hillman C, Rolfe-Tarrant J, Parker MO, Cope JL, Ravarani CNJ, Schmidt MF, Karlsson KÆ. Validation of L-type calcium channel blocker amlodipine as a novel ADHD treatment through cross-species analysis, drug-target Mendelian randomization, and clinical evidence from medical records. Neuropsychopharmacology 2025; 50:1145-1155. [PMID: 39953207 PMCID: PMC12089589 DOI: 10.1038/s41386-025-02062-x] [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: 10/23/2024] [Revised: 01/09/2025] [Accepted: 01/28/2025] [Indexed: 02/17/2025]
Abstract
ADHD is a chronic neurodevelopmental disorder that significantly affects life outcomes, and current treatments often have adverse side effects, high abuse potential, and a 25% non-response rate, highlighting the need for new therapeutics. This study investigates amlodipine, an L-type calcium channel blocker, as a potential foundation for developing a novel ADHD treatment by integrating findings from animal models and human genetic data. Amlodipine reduced hyperactivity in SHR rats and decreased both hyperactivity and impulsivity in adgrl3.1-/- zebrafish. It also crosses the blood-brain barrier, reducing telencephalic activation. Crucially, Mendelian Randomization analysis linked ADHD to genetic variations in L-type calcium channel subunits (α1-C; CACNA1C, β1; CACNB1, α2δ3; CACNA2D3) targeted by amlodipine, while polygenic risk score analysis showed symptom mitigation in individuals with high ADHD genetic liability. With its well-tolerated profile and efficacy across species, supported by genetic evidence, amlodipine shows potential to be refined and developed into a novel treatment for ADHD.
Collapse
Affiliation(s)
| | | | | | | | | | - Courtney Hillman
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, UK
| | - Jude Rolfe-Tarrant
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, UK
| | - Matthew O Parker
- Surrey Sleep Research Centre, School of Biosciences, University of Surrey, Guildford, UK
| | | | | | | | - Karl Æ Karlsson
- 3Z, Menntavegur 1, Reykjavík, Iceland.
- Reykjavik University, Biomedical Engineering, Reykjavik, Iceland.
- Biomedical Center, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
8
|
Tran KN, Sutherland HG, Mallett AJ, Griffiths LR, Lea RA. New composite phenotypes enhance chronic kidney disease classification and genetic associations. PLoS Genet 2025; 21:e1011718. [PMID: 40408443 DOI: 10.1371/journal.pgen.1011718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
Chronic kidney disease (CKD) is a multifactorial condition driven by diverse etiologies that lead to a gradual loss of kidney function. Although genome-wide association studies (GWAS) have identified numerous genetic loci linked to CKD, a large portion of its genetic basis remains unexplained. This knowledge gap may partly arise from the reliance on single biomarkers, such as estimated glomerular filtration rate (eGFR), to assess kidney function. To address this limitation, we developed and applied a novel multi-phenotype approach, combinatorial Principal Component Analysis (cPCA), to better understand the complex genetic architecture of CKD. Using UK Biobank dataset (n = 337,112), we analyzed 21 CKD-related phenotypes, generating over 2 million composite phenotypes (CPs) through cPCA. Nearly 50,000 of these CPs demonstrated significantly higher classification power for clinical CKD compared to individual biomarkers. The top-ranked CP-a combination of albumin, cystatin C, eGFR, gamma-glutamyltransferase, HbA1c, low-density lipoprotein, and microalbuminuria, achieved an AUC of 0.878 (95% CI: 0.873-0.882), significantly outperforming eGFR alone (AUC: 0.830, 95% CI: 0.825-0.835). Genetic association analysis of the ~ 50,000 high-performing CPs identified all major eGFR-associated loci, except for the SH2B3 locus rs3184504, a loss-of-function variant, which was uniquely identified in CPs (p = 3.1[Formula: see text]10-56) but not in eGFR within the same sample size. In addition, SH2B3 locus showed strong evidence of colocalization with eGFR, supporting its role in kidney function. These results highlight the power of the multi-phenotype cPCA approach in understanding the genetic basis of CKD, with potential applications to other complex diseases.
Collapse
Affiliation(s)
- Kim Ngan Tran
- Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Heidi G Sutherland
- Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Andrew J Mallett
- Institute for Molecular Bioscience & Faculty of Medicine, The University of Queensland, St Lucia, Queensland, Australia
- Department of Renal Medicine, Townsville University Hospital, Townsville, Queensland, Australia
- College of Medicine & Dentistry, James Cook University, Townsville, Queensland, Australia
| | - Lyn R Griffiths
- Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Rodney A Lea
- Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| |
Collapse
|
9
|
Noels H, van der Vorst EPC, Rubin S, Emmett A, Marx N, Tomaszewski M, Jankowski J. Renal-Cardiac Crosstalk in the Pathogenesis and Progression of Heart Failure. Circ Res 2025; 136:1306-1334. [PMID: 40403103 DOI: 10.1161/circresaha.124.325488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/14/2025] [Accepted: 03/11/2025] [Indexed: 05/24/2025]
Abstract
Chronic kidney disease (CKD) represents a global health issue with a high socioeconomic impact. Beyond a progressive decline of kidney function, patients with CKD are at increased risk of cardiovascular diseases, including heart failure (HF) and sudden cardiac death. HF in CKD can manifest both as HF with reduced ejection fraction and HF with preserved ejection fraction, with the latter further increasing in relative importance in the more advanced stages of CKD. Typical cardiac remodeling characteristics in uremic cardiomyopathy include left ventricular hypertrophy, myocardial fibrosis, cardiac electrical dysregulation, capillary rarefaction, and microvascular dysfunction, which are triggered by increased cardiac preload, cardiac afterload, and preload and afterload-independent factors. The pathophysiological mechanisms underlying cardiac remodeling in CKD are multifactorial and include neurohormonal activation (with increased activation of the renin-angiotensin-aldosterone system, the sympathetic nervous system, and mineralocorticoid receptor signaling), cardiac steroid activation, mitochondrial dysfunction, inflammation, innate immune activation, and oxidative stress. Furthermore, disturbances in cardiac metabolism and calcium homeostasis, macrovascular and microvascular dysfunction, increased cellular profibrotic responses, the accumulation of uremic retention solutes, and mineral and bone disorders also contribute to cardiovascular disease and HF in CKD. Here, we review the current knowledge of HF in CKD, including the clinical characteristics and pathophysiological mechanisms revealed in animal studies. We also elaborate on the detrimental impact of comorbidities of CKD on HF using hypertension as an example and discuss the clinical characteristics of hypertensive heart disease and the genetic predisposition. Overall, this review aims to increase the understanding of HF in CKD to support future research and clinical translational approaches for improved diagnosis and therapy of this vulnerable patient population.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Biochemistry Department (H.N.), Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Interdisciplinary Center for Clinical Research (IZKF) (E.P.C.v.d.V.), RWTH Aachen University, Germany
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (E.P.C.v.d.V.)
| | - Sébastien Rubin
- L'Institut national de la santé et de la recherche médicale (INSERM), BMC, U1034, University of Bordeaux, Pessac, France (S.R.)
- Renal Unit, University Hospital of Bordeaux, France (S.R.)
| | - Amber Emmett
- Faculty of Medicine, Biology and Health, Division of Cardiovascular Sciences, The University of Manchester, United Kingdom (A.E., M.T.)
| | - Nikolaus Marx
- Department of Internal Medicine I-Cardiology, Angiology and Internal Intensive Care Medicine (N.M.), RWTH Aachen University, Germany
| | - Maciej Tomaszewski
- Faculty of Medicine, Biology and Health, Division of Cardiovascular Sciences, The University of Manchester, United Kingdom (A.E., M.T.)
- British Heart Foundation Manchester Centre of Research Excellence, United Kingdom (M.T.)
- Manchester Academic Health Science Centre, Manchester University National Health Service (NHS) Foundation Trust, United Kingdom (M.T.)
- Signature Research Programme in Health Services and Systems Research, Duke-National University of Singapore (M.T.)
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Biochemistry Department (H.N.), Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
- Pathology Department (J.J.), Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| |
Collapse
|
10
|
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
|
11
|
Le NN, Tran TQB, McClure J, Gill D, Padmanabhan S. Emerging antihypertensive therapies and cardiovascular, kidney, and metabolic outcomes: a Mendelian randomization study. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2025; 11:264-274. [PMID: 39963705 PMCID: PMC12046581 DOI: 10.1093/ehjcvp/pvaf015] [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: 11/30/2024] [Revised: 12/18/2024] [Accepted: 02/14/2025] [Indexed: 05/03/2025]
Abstract
AIMS Emerging antihypertensive drug classes offer new opportunities to manage hypertension; however, their long-term effects on cardiovascular, kidney, and metabolic (CKM) outcomes remain to be elucidated. This study aims to explore the effects of phosphodiesterase type 5 inhibitors (PDE5i), soluble guanylate cyclase stimulators (sGCs), endothelin receptor antagonists (ERAs), and angiotensinogen inhibitors (AGTis) on a range of CKM outcomes. METHODS AND RESULTS Mendelian randomization (MR), summary-based MR (SMR), and colocalization analyses were applied to assess the drug effect on coronary artery disease (CAD), myocardial infarction (MI), ischaemic stroke, atrial fibrillation (AF), heart failure (HF), type 2 diabetes (T2D), and chronic kidney disease (CKD). Genetic association and gene expression summary data were obtained from the largest European-ancestry genome-wide association studies (GWAS) and the genotype-tissue expression version 8 for 29 tissues relevant to the outcomes' pathophysiology.Genetically predicted systolic blood pressure (SBP) reduction was associated with reduced risks of all outcomes. PDE5i was associated with reduced risks of CAD (OR per 10-mmHg decrease in SBP: 0.348[95% confidence interval (CI): 0.199-0.607]) and ischaemic stroke (0.588[0.453-0.763]). sGCs showed protective effects against CAD (0.332[0.236-0.469]), MI (0.238[0.168-0.337]), and CKD (0.55[0.398-0.761]). ERA and AGTi showed protective effects against CAD and ischaemic stroke. SMR and colocalization supported the association of gene expression levels of GUCY1A3 and PDE5A with CAD and MI risk. CONCLUSION Our study highlights the potential of PDE5i, sGCs, ERA, and AGTi in reducing cardiovascular and renal risks. These findings underscore the necessity for targeted clinical trials to validate the efficacy and safety of these therapies.
Collapse
Affiliation(s)
- Nhu Ngoc Le
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Tran Quoc Bao Tran
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - John McClure
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Sandosh Padmanabhan
- BHF Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
12
|
Cho JM, Kim M, Oh J, Koh JH, Cho S, Kim Y, Lee S, Kim K, Kim YC, Han SS, Joo KW, Kim YS, Lee H, Kim DK, Park S. Causal Effects From Kidney Function to Plasma Proteome: Integrated Observational and Mendelian Randomization Analysis With >50,000 UK Biobank Participants. Proteomics Clin Appl 2025; 19:e70002. [PMID: 40014632 DOI: 10.1002/prca.70002] [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: 10/11/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Chronic kidney disease (CKD) causes detrimental systemic effects, including inflammation or apoptosis, which lead to substantial morbidity and mortality. However, the causal effect of reduced kidney function on systemic proteomic signatures is incompletely understood. METHODS We performed an integrated Mendelian randomization (MR) and observational analyses to identify the causal association between kidney function and plasma protein levels, based on 1815 plasma protein profiles in 50,407 UK Biobank participants and the CKDGen Phase 4 genome-wide association study (GWAS) meta-analysis for the genetic instruments of eGFR. RESULTS The MR analysis revealed 383 plasma proteins causally associated with eGFR. Reduced kidney function was found to be causally associated with an increase in the plasma levels of 381 proteins, among which TNF and IGFBP4 were increased, while the level of two proteins, NPHS1 and SPOCK1, decreased. Apoptosis-related pathway was significantly enriched in the gene-set enrichment analysis. In network analysis, TNF was identified as a hub protein with multiple linkages to molecules included in the TNF-signaling pathways, involved in inflammation, fibrosis, and apoptosis. CONCLUSIONS In this proteo-genomic analysis, we identified 383 plasma proteins causally associated with eGFR, highlighting TNF-associated pathways as pathologically relevant processes in kidney disease progression, systemic inflammation, and organ fibrosis, warranting further investigation.
Collapse
Affiliation(s)
- Jeong Min Cho
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, South Korea
| | - Minsang Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jaeik Oh
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jung Hun Koh
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Semin Cho
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, South Korea
| | - Yaerim Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
| | - Soojin Lee
- Department of Internal Medicine, Uijeongbu Eulji University Medical Center, Seoul, South Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Kwon-Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Ki Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Sehoon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
13
|
Lu Z, Ni W, Wu Y, Zhai B, Zhao Q, Zheng T, Liu Q, Ding D. Application of biomarkers in the diagnosis of kidney disease. Front Med (Lausanne) 2025; 12:1560222. [PMID: 40370722 PMCID: PMC12075424 DOI: 10.3389/fmed.2025.1560222] [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: 01/14/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Worldwide, kidney disease has grown to be an important global public health agenda that reduces longevity. Medical institutions around the globe should enhance screening efforts for kidney disease, to facilitate early kidney disease detection, diagnosis, and intervention. Common screening methods for nephropathy encompass renal tissue biopsy, urine dry chemistry tests, urine formed element analysis, and urine-specific protein assays, among others. These methodologies evaluate renal health by scrutinizing a spectrum of biomarkers. Precise classification and quantitative analysis of these biomarkers can assist in determining the site and extent of kidney injury, as well as in assessing treatment efficacy and prognosis. In this paper, we reviewed the methods and biomarkers for kidney disease and also the integration of multiple biomarkers. With the aim of reasonable applying these markers to the early detection, accurate diagnosis, and scientific management of kidney disease, thereby mitigating the threat posed by kidney disease to human health.
Collapse
Affiliation(s)
- Zuohua Lu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Weifeng Ni
- Department of Endocrinology, Rheumatology and Immunology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yuding Wu
- Goldsite Diagnostics Inc., Shenzhen, China
| | - Bin Zhai
- Department of Clinical Laboratory, Baotou Central Hospital, Baotou, China
| | - Qiuyun Zhao
- Department of Clinical Laboratory, Guilin Hospital of Integrated Traditional Chinese and Western Medicine, Guilin, China
| | - Tian Zheng
- Department of Clinical Laboratory, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qianqian Liu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Dapeng Ding
- Department of Clinical Laboratory, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
Schmidt AF, Finan C, van Setten J, Puyol-Antón E, Ruijsink B, Bourfiss M, Alasiri AI, Velthuis BK, Asselbergs FW, Te Riele ASJM. A Mendelian randomization analysis of cardiac MRI measurements as surrogate outcomes for heart failure and atrial fibrillation. COMMUNICATIONS MEDICINE 2025; 5:130. [PMID: 40253538 PMCID: PMC12009341 DOI: 10.1038/s43856-025-00855-1] [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: 06/21/2023] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Drug development and disease prevention of heart failure (HF) and atrial fibrillation (AF) are impeded by a lack of robust early-stage surrogates. We determined to what extent cardiac magnetic resonance (CMR) measurements act as surrogates for the development of HF or AF. METHODS Genetic data were sourced on the association with 21 atrial and ventricular CMR measurements. Mendelian randomization was used to determine CMR associations with AF, HF, non-ischaemic cardiomyopathy (NICM), and dilated cardiomyopathy (DCM), noting that the definition of NICM includes DCM as a subset. Additionally, for the CMR surrogates of AF and HF, we explored their association with non-cardiac traits potentially influenced by cardiac disease liability. RESULTS In total we find that 7 CMR measures (biventricular ejection fraction (EF) and end-systolic volume (ESV), as well as LV systolic volume (SV), end-diastolic volume (EDV), and mass to volume ratio (MVR)) associate with the development of HF, 5 with the development of NICM (biventricular EDV and ESV, LV-EF), 7 with DCM (biventricular EF, ESV, EDV, and LV end-diastolic mass (EDM), and 3 associate with AF (LV-ESV, RV-EF, RV-ESV). Higher EF of both ventricles associate with lower risk of HF and DCM, with biventricular ESV associating with all four cardiac outcomes. Higher values of biventricular EDV associate with lower risk of HF, and DCM. Exploring the associations of these CMR cardiac disease surrogates with non-cardiac traits confirms a strong link with diastolic blood pressure, as well as more specific associations with lung function (LV-ESV), HbA1c (LV-EDM), and type 2 diabetes (LV-SV). CONCLUSIONS The current paper identifies key CMR measurements that may act as surrogate endpoints for the development of HF (including NICM and DCM) or AF.
Collapse
Affiliation(s)
- A F Schmidt
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK.
- UCL BHF Research Accelerator Centre, London, UK.
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | - C Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL BHF Research Accelerator Centre, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - J van Setten
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - E Puyol-Antón
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London, UK
| | - B Ruijsink
- School of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - M Bourfiss
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - A I Alasiri
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Medical Genomics Research Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - B K Velthuis
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - F W Asselbergs
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Institute of Health Informatics, Faculty of Population Health, University College London, London, UK
| | - A S J M Te Riele
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| |
Collapse
|
15
|
D’Antonio M, Arthur TD, Gonzalez Rivera WG, Wu X, Nguyen JP, Gymrek M, Woo-Yeong P, Frazer KA. Genetic analysis of elevated levels of creatinine and cystatin C biomarkers reveals novel genetic loci associated with kidney function. Hum Mol Genet 2025; 34:751-764. [PMID: 39927731 PMCID: PMC12010162 DOI: 10.1093/hmg/ddaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025] Open
Abstract
The rising prevalence of chronic kidney disease (CKD), affecting an estimated 37 million adults in the United States, presents a significant global health challenge. CKD is typically assessed using estimated Glomerular Filtration Rate (eGFR), which incorporates serum levels of biomarkers such as creatinine and cystatin C. However, these biomarkers do not directly measure kidney function; their elevation in CKD results from diminished glomerular filtration. Genome-wide association studies (GWAS) based on eGFR formulas using creatinine (eGFRcre) or cystatin C (eGFRcys) have identified distinct non-overlapping loci, raising questions about whether these loci govern kidney function or biomarker metabolism. In this study, we show that GWAS on creatinine and cystatin C levels in healthy individuals reveal both nonoverlapping genetic loci impacting their metabolism as well as overlapping genetic loci associated with kidney function; whereas GWAS on elevated levels of these biomarkers uncover novel loci primarily associated with kidney function in CKD patients.
Collapse
Affiliation(s)
- Matteo D’Antonio
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Timothy D Arthur
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Wilfredo G Gonzalez Rivera
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Ximei Wu
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Jennifer P Nguyen
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Melissa Gymrek
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
- Department of Computer Science and Engineering, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
- Department of Medicine, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, United States
| | - Park Woo-Yeong
- Division of Nephrology, Department of Internal Medicine, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, 1035 Dalgubeol-daero, Daegu, Republic of Korea
| | - Kelly A Frazer
- Department of Pediatrics, University of California San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, United States
- Institute of Genomic Medicine, University of California San Diego, 9500 Gilman Dr, 9500 Gilman Dr., La Jolla, CA 92093, United States
| |
Collapse
|
16
|
Yu Y, Li J, Yu B, Yu Y, Sun Y, Wang Y, Wang B, Zhang K, Tang M, Lu Y, Wang N. The Identification of Biomarkers and Therapeutic Targets for Diabetic Kidney Disease by Integrating the Proteome with the Genome. Biomedicines 2025; 13:971. [PMID: 40299563 PMCID: PMC12025092 DOI: 10.3390/biomedicines13040971] [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: 02/28/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Background: The blood proteome is a major source of biomarkers and therapeutic targets. We conducted a proteome-wide Mendelian randomization (MR) study to identify cardiometabolic protein markers for diabetic kidney disease (DKD). Methods: We measured all 369 proteins in the Olink Explore 384 Cardiometabolic and Cardiometabolic panel of 500 patients with type 2 diabetes from 11 communities in Shanghai. Protein quantitative trait loci (pQTLs) were derived by coupling genomic and proteomic data. Cis-pQTLs identified for proteins were used as instrumental variables in MR analyses of DKD risk, and the outcome data were obtained from 8401 Japanese individuals with type 2 diabetes (2809 cases and 5592 controls). Replication MR analysis was performed in the UK Biobank Pharma Proteomics Project (UKB-PPP). Colocalization analysis and the Heidi test were used to examine whether the identified proteins and DKD shared causal variants. Results: Among the 369 proteins, we identified 66 independent cis-pQTLs for 64 proteins. MR analysis suggested that two cardiometabolic proteins (UMOD and SIRPA) may play a causal role in increasing DKD risk, with UMOD showing replication in UKB-PPP. Bayesian colocalization further supported the causal effects of these proteins. Additional analyses indicated that UMOD is highly expressed in renal macrophages. Further downstream analyses suggested that UMOD could be a potential novel target and that SIRPA could be a potential repurposing target for DKD; however, further validation is needed. Conclusions: By integrating proteomic and genetic data from patients with type 2 diabetes, we identified two protein biomarkers potentially associated with DKD risk. These findings provide insights into DKD pathophysiology and therapeutic target development, but further replication and functional studies are needed to confirm these associations.
Collapse
Affiliation(s)
- Yuefeng Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Jiang Li
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Bowei Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Yuetian Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Ying Sun
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Yuying Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Bin Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Mengjun Tang
- The 967th Hospital of Joint Logistic Support Force of People’s Liberation Army, Dalian 116011, China;
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.Y.); (J.L.)
| |
Collapse
|
17
|
Lundin R, Melotti R, Barin L, Gögele M, Lombardo S, Fanolla A, Zuech P, Rainer J, Emmert D, Fuchsberger C, Mascalzoni D, De Grandi A, Domingues FS, Hicks AA, Pramstaller PP, Pattaro C. Cohort Profile: the Cooperative Health Research in South Tyrol study. Int J Epidemiol 2025; 54:dyaf064. [PMID: 40436620 PMCID: PMC12119133 DOI: 10.1093/ije/dyaf064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/07/2025] [Indexed: 06/01/2025] Open
Affiliation(s)
- Rebecca Lundin
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | | | - Laura Barin
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
- Centre for Medical Sciences, CISMed, University of Trento, Trento, Italy
| | - Martin Gögele
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Stefano Lombardo
- Provincial Institute for Statistics of the Autonomous Province of Bolzano-South Tyrol (ASTAT), Bolzano, Italy
| | - Antonio Fanolla
- Observatory for Health—Autonomous Province of Bolzano-South Tyrol, Bolzano, Italy
| | - Paola Zuech
- Observatory for Health—Autonomous Province of Bolzano-South Tyrol, Bolzano, Italy
| | | | - David Emmert
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | | | - Deborah Mascalzoni
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
- Center for Research Ethics and Bioethics, Uppsala University, Uppsala, Sweden
| | | | | | - Andrew A Hicks
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | | | | |
Collapse
|
18
|
Liu M, Liu Y, Sun M, Zeng Z, Song Y, Jia E, Wu S. Sedentary lifestyle and hallux valgus: Unraveling the causal pathways and the mediating role of calcium homeostasis through mendelian randomization. J Foot Ankle Surg 2025:S1067-2516(25)00112-7. [PMID: 40209937 DOI: 10.1053/j.jfas.2025.04.003] [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: 12/04/2024] [Revised: 03/29/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
The causal relationships between hallux valgus (HV), sedentary behavior and calcium homeostasis remain unclear. This study aimed to explore these associations using Mendelian randomization (MR) analysis. Leisure screen time (LST) was used as an indicator of sedentary behavior, while seven traits and three diseases were selected to represent calcium homeostasis. Two-sample MR was performed to assess the causal effect of sedentary behavior and calcium homeostasis on HV. Two-step MR was conducted to quantify the mediating roles of calcium homeostasis-related traits in the association between sedentary behavior and HV. Our results showed that longer LST was strongly associated with higher risk of HV (odds ratio (OR) = 1.1902, 95 %CI = 1.0129-1.3986, p = 0.0343). By contrast, serum calcium (S-Ca) levels were negatively associated with HV risk (OR = 0.7530, 95 %CI = 0.5675-0.9992, p = 0.0494). Mediation analyses found that S-Ca played an important mediating role in the effect of LST on HV (proportion mediated = 10.4 %). Our results extend the understanding of the pathogenesis of HV, and highlight the importance of preoperative metabolic optimization and postoperative behavioral and calcium supplementation strategies to enhance surgical outcomes and mitigate recurrence.
Collapse
Affiliation(s)
- Maolin Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
| | - Yan Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
| | - Miao Sun
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
| | - Zhongyao Zeng
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
| | - Yuanzhi Song
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
| | - Erlong Jia
- Department of Orthopedics, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, PR China.
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China.
| |
Collapse
|
19
|
Jin X, Wang Y, Zeng S, Cai J, Wang K, Ge Q, Zhang L, Li X, Zhang L, Tong Y, Luo X, Yang M, Zhang W, Yu C, Xiao C, Liu Z. Preschool age-specific obesity and later-life kidney health: a Mendelian randomization and colocalization study. Int J Obes (Lond) 2025; 49:649-657. [PMID: 39572765 DOI: 10.1038/s41366-024-01686-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 04/17/2025]
Abstract
OBJECTIVES While the association between obesity and kidney diseases has been found in previous studies, the relationship between preschool-age obesity and later-life kidney health remains unclear, posing challenges for effective interventions in this critical life period. METHODS Utilizing the hitherto largest genome-wide association studies, we conducted two-sample mendelian randomization (MR) to estimate the association of preschool age-specific obesity on kidney health and diseases, including blood urea nitrogen (BUN), eGFRcrea, eGFRcys, chronic kidney disease (CKD), IgA nephropathy, and diabetic nephropathy. Then, we applied multivariable Mendelian randomization (MVMR) and stepwise MR to elucidate the role of adult obesity and 12 other potential factors in the pathway between preschool age-specific obesity and kidney health. Finally, we employed colocalization analysis to understand the mechanism of preschool age-specific obesity and kidney damage further by detecting shared causal variants. RESULTS Our two-sample MR results indicated that preschool obesity could be associated with kidney health and disease. In addition, we observed a switch in the direction of associations between age-specific body mass index (BMI) and CKD, manifesting as negative associations before 3 years old and positive associations after 3 years old. Furthermore, MVMR and stepwise MR results suggested potential pathways linking preschool obesity to kidney health, involving factors such as adult BMI, circulating high-density lipoprotein cholesterol levels, and circulating C-reactive protein levels. Finally, we detected that preschool-age BMI and kidney function could share causal variants such as rs76111507, rs62107261, rs77165542 in the region of chromosome 2, and rs571312 in the region of chromosome 18. CONCLUSION Our study supports the association between preschool obesity and kidney health, emphasizing the role of adult BMI in this relationship. These findings underscore the importance of interventions starting in early childhood and continuing through adulthood to reduce the long-term risk of obesity-related kidney damage.
Collapse
Affiliation(s)
- Xin Jin
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yujue Wang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sixuan Zeng
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiarui Cai
- Faculty of Medicine, Imperial College London, London, UK
| | - Kerui Wang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiaoyue Ge
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Zhang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xinxi Li
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ling Zhang
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
- Med-X center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Yu Tong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoli Luo
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Menghan Yang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weidong Zhang
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuan Yu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Chenghan Xiao
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhenmi Liu
- Department of Maternal and Child Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
Yang X, Xiao R, Liu B, Xie B, Yang Z. The causal relationship of inflammation-related factors with osteoporosis: A Mendelian Randomization Analysis. Exp Gerontol 2025; 202:112715. [PMID: 39983802 DOI: 10.1016/j.exger.2025.112715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/10/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND We used Mendelian randomization (MR) approach to examine whether genetically determined inflammation-related risk factors play a role in the onset of osteoporosis (OP) in the European population. METHODS Genome-wide association studies (GWASs) summary statistics of estimated bone mineral density (eBMD) obtained from the public database GEnetic Factors for OSteoporosis Consortium (GEFOS) including 142,487 European people. For exposures, we utilized GWAS data of 9 risk factors including diseases chronic kidney disease (CKD) (41,395 cases and 439,303 controls), type 2 diabetes (T2D) (88,427 cases and 566,778 controls), Alzheimer's disease (AD) (71,880 cases, 383,378 controls) and major depression disorder (MDD) (9240 cases and 9519 controls) and lifestyle behaviors are from different consortiums. Inverse variance weighted (IVW) analysis was principal method in this study and random effect model was applied; MR-Egger method and weighted median method were also performed for reliable results. Cochran's Q test and MR-Egger regression were used to detect heterogeneity and pleiotropy and leave-one-out analysis was performed to find out whether there are influential SNPs. RESULTS We found that T2D (IVW: β = 0.05, P = 0.0014), FI (IVW: β = -0.22, P < 0.001), CKD (IVW: β = 0.02, P = 0.009), ALZ (IVW: β = 0.06, P = 0.005), Coffee consumption (IVW: β = 0.11, P = 0.003) were causally associated with OP (P<0.006after Bonferroni correction). CONCLUSIONS Our study revealed that T2D, FI, CKD, ALZ and coffee consumption are causally associated with OP. Future interventions targeting factors above could provide new clinical strategies for the personalized prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Xinyue Yang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Rui Xiao
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, China
| | - Bo Xie
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China.
| | - Zhao Yang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China.
| |
Collapse
|
21
|
Lyu Y, Li H, Liu X, Zhang X, Chen Y, Fan G, Zhang H, Han Z, Guo Z, Weng H, Hu H, Li X, Zhang Z, Zhang Y, Xu F, Wang C, Wang D, Yang P, Zhai Z. Estimated Glomerular Filtration Rate Decline is Causally Associated with Acute Pulmonary Embolism: A Nested Case-Control and Mendelian Randomization Study. Thromb Haemost 2025. [PMID: 39401521 DOI: 10.1055/a-2439-5200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2025]
Abstract
Renal dysfunction is highly prevalent among patients with pulmonary embolism (PE). This study combined population-based study and Mendelian randomization (MR) to observe the relationship between renal function and PE.A nested case-control study were performed using data of PE patients and controls were from two nationwide cohorts, the China pUlmonary thromboembolism REgistry Study (CURES) and China Health and Retirement Longitudinal Survey (CHARLS). Baseline characteristics were balanced using propensity score matching and inverse probability of treatment weighting. Restricted cubic spline models were applied for the relationship between estimated glomerular filtration rate (eGFR) decline and the risk of PE. Bidirectional two-sample MR analyses were performed using genome-wide association study summary statistics for eGFR involving 1,201,909 individuals and for PE from the FinnGen consortium.The nested case-control study including 17,547 participants (6,322 PE patients) found that eGFR distribution was significantly different between PE patients and controls (p < 0.001), PE patients had a higher proportion of eGFR < 60 mL/min/1.73 m2. eGFR below 88 mL/min/1.73 m2 was associated with a steep elevation in PE risk. MR analyses indicated a potential causal effect of eGFR decline on PE (odds ratio = 4·26, 95% confidence interval: 2·07-8·79), with no evidence of horizontal pleiotropy and reverse causality.Our findings support the hypothesis that renal function decline contributes to an elevated PE risk. Together with the high prevalence of chronic kidney diseases globally, there arises the necessity for monitoring and modulation of renal function in effective PE prevention.
Collapse
Affiliation(s)
- Yanshuang Lyu
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Haobo Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xin Liu
- Department of Nephrology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Xiaomeng Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yinong Chen
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Guohui Fan
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Data and Project Management Unit, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Hong Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhifa Han
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Zhuangjie Guo
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Haoyi Weng
- Shenzhen WeGene Clinical Laboratory, Shenzhen, China
- Wegene Shenzhen Zaozhidao Technology Co., Ltd, Shenzhen, China
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and ngineering, Central South University, Changsha, China
| | - Huiyuan Hu
- First Clinical College, Xi'an Jiaotong University, Xi'an, ShaanXi, China
| | - Xincheng Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhu Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yu Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- China China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Feiya Xu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Chen Wang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- Data and Project Management Unit, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
- First Clinical College, Xi'an Jiaotong University, Xi'an, ShaanXi, China
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- China China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Dingyi Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Data and Project Management Unit, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhenguo Zhai
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- Data and Project Management Unit, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
- First Clinical College, Xi'an Jiaotong University, Xi'an, ShaanXi, China
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- China China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Fang X, Zhong Y, Zheng R, Wu Q, Liu Y, Zhang D, Wang Y, Ding W, Wang K, Zhong F, Lin K, Yao X, Hu Q, Li X, Xu G, Liu N, Nie J, Li D, Geng H, Guan Y. PPDPF preserves integrity of proximal tubule by modulating NMNAT activity in chronic kidney diseases. SCIENCE ADVANCES 2025; 11:eadr8648. [PMID: 40106551 PMCID: PMC11922016 DOI: 10.1126/sciadv.adr8648] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/24/2024] [Indexed: 03/22/2025]
Abstract
Genome-wide association studies (GWAS) have identified loci associated with kidney diseases, but the causal variants, genes, and pathways involved remain elusive. Here, we identified a kidney disease gene called pancreatic progenitor cell differentiation and proliferation factor (PPDPF) through integrating GWAS on kidney function and multiomic analysis. PPDPF was predominantly expressed in healthy proximal tubules of human and mouse kidneys via single-cell analysis. Further investigations revealed that PPDPF functioned as a thiol-disulfide oxidoreductase to maintain cellular NAD+ levels. Deficiency in PPDPF disrupted NAD+ and mitochondrial homeostasis by impairing the activities of nicotinamide mononucleotide adenylyl transferases (NMNATs), thereby compromising the function of proximal tubules during injuries. Consequently, knockout of PPDPF notably accelerated the progression of chronic kidney disease (CKD) in mouse models induced by aging, chemical exposure, and obstruction. These findings strongly support targeting PPDPF as a potential therapy for kidney fibrosis, offering possibilities for future CKD interventions.
Collapse
Affiliation(s)
- Xiaoliang Fang
- Department of Urology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Yi Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Rui Zheng
- Department of Urology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Qihui Wu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Yu Liu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Dexin Zhang
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuwei Wang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Wubing Ding
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kaiyuan Wang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fengbo Zhong
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kai Lin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiaohui Yao
- Qingdao Innovation and Development Center, Harbin Engineering University, Qingdao, Shandong, 266000, China
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, Heilongjiang, 150001, China
| | - Qingxun Hu
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiaofei Li
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, 17164, Sweden
| | - Guofeng Xu
- Department of Pediatric Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jing Nie
- Biobank of Peking University First Hospital, Peking University First Hospital, Peking University, Beijing, 100034, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Hongquan Geng
- Department of Urology, Children’s Hospital of Fudan University, Shanghai, 201102, China
| | - Yuting Guan
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China
| |
Collapse
|
23
|
Wuttke M, Pattaro C. GWAS scorecard prioritizes kidney genes using coding and regulatory variants. Nat Rev Nephrol 2025:10.1038/s41581-025-00952-3. [PMID: 40113942 DOI: 10.1038/s41581-025-00952-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Affiliation(s)
- Matthias Wuttke
- Institute of Genetic Epidemiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|
24
|
Goovaerts S, Naqvi S, Hoskens H, Herrick N, Yuan M, Shriver MD, Shaffer JR, Walsh S, Weinberg SM, Wysocka J, Claes P. Enhanced insights into the genetic architecture of 3D cranial vault shape using pleiotropy-informed GWAS. Commun Biol 2025; 8:439. [PMID: 40087503 PMCID: PMC11909261 DOI: 10.1038/s42003-025-07875-6] [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: 06/11/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Large-scale GWAS studies have uncovered hundreds of genomic loci linked to facial and brain shape variation, but only tens associated with cranial vault shape, a largely overlooked aspect of the craniofacial complex. Surrounding the neocortex, the cranial vault plays a central role during craniofacial development and understanding its genetics are pivotal for understanding craniofacial conditions. Experimental biology and prior genetic studies have generated a wealth of knowledge that presents opportunities to aid further genetic discovery efforts. Here, we use the conditional FDR method to leverage GWAS data of facial shape, brain shape, and bone mineral density to enhance SNP discovery for cranial vault shape. This approach identified 120 independent genomic loci at 1% FDR, nearly tripling the number discovered through unconditioned analysis and implicating crucial craniofacial transcription factors and signaling pathways. These results significantly advance our genetic understanding of cranial vault shape and craniofacial development more broadly.
Collapse
Affiliation(s)
- Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
| | - Sahin Naqvi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Genetics and Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hanne Hoskens
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
- Department of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research, Institute, University of Calgary, Calgary, AB, Canada
| | - Noah Herrick
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meng Yuan
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Mark D Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - John R Shaffer
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
| | - Seth M Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anthropology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium.
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| |
Collapse
|
25
|
Zoccali C, Mallamaci F, Rosenberg K, Unwin R, Silva PI, Simeoni MA, Hafez G, Capasso G, Nitsch D. Big databases and biobanks for studying the links between CKD, cognitive impairment, and dementia. Nephrol Dial Transplant 2025; 40:ii37-ii45. [PMID: 40080089 PMCID: PMC11905747 DOI: 10.1093/ndt/gfae255] [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: 07/01/2024] [Indexed: 03/15/2025] Open
Abstract
Research on cognitive function in individuals with chronic kidney disease (CKD) is critical due to the significant public health challenge posed by both CKD and cognitive impairment. CKD affects approximately 10-15% of the adult population, with higher prevalence in the elderly, who are already at increased risk for cognitive decline. Cognitive impairment is notably higher in CKD patients, particularly those with severe stages of the disease, and progresses more rapidly in those on dialysis. This review explores how data from large biobank studies such as the Alzheimer's Disease Neuroimaging Initiative, UK Biobank, and others could be used to enhance understanding the progression and interplay between CKD and cognitive decline. Each of these data sources has specific strengths and limitations. Strengths include large sample sizes and longitudinal data across different groups, and in different settings. Addressing limitations leads to challenges in dealing with heterogeneous data collection methods, and addressing missing data, which requires the use of sophisticated statistical techniques. Combining data from multiple databases can mitigate individual study limitations, particularly via the 'epidemiological triangulation' concept. Using such data appropriately holds immense potential to better understand the pathobiology underlying CKD and cognitive impairment. Addressing the inherent challenges with a clear strategy is crucial for advancing our understanding and improving the lives of those affected by both CKD and cognitive impairment.
Collapse
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, New York, NY, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino (AV), Italy
- Associazione Ipertensione Nefrologia Trapianto Renal (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Francesca Mallamaci
- IFC-CNR, Institute of Clinical Physiology of Reggio Calabria, Italy
- Nephrology and Transplantation Unit, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Kerry Rosenberg
- Department of Renal Medicine, University College London, London, UK
| | | | - Pedro Imenez Silva
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center, Rotterdam, The Netherlands
| | - Maria Adelina Simeoni
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, Caserta, Italy
| | - Gaye Hafez
- Department of Pharmacology, Faculty of Pharmacy, Altinbas University, Istanbul, Turkey
| | | | - Dorothea Nitsch
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
26
|
Xie K, Cao H, Ling S, Zhong J, Chen H, Chen P, Huang R. Global, regional, and national burden of chronic kidney disease, 1990-2021: a systematic analysis for the global burden of disease study 2021. Front Endocrinol (Lausanne) 2025; 16:1526482. [PMID: 40110544 PMCID: PMC11919670 DOI: 10.3389/fendo.2025.1526482] [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: 11/11/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Background Chronic kidney disease (CKD) continues to represent a significant public health concern, with both prevalence and incidence rates on the rise globally. Therefore, the study employed the Global Burden of Disease (GBD) database to investigate the global burden of CKD from 1990 to 2021. Methods This study utilized data from the GBD 2021. Join-point regression models were developed for the estimation of the average annual percentage change (AAPC) in the prevalence and mortality rates of CKD. Subsequently, stepwise multiple linear regression analysis was conducted to examine the trends in disability adjusted life years (DALYs) and DALYs rate for CKD across diverse populations between 1990 and 2021. Moreover, the influence of age, gender, and socio-demographic index (SDI) on the burden of CKD among patients from 1990 to 2021 was examined. Furthermore, the projection of the burden of CKD from 2022 to 2032 was also conducted. Results The AAPC for prevalence and mortality rates across the entire period spanning 1990 to 2021 was 0.92 and 2.66, respectively. A notable increase in the DALYs and DALYs rate for CKD was demonstrated over time, indicating a growing CKD burden on society since 1990. Furthermore, the DALYs rates for CKD were lowest in the 5-9 year age group for both genders, rising thereafter with age. Notably, the DALYs rate for CKD was higher in males than in females. Regions with higher SDI, generally exhibited a lower burden of CKD, while less developed regions, demonstrated the opposite pattern. Additionally, the age-standardized prevalence and mortality rates for CKD would be projected to increase to 8,773.85 and 21.26 per 100,000 individuals, respectively, by 2032. Conclusion The research indicated a gradual increase in the global prevalence and mortality rates of CKD over time, which might prompt the formulation of more efficient health policies to alleviate its burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Renfa Huang
- Nephropathy Department, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
27
|
Xiu F, Gai Z, Gehrig P, Wolski WE, Lone MA, Visentin M. The landscape of renal protein S-acylation in mice with lipid-induced nephrotoxicity. Sci Rep 2025; 15:7689. [PMID: 40044913 PMCID: PMC11882957 DOI: 10.1038/s41598-025-92530-7] [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: 10/22/2024] [Accepted: 02/28/2025] [Indexed: 03/09/2025] Open
Abstract
Excess fat intake is associated with kidney toxicity and dysfunction. Because fatty acids can also be reversibly attached onto cysteine residues and modulate the function of several membrane-bound proteins, we studied the effect of high-fat diet (HFD) on the S-acylated proteome of mouse kidneys to uncover novel biochemical changes that might contribute to lipid-induced nephrotoxicity. We compared the S-acylated proteome of kidneys from mice fed a chow diet (CD) or a HFD. HFD caused albuminuria. The HFD intervention induced a large-scale repression of protein S-acylation as well as of the most abundant ceramides and sphingomyelin species, which are highly suggestive of a reduction in acyl-CoA availability. The HFD-induced S-acylation repression mostly affected proteins involved in endocytosis and intracellular transport. Notably, the kidneys of mice fed a HFD displayed a marked decrease in the total amount and in the S-acylated form of megalin, the main tubular protein retrieval system. Further in vitro experiments indicated that S-acylation inhibition results in a reduction of megalin protein level. We conclude that diet-induced derangement of fatty acid metabolism modifies the renal landscape of the S-acylated proteome during the early stages of the kidney injury, which might reduce the efficiency of protein reabsorption by the proximal tubule.
Collapse
Affiliation(s)
- Fangrui Xiu
- Affiliated Hospital, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, 8006, Zurich, Switzerland
| | - Zhibo Gai
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, 8006, Zurich, Switzerland
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Peter Gehrig
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Witold E Wolski
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Museer A Lone
- Institute of Clinical Chemistry, University Hospital Zurich, Wagistrasse 14, 8952 Schlieren, 8006, Zurich, Switzerland.
| | - Michele Visentin
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, 8006, Zurich, Switzerland.
| |
Collapse
|
28
|
Collins KE, Gilbert E, Mauduit V, Benson KA, Elhassan EAE, O’Seaghdha C, Hill C, McKnight AJ, Maxwell AP, van der Most PJ, de Borst MH, Guan W, Jacobson PA, Israni AK, Keating BJ, Lord GM, Markkinen S, Helanterä I, Hyvärinen K, Partanen J, Madden SF, Lanktree MB, Limou S, Cavalleri GL, Conlon PJ. Donor and Recipient Polygenic Risk Scores Influence Kidney Transplant Function. Transpl Int 2025; 38:14171. [PMID: 40104404 PMCID: PMC11913612 DOI: 10.3389/ti.2025.14171] [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: 12/06/2024] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Kidney transplant outcomes are influenced by donor and recipient age, sex, HLA mismatch, donor type, anti-rejection medication adherence and disease recurrence, but variability in transplant outcomes remains unexplained. We hypothesise that donor and recipient polygenic burden for traits related to kidney function may also influence graft function. We assembled a cohort of 6,060 living and deceased kidney donor-recipient pairs. We calculated polygenic risk scores (PRSs) for kidney function-related traits in both donors and recipients. We investigated the association between these PRSs and recipient eGFR at 1- and 5-year post-transplant as well as graft failure. Donor: hypertension PRS (P < 0.001), eGFR PRS (P < 0.001), and intracranial aneurysm PRS (P = 0.01), along with recipient eGFR PRS (P = 0.001) were associated with eGFR at 1-year post-transplantation. Clinical factors explained 25% of the variation in eGFR at 1-year and 13% at 5-year, with PRSs cumulatively adding 1% in both cases. PRSs were not associated with long-term graft survival. We demonstrate a small, but statistically significant association between donor and recipient PRSs and recipient graft function at 1- and 5-year post-transplant. This effect is, at present, unlikely to have clinical application and further research is required to improve PRS performance.
Collapse
Affiliation(s)
- Kane E. Collins
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
- SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Edmund Gilbert
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
| | - Vincent Mauduit
- Ecole Centrale Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR1064, Nantes University, Nantes, France
| | - Katherine A. Benson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
| | - Elhussein A. E. Elhassan
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Conall O’Seaghdha
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Claire Hill
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
| | - Amy Jayne McKnight
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
| | | | - Peter J. van der Most
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martin H. de Borst
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Pamala A. Jacobson
- College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Ajay K. Israni
- Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Brendan J. Keating
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Graham M. Lord
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Salla Markkinen
- Finnish Red Cross Blood Service, Research and Development, Biomedicum 1, Helsinki, Finland
| | - Ilkka Helanterä
- Helsinki University Hospital, Transplantation and Liver Surgery, Helsinki, Finland
| | - Kati Hyvärinen
- Finnish Red Cross Blood Service, Research and Development, Biomedicum 1, Helsinki, Finland
| | - Jukka Partanen
- Finnish Red Cross Blood Service, Research and Development, Biomedicum 1, Helsinki, Finland
| | - Stephen F. Madden
- Data Science Centre, Beaux Lane House, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Matthew B. Lanktree
- Division of Nephrology, Departments of Medicine and Health Research Methodology, Evidence and Impact, St. Joseph’s Healthcare Hamilton, McMaster University and Population Health Research Institute, Hamilton, ON, Canada
| | - Sophie Limou
- Ecole Centrale Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR1064, Nantes University, Nantes, France
| | - Gianpiero L. Cavalleri
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
- SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Peter J. Conlon
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
29
|
Collins KE, Gilbert E, Mauduit V, Gaheer P, Elhassan EAE, Benson KA, Osman SM, Hill C, McKnight AJ, Maxwell AP, van der Most PJ, de Borst MH, Guan W, Jacobson PA, Israni AK, Keating BJ, Lord GM, Markkinen S, Helanterä I, Hyvärinen K, Partanen J, Madden SF, Storrar J, Sinha S, Kalra PA, Lanktree MB, Limou S, Cavalleri GL, Conlon PJ. Polygenic risk scores for eGFR are associated with age at kidney failure. J Nephrol 2025:10.1007/s40620-025-02207-7. [PMID: 40029548 DOI: 10.1007/s40620-025-02207-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/02/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND The genetic architecture of chronic kidney disease (CKD) is complex, including monogenic and polygenic contributions. CKD progression to kidney failure is influenced by factors including male sex, baseline estimated glomerular filtration rate (eGFR), hypertension, diabetes, proteinuria, and the underlying kidney disease. These traits all have strong genetic components, which can be partially quantified using polygenic risk scores. This paper examines the association between polygenic risk scores for CKD-related traits and age at kidney failure development. METHODS Genome-wide genotype data from 10,586 patients with kidney failure were compiled from 12 cohorts. Polygenic risk scores for hypertension, albuminuria, rapid decline in eGFR, decreased total kidney volume, and decreased eGFR were calculated using weights from published independent population-scale genome-wide association studies. The association between each polygenic risk score and age at kidney failure was investigated using logistic regression models. The association between polygenic risk score and age at kidney failure was also investigated separately for each primary kidney disease. RESULTS Individuals in the highest 10% of polygenic risk score for decreased eGFR developed kidney failure 2 years earlier than those in the bottom 90% (49.9 years and 47.9 years, P = 5e-5). A standard deviation increase in decreased eGFR polygenic risk score was associated with increased odds of developing kidney failure before the age of 60 years (Odds ratio (OR) = 1.05; 95% CI 1.01-1.10; P = 0.01), as was high decreased eGFR polygenic risk score (OR = 1.26; 95% CI 1.08-1.46; P = 0.003). CONCLUSIONS We conclude that decreased eGFR polygenic risk score explains a portion of the variation in age at development of kidney failure.
Collapse
Affiliation(s)
- Kane E Collins
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
- SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Edmund Gilbert
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
| | - Vincent Mauduit
- Nantes University, Ecole Centrale Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR1064, Nantes, France
| | - Pukhraj Gaheer
- Division of Nephrology, Departments of Medicine and Health Research Methodology, Evidence and Impact, St. Joseph's Healthcare Hamilton, McMaster University and Population Health Research Institute, Hamilton, ON, Canada
| | - Elhussein A E Elhassan
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Katherine A Benson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
| | - Shohdan Mohamad Osman
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Claire Hill
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | | | | | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamala A Jacobson
- Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Ajay K Israni
- University of Texas Medical Branch, Galveston, TX, USA
| | - Brendan J Keating
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Graham M Lord
- School of Immunology and Microbial Sciences, University College London, London, UK
| | - Salla Markkinen
- Finnish Red Cross Blood Service, Research and Development, Biomedicum 1, Helsinki, Finland
| | - Ilkka Helanterä
- Helsinki University Hospital, Transplantation and Liver Surgery, Helsinki, Finland
| | - Kati Hyvärinen
- Finnish Red Cross Blood Service, Research and Development, Biomedicum 1, Helsinki, Finland
| | - Jukka Partanen
- Finnish Red Cross Blood Service, Research and Development, Biomedicum 1, Helsinki, Finland
| | - Stephen F Madden
- Data Science Centre, Beaux Lane House, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Joshua Storrar
- Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
- University of Manchester, Manchester, UK
| | - Smeeta Sinha
- Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
- University of Manchester, Manchester, UK
| | - Philip A Kalra
- Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
- University of Manchester, Manchester, UK
| | - Matthew B Lanktree
- Division of Nephrology, Departments of Medicine and Health Research Methodology, Evidence and Impact, St. Joseph's Healthcare Hamilton, McMaster University and Population Health Research Institute, Hamilton, ON, Canada
| | - Sophie Limou
- Nantes University, Ecole Centrale Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR1064, Nantes, France
| | - Gianpiero L Cavalleri
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- The Science Foundation Ireland FutureNeuro Centre of Excellence, Dublin, Ireland
- SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Peter J Conlon
- Division of Nephrology, Departments of Medicine and Health Research Methodology, Evidence and Impact, St. Joseph's Healthcare Hamilton, McMaster University and Population Health Research Institute, Hamilton, ON, Canada.
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
30
|
Zanoni F, Obayemi JE, Gandla D, Castellano G, Keating BJ. Emerging role of genetics in kidney transplantation. Kidney Int 2025; 107:424-433. [PMID: 39710162 DOI: 10.1016/j.kint.2024.09.026] [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: 02/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 12/24/2024]
Abstract
The advent of more affordable genomic analytical pipelines has facilitated the expansion of genetic studies in kidney transplantation. Advances in genetic sequencing have allowed for a greater understanding of the genetic basis of chronic kidney disease, which has helped to guide transplant management and address issues related to living donation in specific disease settings. Recent efforts have shown significant effects of genetic ancestry and donor APOL1 risk genotypes in determining worse allograft outcomes and increased donation risks. Genetic studies in kidney transplantation outcomes have started to assess the effects of donor and recipient genetics in primary disease recurrence and transplant-related comorbidities, while genome-wide donor-recipient genetic incompatibilities have been shown to represent an important determinant of alloimmunity. Future large-scale comprehensive studies will shed light on the clinical utility of integrative genomics in the kidney transplantation setting.
Collapse
Affiliation(s)
- Francesca Zanoni
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Division of Transplantation, Department of Surgery, New York University Langone Health, Grossman School of Medicine, New York, New York, USA
| | - Joy E Obayemi
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA; Comprehensive Transplant Center, Department of Surgery, Northwestern University, Chicago Illinois, USA
| | - Divya Gandla
- Division of Transplantation, Department of Surgery, New York University Langone Health, Grossman School of Medicine, New York, New York, USA
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Brendan J Keating
- Institute of Systems Genetics, New York University Langone Health, Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
31
|
Thompson AS, Tresserra-Rimbau A, Jennings A, Bondonno NP, Candussi CJ, O'Neill JK, Hill C, Gaggl M, Cassidy A, Kühn T. Adherence to a Healthful Plant-Based Diet and Risk of Chronic Kidney Disease Among Individuals with Diabetes. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025; 44:212-222. [PMID: 39466646 DOI: 10.1080/27697061.2024.2415917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE Chronic kidney disease (CKD) is highly prevalent among people with diabetes. While identifying modifiable risk factors to prevent a decline in kidney function among those living with diabetes is pivotal, there is limited evidence on dietary risk factors for CKD. In this study, we examined the associations between healthy and less healthy plant-based diets (PBDs) and the risk of CKD among those with diabetes, and to identify potential underlying mechanisms. METHODS We conducted a prospective analysis among 7,747 UK Biobank participants with prevalent diabetes. Multivariable Cox proportional hazard regression models were used to examine the associations between healthful and unhealthful PBDs and the risk of CKD. Causal mediation analyses were further employed to explore the underlying mechanisms of the observed associations. RESULTS Among 7,747 study participants with diabetes, 1,030 developed incident CKD over 10.2 years of follow-up. Higher adherence to a healthy PBD was associated with a 24% lower CKD risk (HRQ4 versus Q1: 0.76 [95%CI: 0.63-0.92], ptrend = 0.002), while higher adherence to an unhealthy PBD was associated with a 35% higher risk (HRQ4 versus Q1: 1.35 [95%CI: 1.11-1.65], ptrend = 0.006). The observed associations were predominantly mediated by markers of body fatness (proportion mediated: 11-25%) and kidney function (23-89%). CONCLUSIONS In this prospective cohort study of middle-aged adults with diabetes, adherence to a healthy PBD was associated with lower CKD risk, whereas adherence to an unhealthy PBD was associated with a higher CKD risk. Associations were primarily mediated by markers of lower body fatness and improved kidney function.
Collapse
Affiliation(s)
- Alysha S Thompson
- The Co-Centre for Sustainable Food Systems and The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Anna Tresserra-Rimbau
- The Co-Centre for Sustainable Food Systems and The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
- Department of Nutrition, Food Science and Gastronomy, XIA, School of Pharmacy and Food Sciences, INSA, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Amy Jennings
- The Co-Centre for Sustainable Food Systems and The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Nicola P Bondonno
- The Co-Centre for Sustainable Food Systems and The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
- Danish Cancer Institute, Copenhagen, Denmark
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Catharina J Candussi
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Joshua K O'Neill
- The Co-Centre for Sustainable Food Systems and The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Claire Hill
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Martina Gaggl
- Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Aedín Cassidy
- The Co-Centre for Sustainable Food Systems and The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Tilman Kühn
- The Co-Centre for Sustainable Food Systems and The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
- Center for Public Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Wang Y, Cheng F, Hou N, Tan Y, Zhang S, Hou Y, Guo W, Peng J, Li W, Wu J. Increased risk of chronic diseases and multimorbidity in middle-aged and elderly individuals with early vision, hearing, or dual sensory impairments: insights from prospective cohort studies and Mendelian randomization analysis. BMC Med 2025; 23:118. [PMID: 40001102 PMCID: PMC11863693 DOI: 10.1186/s12916-025-03857-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/08/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Sensory impairments (SI), including vision (VI), hearing (HI), and dual sensory impairments (DSI), are prevalent with aging, but their impact on disease risk remains unclear. This study investigates the epidemiological and genetic associations between SIs and 10 chronic disease categories and multimorbidity. METHODS Using the CHARLS study, participants were classified by their self-reported VI/HI/DSI status in 2011 and 2013 into groups: "new onset, remission, persistent, and no SI." Their chronic disease incidence was tracked until 2018 in sub-cohorts respectively. Mendelian randomization (MR) analyses used genetic instruments from UK Biobank GWAS data on 88,250/504,307 individuals for vision/hearing loss, with outcome datasets from consortia including FinnGen, DIAMANTE, CKDGen, PGC, GWAS Catalog, and International Parkinson's Disease Genomics Consortium. RESULTS The cohort study revealed that persistent HI significantly increased the risk of heart disease (P < 0.001, HR 1.63, 95% CI 1.31-2.03), stroke (P 0.004, HR 1.59, 95% CI 1.16-2.18), chronic lung disease (P 0.002, HR 1.53, 95% CI 1.17-1.99), and emotional, nervous, or psychiatric problems (P 0.016, HR 2.03, 95% CI 1.14-3.60). Persistent VI was significantly associated with diabetes or high blood sugar (DM/Hglu) (P 0.012, HR 1.63, 95% CI 1.11-2.38) and chronic lung disease (P 0.042, HR 1.53, 95% CI 1.02-2.31). MR confirmed these strong or suggestive associations, indicating that HI significantly increased the risk of cardiovascular and cerebrovascular events by 61-170%, bronchitis by 160%, and schizophrenia by 36%. In addition, VI significantly raised the risk of hyperglycemia or diabetes by 2-4% and the risk of lung function decline. Additionally, cohort studies confirmed that early DSI significantly raised the risk of multiple diseases, while MR identified genetic links between VI and hepatic failure, Parkinson's, and Alzheimer's disease, and between HI and hypertension, chronic kidney disease, and renal failure. CONCLUSIONS This study provides evidence from epidemiological or genetic perspectives demonstrates that early exposure to HI/VI/DSI increases the risk of developing chronic diseases. These findings underscore the need for continuous monitoring and timely intervention for SI to manage chronic disease risks in aging populations.
Collapse
Affiliation(s)
- Yaoling Wang
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, Sichuan, 610041, China
| | - Fang Cheng
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 1277, Jiefang Avenue, Wuhan, 430000, China
| | - Niuniu Hou
- Department of General Surgery, Air Force 986(Th) Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Yuting Tan
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shaomin Zhang
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, Sichuan, 610041, China
| | - Yanbing Hou
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, Sichuan, 610041, China
| | - Wen Guo
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, Sichuan, 610041, China
| | - Jin Peng
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, Sichuan, 610041, China
| | - Wei Li
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No 1277, Jiefang Avenue, Wuhan, 430000, China.
| | - Jinhui Wu
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
33
|
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
|
34
|
Li H, Li C, Zhang C, Ying Z, Wu C, Zeng X, Bao J. Psychiatric disorders and following risk of chronic kidney disease: a prospective cohort study from UK Biobank. BMC Psychiatry 2025; 25:109. [PMID: 39934692 PMCID: PMC11816523 DOI: 10.1186/s12888-024-06461-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/27/2024] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Psychiatric disorders have been reported to influence many health outcomes, but evidence about their impact on chronic kidney disease (CKD) has not been fully explored, as well as possible mechanisms implicated are still unclear. METHODS Four hundred forty-one thousand eight hundred ninety-three participants from UK Biobank were included in this study. To assess the association between psychiatric disorders mainly including depression, anxiety, stress-related disorders, substance misuse as well as psychotic disorder, and CKD, a Cox regression model using age as the underlying time scale was employed. This approach considers the age progression of participants from the beginning to the end of the study as the elapsed time. Flexible nonparametric smoothing model was conducted to illustrate the temporal patterns. Subgroup analyses were performed by stratification of gender, genetic susceptibility to CKD, age at entry or exit the cohort, follow-up duration, and the number of psychiatric disorders at baseline. Mediation analysis was implemented to evaluate the roles of body mass index (BMI), hypertension, and diabetes. RESULTS Compared with individuals without psychiatric disorders, an increased risk of CKD was observed in patients with psychiatric disorders (hazard ratios (HR) = 1.52, 95% confidence intervals (CI): 1.40-1.65, p-value < 0.001). The hazard ratio among psychiatric patients gradually increased, and became significant after about 10 years follow-ups. The HR for patients followed up for 10-12 years was 1.60 (95% CI: 1.34-1.91, p-value < 0.001), and the HR was 1.66 (95% CI: 1.29-2.13, p-value < 0.001) for patients followed up for 12-13 years. Five distinct psychiatric disorders were found to be significantly associated with an increased risk of developing CKD. The highest HR was observed between stress-related disorder and CKD (HR = 1.95, 95%CI: 1.28-2.97, p-value = 0.002). When adjusting genetic susceptibility to CKD, the HR for the association between stress-related disorders and CKD became 1.86 (95%CI: 1.14-3.04, p-value = 0.013). Although these associations were nominally significant, they did not reach statistical significance after applying the Bonferroni multiple corrections, potentially due to the limited sample size. Subgroup analysis revealed that psychiatric patients who are under age 60, with multiple psychiatric morbidities or having been diagnosed with psychiatric disorders for over 10 years may be high-risk populations. Hypertension, BMI and diabetes mediated 49.13% (95% CI: 37.60%-67.08%), 12.11% (95% CI: 8.49%-17.24%) and 3.78% (95% CI: 1.58%-6.52%) of the total effect, respectively. CONCLUSIONS Psychiatric disorders were associated with a delayed onset of an elevated risk for CKD, this association was only observed in patients with psychiatric disorders for more than 10 years. Our study highlights the significance of lifestyle interventions, routine monitoring of kidney function, early screening for CKD, and personalized management strategies for psychiatric patients as potential approaches to the precise prevention of CKD.
Collapse
Affiliation(s)
- Hanfei Li
- Division of Nephrology and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- College of Life Science, SiChuan University, Chengdu, 610064, China
| | - Chunyang Li
- Division of Nephrology and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610065, China
| | - Chao Zhang
- Division of Nephrology and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610065, China
| | - Zhiye Ying
- Division of Nephrology and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610065, China
| | - Chuanfang Wu
- College of Life Science, SiChuan University, Chengdu, 610064, China
| | - Xiaoxi Zeng
- Division of Nephrology and West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Med-X Center for Informatics, Sichuan University, Chengdu, 610065, China.
| | - Jinku Bao
- College of Life Science, SiChuan University, Chengdu, 610064, China.
| |
Collapse
|
35
|
Wang S, Cheng Y, Zhang Z, Liu W, Ou M, Yin T, Meng Y, Ban H, Gu W, Meng X, Zhang L, Du Y. Association between obstructive sleep apnea and chronic kidney disease: A cross-sectional and Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41437. [PMID: 39928765 PMCID: PMC11812998 DOI: 10.1097/md.0000000000041437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/04/2025] [Accepted: 01/16/2025] [Indexed: 02/12/2025] Open
Abstract
Previous observational studies have shown that obstructive sleep apnea (OSA) was associated with chronic kidney disease(CKD). Early diagnosis of OSA usually helps better prevent the occurrence of CKD. This cross-sectional investigation was conducted using data from the National Health and Nutrition Examination Survey, which was carried out between 2007 to 2008 and 2015 to 2016. Logistic regression model was employed to assess the impact of OSA on CKD. We did a mediation analysis to assess how much of the effect of OSA on CKD was mediated through mediators. Additionally, Mendelian randomization (MR) analysis assessed the causal link between OSA and various measures of renal impairment and possible mediators: obesity, hypertension and type 2 diabetes mellitus. In the cross-sectional study, the results of unadjusted model showed that participants with OSA had a higher risk of CKD compared to non-OSA (OR = 1.14, 95% confidence intervals [CI]: 1.01-1.28, P < .05). In mediation analysis, the proportion of hypertension and obesity mediating the effect of OSA on CKD was 41.83% and 30.74%, respectively. Univariate MR analysis results showed that: genetically predicted OSA was associated with decreased estimated glomerular filtration ratecystatin c (eGFRcystatin c) level (OR = 0.997, 95% CI: 0.995-0.999, P < .05), increased blood urea nitrogen (BUN) levels (OR = 1.023, 95% CI: 1.008-1.038, P < .05), increased serum creatinine levels (OR = 1.010, 95% CI: 1.002-1.018, P < .05), increased serum cystatin C levels (OR = 1.015, 95% CI: 1.005-1.026, P < .05). Multivariable MR results showed that obesity mediated the causal effect of OSA on eGFRcystatin c, BUN levels and serum cystatin C levels. The cross-sectional study revealed a positive relationship between OSA and CKD, which was mediated by hypertension and obesity. The MR analysis suggest that OSA was associated with several measures of renal impairment, which was mediated by obesity. These findings may inform prevention and intervention strategies against CKD.
Collapse
Affiliation(s)
- Shaokang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yupei Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhe Zhang
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Mi Ou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Tianlong Yin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yalu Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Haipeng Ban
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wenlong Gu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xianggang Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lili Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuzheng Du
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
36
|
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
|
37
|
Stoneman HR, Price AM, Trout NS, Lamont R, Tifour S, Pozdeyev N, Crooks K, Lin M, Rafaels N, Gignoux CR, Marker KM, Hendricks AE. Characterizing substructure via mixture modeling in large-scale genetic summary statistics. Am J Hum Genet 2025; 112:235-253. [PMID: 39824191 PMCID: PMC11866976 DOI: 10.1016/j.ajhg.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/20/2025] Open
Abstract
Genetic summary data are broadly accessible and highly useful, including for risk prediction, causal inference, fine mapping, and incorporation of external controls. However, collapsing individual-level data into summary data, such as allele frequencies, masks intra- and inter-sample heterogeneity, leading to confounding, reduced power, and bias. Ultimately, unaccounted-for substructure limits summary data usability, especially for understudied or admixed populations. There is a need for methods to enable the harmonization of summary data where the underlying substructure is matched between datasets. Here, we present Summix2, a comprehensive set of methods and software based on a computationally efficient mixture model to enable the harmonization of genetic summary data by estimating and adjusting for substructure. In extensive simulations and application to public data, we show that Summix2 characterizes finer-scale population structure, identifies ascertainment bias, and scans for potential regions of selection due to local substructure deviation. Summix2 increases the robust use of diverse, publicly available summary data, resulting in improved and more equitable research.
Collapse
Affiliation(s)
- Hayley R Stoneman
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Adelle M Price
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA
| | - Nikole Scribner Trout
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA
| | - Riley Lamont
- Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA
| | - Souha Tifour
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA
| | - Nikita Pozdeyev
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristy Crooks
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Meng Lin
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicholas Rafaels
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher R Gignoux
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katie M Marker
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Audrey E Hendricks
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
38
|
Yang LZ, Yang Y, Hong C, Wu QZ, Shi XJ, Liu YL, Chen GZ. Systematic Mendelian Randomization Exploring Druggable Genes for Hemorrhagic Strokes. Mol Neurobiol 2025; 62:1359-1372. [PMID: 38977622 PMCID: PMC11772512 DOI: 10.1007/s12035-024-04336-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
Patients with hemorrhagic stroke have high rates of morbidity and mortality, and drugs for prevention are very limited. Mendelian randomization (MR) analysis can increase the success rate of drug development by providing genetic evidence. Previous MR analyses only analyzed the role of individual drug target genes in hemorrhagic stroke; therefore, we used MR analysis to systematically explore the druggable genes for hemorrhagic stroke. We sequentially performed summary-data-based MR analysis and two-sample MR analysis to assess the associations of all genes within the database with intracranial aneurysm, intracerebral hemorrhage, and their subtypes. Validated genes were further analyzed by colocalization. Only genes that were positive in all three analyses and were druggable were considered desirable genes. We also explored the mediators of genes affecting hemorrhagic stroke incidence. Finally, the associations of druggable genes with other cardiovascular diseases were analyzed to assess potential side effects. We identified 56 genes that significantly affected hemorrhagic stroke incidence. Moreover, TNFSF12, SLC22A4, SPARC, KL, RELT, and ADORA3 were found to be druggable. The inhibition of TNFSF12, SLC22A4, and SPARC can reduce the risk of intracranial aneurysm, subarachnoid hemorrhage, and intracerebral hemorrhage. Gene-induced hypertension may be a potential mechanism by which these genes cause hemorrhagic stroke. We also found that blocking these genes may cause side effects, such as ischemic stroke and its subtypes. Our study revealed that six druggable genes were associated with hemorrhagic stroke, and the inhibition of TNFSF12, SLC22A4, and SPARC had preventive effects against hemorrhagic strokes.
Collapse
Affiliation(s)
- Lun-Zhe Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chuan Hong
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qi-Zhe Wu
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiong-Jie Shi
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Lin Liu
- Department of Neurosurgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guang-Zhong Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
39
|
Li X, Zhao W, Pan H, Wang D. Separating the Effects of Early-Life and Adult Body Size on Chronic Kidney Disease Risk: A Mendelian Randomization Study. J Obes Metab Syndr 2025; 34:65-74. [PMID: 39800332 PMCID: PMC11799605 DOI: 10.7570/jomes24018] [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: 05/08/2024] [Revised: 05/27/2024] [Accepted: 09/19/2024] [Indexed: 01/25/2025] Open
Abstract
Background Whether there is a causal relationship between childhood obesity and increased risk of chronic kidney disease (CKD) remains controversial. This study sought to explore how body size in childhood and adulthood independently affects CKD risk in later life using a Mendelian randomization (MR) approach. Methods Univariate and multivariate MR was used to estimate total and independent effects of body size exposures. Genetic associations with early-life and adult body size were obtained from a genome-wide association study of 453,169 participants in the U.K. Biobank, and genetic associations with CKD were obtained from the CKDGen and FinnGen consortia. Results A larger genetically predicted early-life body size was associated with an increased risk of CKD (odds ratio [OR], 1.27; 95% confidence interval [CI], 1.14 to 1.41; P=1.70E-05) and increased blood urea nitrogen (BUN) levels (β=0.010; 95% CI, 0.005 to 0.021; P=0.001). However, the association between the impact of early-life body size on CKD (OR, 1.12; 95% CI, 0.95 to 1.31; P=0.173) and BUN level (β=0.001; 95% CI, -0.010 to 0.012; P=0.853) did not remain statistically significant after adjustment for adult body size. Larger genetically predicted adult body size was associated with an increased risk of CKD (OR, 1.37; 95% CI, 1.21 to 1.54; P=4.60E-07), decreased estimated glomerular filtration rate (β=-0.011; 95% CI, -0.017 to -0.006; P=5.79E-05), and increased BUN level (β=0.010; 95% CI, 0.002 to 0.019; P=0.018). Conclusion Our research indicates that the significant correlation between early-life body size and CKD risk is likely due to maintaining a large body size into adulthood.
Collapse
Affiliation(s)
- Xunliang Li
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenman Zhao
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haifeng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Deguang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
40
|
Gorski M, Wiegrebe S, Burkhardt R, Behr M, Küchenhoff H, Stark KJ, Böger CA, Heid IM. Bias-corrected serum creatinine from UK Biobank electronic medical records generates an important data resource for kidney function trajectories. Sci Rep 2025; 15:3540. [PMID: 39875408 PMCID: PMC11775100 DOI: 10.1038/s41598-025-85391-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Loss of kidney function is a substantial personal and public health burden. Kidney function is typically assessed as estimated glomerular filtration rate (eGFR) based on serum creatinine. UK Biobank provides serum creatinine measurements from study center assessments (SC, n = 425,147 baseline, n = 15,314 with follow-up) and emerging electronic Medical Records (eMR, "GP-clinical") present a promising resource to augment this data longitudinally. However, it is unclear whether eMR-based and SC-based creatinine values can be used jointly for research on eGFR decline. When comparing eMR-based with SC-based creatinine by calendar year (n = 70,231), we found a year-specific multiplicative bias for eMR-based creatinine that decreased over time (factor 0.84 for 2007, 0.97 for 2013). Deriving eGFR based on SC- and bias-corrected eMR-creatinine yielded 454,907 individuals with ≥ 1eGFR assessment (2,102,174 assessments). This included 206,063 individuals with ≥ 2 assessments over up to 60.2 years (median 6.00 assessments, median time = 8.7 years), where we also obtained eMR-based information on kidney disease or renal replacement therapy. We found an annual eGFR decline of 0.11 (95%-CI = 0.10-0.12) versus 1.04 mL/min/1.73m2/year (95%-CI = 1.03-1.05) without and with bias-correction, the latter being in line with literature. In summary, our bias-corrected eMR-based creatinine values enabled a 4-fold increased number of eGFR assessments in UK Biobank suitable for kidney function research.
Collapse
Affiliation(s)
- Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Simon Wiegrebe
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Merle Behr
- Faculty of Informatics and Data Science, University of Regensburg, Regensburg, Germany
| | - Helmut Küchenhoff
- Statistical Consulting Unit StaBLab, Department of Statistics, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Center for Machine Learning (MCML), Munich, Germany
| | - Klaus J Stark
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Nephrology, Diabetology and Rheumatology, Kliniken Südostbayern, Traunstein, Germany
- KfH Kidney Center Traunstein, Traunstein, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
41
|
Qi X, Wang J, Wang T, Wang W, Zhang D. Epigenome-wide association study of Chinese monozygotic twins identifies DNA methylation loci associated with estimated glomerular filtration rate. J Transl Med 2025; 23:101. [PMID: 39844292 PMCID: PMC11752939 DOI: 10.1186/s12967-025-06067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/05/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND DNA methylation (DNAm) has been shown in multiple studies to be associated with the estimated glomerular filtration rate (eGFR). However, studies focusing on Chinese populations are lacking. We conducted an epigenome-wide association study to investigate the association between DNAm and eGFR in Chinese monozygotic twins. METHODS Genome-wide DNAm level was detected using Reduced Representation Bisulfite Sequencing test. Generalized estimation equation (GEE) was used to examine the association between Cytosine-phosphate-Guanines (CpGs) DNAm and eGFR. Inference about Causation from Examination of FAmiliaL CONfounding was employed to infer the causal relationship. The comb-p was used to identify differentially methylated regions (DMRs). GeneMANIA was used to analyze the gene interaction network. The Genomic Regions Enrichment of Annotations Tool enriched biological functions and pathways. Gene expression profiling sequencing was employed to measure mRNA expression levels, and the GEE model was used to investigate the association between gene expression and eGFR. The candidate gene was validated in a community population by calculating the methylation risk score (MRS). RESULTS A total of 80 CpGs and 28 DMRs, located at genes such as OLIG2, SYNGR3, LONP1, CDCP1, and SHANK1, achieved genome-wide significance level (FDR < 0.05). The causal effect of DNAm on eGFR was supported by 12 CpGs located at genes such as SYNGR3 and C9orf3. In contrast, the causal effect of eGFR on DNAm is proved by 13 CpGs located at genes such as EPHB3 and MLLT1. Enrichment analysis revealed several important biological functions and pathways related to eGFR, including alpha-2A adrenergic receptor binding pathway and corticotropin-releasing hormone receptor activity pathway. GeneMANIA results showed that SYNGR3 was co-expressed with MLLT1 and had genetic interactions with AFF4 and EDIL3. Gene expression analysis found that SYNGR3 expression was negatively associated with eGFR. Validation analysis showed that the MRS of SYNGR3 was positively associated with low eGFR levels. CONCLUSIONS We identified a set of CpGs, DMRs, and pathways potentially associated with eGFR, particularly in the SYNGR3 gene. These findings provided new insights into the epigenetic modifications related to the decline in eGFR and chronic kidney disease.
Collapse
Affiliation(s)
- Xueting Qi
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Jingjing Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Tong Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, People's Republic of China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, People's Republic of China.
| |
Collapse
|
42
|
Zhang Y, Ou G, Peng L, Pan J, Zhang S, Shi J. Genetic association analysis of lipid-lowering drug target genes in chronic kidney disease. Front Endocrinol (Lausanne) 2025; 15:1434145. [PMID: 39877840 PMCID: PMC11772207 DOI: 10.3389/fendo.2024.1434145] [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: 06/13/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Objective The impact of lipid-lowering medications on chronic kidney disease (CKD) remains a subject of debate. This Mendelian randomization (MR) study aims to elucidate the potential effects of lipid-lowering drug targets on CKD development. Methods We extracted 11 genetic variants encoding targets of lipid-lowering drugs from published genome-wide association study (GWAS) summary statistics, encompassing LDLR, HMGCR, PCSK9, NPC1L1, APOB, ABCG5/ABCG8, LPL, APOC3, ANGPTL3, and PPARA. A Mendelian randomization analysis was conducted targeting these drug-related genes. CKD risk was designated as the primary outcome, while estimated glomerular filtration rate (eGFR) and blood urea nitrogen (BUN) were assessed as secondary outcomes. Additionally, mediation analysis was performed utilizing 731 immune cell phenotypes to identify potential mediators. Results The meta-analysis revealed a significant association between ANGPTL3 inhibitors and a reduced risk of CKD (OR [95% CI] = 0.85 [0.75-0.96]). Conversely, LDLR agonists were significantly linked to an increased risk of CKD (OR [95% CI] = 1.11 [1.02-1.22]). Regarding secondary outcomes, lipid-lowering drugs did not significantly affect eGFR and BUN levels. Mediation analysis indicated that the reduction in CKD risk by ANGPTL3 inhibitors was mediated through modulation of the immune cell phenotype, specifically HLA-DR on CD14+ CD16+ monocytes (Mediated proportion: 4.69%; Mediated effect: -0.00899). Conclusion Through drug-targeted MR analysis, we identified a causal relationship between lipid-lowering drug targets and CKD. ANGPTL3 and LDLR may represent promising candidate drug targets for CKD treatment.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Medical University, Jinzhou, Liaoning, China
- Department of Urology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Guangyang Ou
- Department of Cardiology, Hunan University of Chinese Medicine, Changsha, China
| | - Lei Peng
- Motor Robotics Institute (MRI), South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jian Pan
- Motor Robotics Institute (MRI), South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Shaohua Zhang
- Motor Robotics Institute (MRI), South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jianguo Shi
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
43
|
Valo E, Richmond A, Mutter S, Dahlström EH, Campbell A, Porteous DJ, Wilson JF, Groop PH, Hayward C, Sandholm N. Genome-wide characterization of 54 urinary metabolites reveals molecular impact of kidney function. Nat Commun 2025; 16:325. [PMID: 39746953 PMCID: PMC11696681 DOI: 10.1038/s41467-024-55182-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Dissecting the genetic mechanisms underlying urinary metabolite concentrations can provide molecular insights into kidney function and open possibilities for causal assessment of urinary metabolites with risk factors and disease outcomes. Proton nuclear magnetic resonance metabolomics provides a high-throughput means for urinary metabolite profiling, as widely applied for blood biomarker studies. Here we report a genome-wide association study meta-analysed for 3 European cohorts comprising 8,011 individuals, covering both people with type 1 diabetes and general population settings. We identify 54 associations (p < 9.3 × 10-10) for 19 of 54 studied metabolite concentrations. Out of these, 33 were not reported previously for relevant urinary or blood metabolite traits. Subsequent two-sample Mendelian randomization analysis suggests that estimated glomerular filtration rate causally affects 13 urinary metabolite concentrations whereas urinary ethanolamine, an initial precursor for phosphatidylcholine and phosphatidylethanolamine, was associated with higher eGFR lending support for a potential protective role. Our study provides a catalogue of genetic associations for 53 metabolites, enabling further investigation on how urinary metabolites are linked to human health.
Collapse
Grants
- Wellcome Trust
- MC_UU_00007/10 Medical Research Council
- Folkhälsan Research Foundation, Wilhelm and Else Stockmann Foundation, Liv och Hälsa Society, Helsinki University Hospital Research Funds (EVO TYH2018207), Academy of Finland (299200, and 316664), Novo Nordisk Foundation (NNF OC0013659, NNF23OC0082732), Sigrid Jusélius Foundation, and Finnish Diabetes Research Foundation. Genotyping of the FinnDiane GWAS data was funded by the Juvenile Diabetes Research Foundation (JDRF) within the Diabetic Nephropathy Collaborative Research Initiative (DNCRI; Grant 17-2013-7), with GWAS quality control and imputation performed at University of Virginia. Generation Scotland received core support from the Chief Scientist Office of the Scottish Government Health Directorates [CZD/16/6] and the Scottish Funding Council [HR03006] and is currently supported by the Wellcome Trust [216767/Z/19/Z]. Genotyping of the GS:SFHS samples was carried out by the Genetics Core Laboratory at the Edinburgh Clinical Research Facility, University of Edinburgh, Scotland and was funded by the Medical Research Council UK and the Wellcome Trust (Wellcome Trust Strategic Award “STratifying Resilience and Depression Longitudinally” (STRADL) Reference 104036/Z/14/Z). CH was supported by the MRC Human Genetics Unit quinquennial programme grant “QTL in Health and Disease” (MC_UU_00007/10.) The Viking Health Study – Shetland (VIKING) was supported by the MRC Human Genetics Unit quinquennial programme grant “QTL in Health and Disease” (MC_UU_00007/10).
Collapse
Affiliation(s)
- Erkka Valo
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anne Richmond
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Stefan Mutter
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emma H Dahlström
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - James F Wilson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Per-Henrik Groop
- Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Caroline Hayward
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK.
| | - Niina Sandholm
- Folkhälsan Research Center, Helsinki, Finland.
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
44
|
Shivakumar M, Kim Y, Jung SH, Woerner J, Kim D. Frequency of adding salt is a stronger predictor of chronic kidney disease in individuals with genetic risk. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2025; 30:551-564. [PMID: 39670395 PMCID: PMC12008778 DOI: 10.1142/9789819807024_0039] [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] [Indexed: 05/16/2025]
Abstract
The incidence of chronic kidney disease (CKD) is increasing worldwide, but there is no specific treatment available. Therefore, understanding and controlling the risk factors for CKD are essential for preventing disease occurrence. Salt intake raises blood pressure by increasing fluid volume and contributes to the deterioration of kidney function by enhancing the renin-angiotensin system and sympathetic tone. Thus, a low-salt diet is important to reduce blood pressure and prevent kidney diseases. With recent advancements in genetic research, our understanding of the etiology and genetic background of CKD has deepened, enabling the identification of populations with a high genetic predisposition to CKD. It is thought that the impact of lifestyle or environmental factors on disease occurrence or prevention may vary based on genetic factors. This study aims to investigate whether frequency of adding salt has different effects depending on genetic risk for CKD. CKD polygenic risk scores (PRS) were generated using CKDGen Consortium GWAS (N= 765,348) summary statics. Then we applied the CKD PRS to UK Biobank subjects. A total of 331,318 European individuals aged 40-69 without CKD were enrolled in the study between 2006-2010. The average age at enrollment of the participants in this study was 56.69, and 46% were male. Over an average follow-up period of 8 years, 12,279 CKD cases were identified. The group that developed CKD had a higher percentage of individuals who added salt (46.37% vs. 43.04%) and higher CKD high-risk PRS values compared to the group that did not develop CKD (23.53% vs. 19.86%). We classified the individuals into four groups based on PRS: low (0-19%), intermediate (20-79%), high (80-94%), very high (≥ 95%). Incidence of CKD increased incrementally according to CKD PRS even after adjusting for age, sex, race, Townsend deprivation index, body mass index, estimated glomerular filtration rate, smoking, alcohol, physical activity, diabetes mellitus, dyslipidemia, hypertension, coronary artery diseases, cerebrovascular diseases at baseline. Compared to the "never/rarely" frequency of adding salt group, "always" frequency of adding salt group had an increasing incidence of CKD proportionate to the degree of frequency of adding salt. However, the significant association of "always" group on incident CKD disappeared in the low PRS group. This study validated the signal from PRSs for CKD across a large cohort and confirmed that frequency of adding salt contributes to the occurrence of CKD. Additionally, it confirmed that the effect of frequency of "always" adding salt on CKD incidence is greater in those with more than intermediate CKD-PRS. This study suggests that increased salt intake is particularly concerning for individuals with genetic risk factors for CKD, underscoring the clinical importance of reducing salt intake for these individuals.
Collapse
Affiliation(s)
- Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanggyun Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakob Woerner
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA,
| |
Collapse
|
45
|
Guo X, Feng Y, Ji X, Jia N, Maimaiti A, Lai J, Wang Z, Yang S, Hu S. Shared genetic architecture and bidirectional clinical risks within the psycho-metabolic nexus. EBioMedicine 2025; 111:105530. [PMID: 39731856 PMCID: PMC11743124 DOI: 10.1016/j.ebiom.2024.105530] [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: 09/05/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Increasing evidence suggests a complex interplay between psychiatric disorders and metabolic dysregulations. However, most research has been limited to specific disorder pairs, leaving a significant gap in our understanding of the broader psycho-metabolic nexus. METHODS This study leveraged large-scale cohort data and genome-wide association study (GWAS) summary statistics, covering 8 common psychiatric disorders and 43 metabolic traits. We introduced a comprehensive analytical strategy to identify shared genetic bases sequentially, from key genetic correlation regions to local pleiotropy and pleiotropic genes. Finally, we developed polygenic risk score (PRS) models to translate these findings into clinical applications. FINDINGS We identified significant bidirectional clinical risks between psychiatric disorders and metabolic dysregulations among 310,848 participants from the UK Biobank. Genetic correlation analysis confirmed 104 robust trait pairs, revealing 1088 key genomic regions, including critical hotspots such as chr3: 47588462-50387742. Cross-trait meta-analysis uncovered 388 pleiotropic single nucleotide variants (SNVs) and 126 shared causal variants. Among variants, 45 novel SNVs were associated with psychiatric disorders and 75 novel SNVs were associated with metabolic traits, shedding light on new targets to unravel the mechanism of comorbidity. Notably, RBM6, a gene involved in alternative splicing and cellular stress response regulation, emerged as a key pleiotropic gene. When psychiatric and metabolic genetic information were integrated, PRS models demonstrated enhanced predictive power. INTERPRETATION The study highlights the intertwined genetic and clinical relationships between psychiatric disorders and metabolic dysregulations, emphasising the need for integrated approaches in diagnosis and treatment. FUNDING The National Key Research and Development Program of China (2023YFC2506200, SHH). The National Natural Science Foundation of China (82273741, SY).
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Feng
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne, Carlton South, VIC, Australia
| | - Xiaolong Ji
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ningning Jia
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Aierpati Maimaiti
- Department of Neurosurgery, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Jianbo Lai
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zheng Wang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Sheng Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Nanhu Brain-Computer Interface Institute, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Precision Psychiatry, Hangzhou, 310003, China; Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China; Brain Research Institute of Zhejiang University, Hangzhou, 310058, China; MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058, China; Department of Psychology and Behavioral Sciences, Graduate School, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
46
|
Khan A, Kiryluk K. Polygenic scores and their applications in kidney disease. Nat Rev Nephrol 2025; 21:24-38. [PMID: 39271761 DOI: 10.1038/s41581-024-00886-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/15/2024]
Abstract
Genome-wide association studies (GWAS) have uncovered thousands of risk variants that individually have small effects on the risk of human diseases, including chronic kidney disease, type 2 diabetes, heart diseases and inflammatory disorders, but cumulatively explain a substantial fraction of disease risk, underscoring the complexity and pervasive polygenicity of common disorders. This complexity poses unique challenges to the clinical translation of GWAS findings. Polygenic scores combine small effects of individual GWAS risk variants across the genome to improve personalized risk prediction. Several polygenic scores have now been developed that exhibit sufficiently large effects to be considered clinically actionable. However, their clinical use is limited by their partial transferability across ancestries and a lack of validated models that combine polygenic, monogenic, family history and clinical risk factors. Moreover, prospective studies are still needed to demonstrate the clinical utility and cost-effectiveness of polygenic scores in clinical practice. Here, we discuss evolving methods for developing polygenic scores, best practices for validating and reporting their performance, and the study designs that will empower their clinical implementation. We specifically focus on the polygenic scores relevant to nephrology and other chronic, complex diseases and review their key limitations, necessary refinements and potential clinical applications.
Collapse
Affiliation(s)
- Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
47
|
Weng LC, Rämö JT, Jurgens SJ, Khurshid S, Chaffin M, Hall AW, Morrill VN, Wang X, Nauffal V, Sun YV, Beer D, Lee S, Nadkarni GN, Duong T, Wang B, Czuba T, Austin TR, Yoneda ZT, Friedman DJ, Clayton A, Hyman MC, Judy RL, Skanes AC, Orland KM, Treu TM, Oetjens MT, Alonso A, Soliman EZ, Lin H, Lunetta KL, van der Pals J, Issa TZ, Nafissi NA, May HT, Leong-Sit P, Roselli C, Choi SH, Khan HR, Knight S, Karlsson Linnér R, Bezzina CR, Ripatti S, Heckbert SR, Gaziano JM, Loos RJF, Psaty BM, Smith JG, Benjamin EJ, Arking DE, Rader DJ, Shah SH, Roden DM, Damrauer SM, Eckhardt LL, Roberts JD, Cutler MJ, Shoemaker MB, Haggerty CM, Cho K, Palotie A, Wilson PWF, Ellinor PT, Lubitz SA. The impact of common and rare genetic variants on bradyarrhythmia development. Nat Genet 2025; 57:53-64. [PMID: 39747593 PMCID: PMC11735381 DOI: 10.1038/s41588-024-01978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/09/2024] [Indexed: 01/04/2025]
Abstract
To broaden our understanding of bradyarrhythmias and conduction disease, we performed common variant genome-wide association analyses in up to 1.3 million individuals and rare variant burden testing in 460,000 individuals for sinus node dysfunction (SND), distal conduction disease (DCD) and pacemaker (PM) implantation. We identified 13, 31 and 21 common variant loci for SND, DCD and PM, respectively. Four well-known loci (SCN5A/SCN10A, CCDC141, TBX20 and CAMK2D) were shared for SND and DCD, while others were more specific for SND or DCD. SND and DCD showed a moderate genetic correlation (rg = 0.63). Cardiomyocyte-expressed genes were enriched for contributions to DCD heritability. Rare-variant analyses implicated LMNA for all bradyarrhythmia phenotypes, SMAD6 and SCN5A for DCD and TTN, MYBPC3 and SCN5A for PM. These results show that variation in multiple genetic pathways (for example, ion channel function, cardiac developmental programs, sarcomeric structure and cellular homeostasis) appear critical to the development of bradyarrhythmias.
Collapse
Grants
- R01 HL141901 NHLBI NIH HHS
- R01 HL139738 NHLBI NIH HHS
- 18SFRN34250007 American Heart Association (American Heart Association, Inc.)
- TNE FANTASY 19CV03 Fondation Leducq
- R01 HL092577 NHLBI NIH HHS
- R01 HL105756 NHLBI NIH HHS
- R01 HL157635 NHLBI NIH HHS
- R01 HL139731 NHLBI NIH HHS
- U01 AG068221 NIA NIH HHS
- 23CDA1050571 American Heart Association (American Heart Association, Inc.)
- T32 HL007101 NHLBI NIH HHS
- R01 AG083735 NIA NIH HHS
- 18SFRN34110082 American Heart Association (American Heart Association, Inc.)
- 18SFRN34230127 American Heart Association (American Heart Association, Inc.)
- IK2 CX001780 CSRD VA
- 75N92019D00031 NHLBI NIH HHS
- K23 HL169839 NHLBI NIH HHS
- 03-007-2022-0035 Hartstichting (Dutch Heart Foundation)
- R21 HL175584 NHLBI NIH HHS
- R01 HL163987 NHLBI NIH HHS
- National Institutes of Health:R01HL139731 & R01HL157635
- Sigrid Juséliuksen Säätiö (Sigrid Jusélius Foundation)
- National Institutes of Health: K23HL169839
- National Institutes of Health: RO1HL092577
- National Institutes of Health: T32HL007101
- Swedish Heart-Lung Foundation (2022-0344, 2022-0345), the Swedish Research Council (2021-02273), the European Research Council (ERC-STG-2015-679242), Gothenburg University, Skane University Hospital, the Scania county, governmental funding of clinical research within the Swedish National Health Service, a generous donation from the Knut and Alice Wallenberg foundation to the Wallenberg Center for Molecular Medicine in Lund, and funding from the Swedish Research Council (Linnaeus grant Dnr 349-2006-237, Strategic Research Area Exodiab Dnr 2009-1039) and Swedish Foundation for Strategic Research (Dnr IRC15-0067) to the Lund University Diabetes Center.
- US Department of Veterans Affairs Clinical Research and Development award IK2-CX001780
- National Institutes of Health: R01HL163987-01 and R01HL139738-01
- Academy of Finland Centre of Excellence in Complex Disease Genetics (grant no. 312074 and 336824)
- National Institutes of Health: R01HL139731, R01HL157635, and RO1HL092577 European Union: MAESTRIA 965286
Collapse
Affiliation(s)
- Lu-Chen Weng
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Joel T Rämö
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean J Jurgens
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Shaan Khurshid
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Mark Chaffin
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amelia Weber Hall
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Valerie N Morrill
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xin Wang
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Victor Nauffal
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Yan V Sun
- VA Atlanta Healthcare System, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | | | - Simon Lee
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | | | - ThuyVy Duong
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Biqi Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Tomasz Czuba
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Thomas R Austin
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Zachary T Yoneda
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel J Friedman
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Anne Clayton
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
| | - Matthew C Hyman
- Division of Cardiac Electrophysiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Allan C Skanes
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, Ontario, Canada
| | - Kate M Orland
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Matthew T Oetjens
- Autism and Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center, Section on Cardiovascular Medicine, Department of Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jesper van der Pals
- Department of Cardiology, Clinical Sciences, Lund University and Skane University Hospital, Lund, Sweden
| | - Tariq Z Issa
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Navid A Nafissi
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Heidi T May
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
| | - Peter Leong-Sit
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, Ontario, Canada
| | - Carolina Roselli
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Habib R Khan
- Section of Cardiac Electrophysiology, Western University, London, Ontario, Canada
| | - Stacey Knight
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
- Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Richard Karlsson Linnér
- Autism and Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
- Department of Economics, Leiden Law School, Leiden University, Leiden, The Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Center, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - J Michael Gaziano
- VA Boston Healthcare System, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, USA
| | - J Gustav Smith
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Cardiology, Clinical Sciences, Lund University and Skane University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Emelia J Benjamin
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- NHLBI and BU's Framingham Heart Study, Framingham, MA, USA
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel J Rader
- Departments of Medicine and Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Svati H Shah
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dan M Roden
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics and Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lee L Eckhardt
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jason D Roberts
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Western University, London, Ontario, Canada
| | - Michael J Cutler
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
| | - M Benjamin Shoemaker
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher M Haggerty
- Heart Institute, Geisinger, Danville, PA, USA
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA, USA
| | - Kelly Cho
- VA Boston Healthcare System, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Peter W F Wilson
- VA Atlanta Healthcare System, Decatur, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Steven A Lubitz
- Telemachus and Irene Demoulas Family Foundation Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
48
|
Bazua-Valenti S, Brown MR, Zavras J, Riedl Khursigara M, Grinkevich E, Sidhom EH, Keller KH, Racette M, Dvela-Levitt M, Quintanova C, Demirci H, Sewerin S, Goss AC, Lin J, Yoo H, Vaca Jacome AS, Papanastasiou M, Udeshi N, Carr SA, Himmerkus N, Bleich M, Mutig K, Bachmann S, Halbritter J, Kmoch S, Živná M, Kidd K, Bleyer AJ, Weins A, Alper SL, Shaw JL, Kost-Alimova M, Pablo JLB, Greka A. Disrupted uromodulin trafficking is rescued by targeting TMED cargo receptors. J Clin Invest 2024; 134:e180347. [PMID: 39680459 PMCID: PMC11645142 DOI: 10.1172/jci180347] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 10/22/2024] [Indexed: 12/18/2024] Open
Abstract
The trafficking dynamics of uromodulin (UMOD), the most abundant protein in human urine, play a critical role in the pathogenesis of kidney disease. Monoallelic mutations in the UMOD gene cause autosomal dominant tubulointerstitial kidney disease (ADTKD-UMOD), an incurable genetic disorder that leads to kidney failure. The disease is caused by the intracellular entrapment of mutant UMOD in kidney epithelial cells, but the precise mechanisms mediating disrupted UMOD trafficking remain elusive. Here, we report that transmembrane Emp24 protein transport domain-containing (TMED) cargo receptors TMED2, TMED9, and TMED10 bind UMOD and regulate its trafficking along the secretory pathway. Pharmacological targeting of TMEDs in cells, in human kidney organoids derived from patients with ADTKD-UMOD, and in mutant-UMOD-knockin mice reduced intracellular accumulation of mutant UMOD and restored trafficking and localization of UMOD to the apical plasma membrane. In vivo, the TMED-targeted small molecule also mitigated ER stress and markers of kidney damage and fibrosis. Our work reveals TMED-targeting small molecules as a promising therapeutic strategy for kidney proteinopathies.
Collapse
Affiliation(s)
- Silvana Bazua-Valenti
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Departamento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, México
| | - Matthew R. Brown
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Jason Zavras
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Magdalena Riedl Khursigara
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Elizabeth Grinkevich
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Eriene-Heidi Sidhom
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keith H. Keller
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew Racette
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Moran Dvela-Levitt
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Hasan Demirci
- Institute of Translational Physiology and
- Department of Anatomy, Charité - Universitätsmedizin, Berlin, Germany
| | - Sebastian Sewerin
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Alissa C. Goss
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - John Lin
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Hyery Yoo
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Alvaro S. Vaca Jacome
- Proteomics Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Malvina Papanastasiou
- Proteomics Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Namrata Udeshi
- Proteomics Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Steven A. Carr
- Proteomics Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nina Himmerkus
- Institute of Physiology, Christian - Albrechts - Universität, Kiel, Germany
| | - Markus Bleich
- Institute of Physiology, Christian - Albrechts - Universität, Kiel, Germany
| | - Kerim Mutig
- Institute of Translational Physiology and
- Department of Anatomy, Charité - Universitätsmedizin, Berlin, Germany
| | - Sebastian Bachmann
- Institute of Translational Physiology and
- Department of Anatomy, Charité - Universitätsmedizin, Berlin, Germany
| | - Jan Halbritter
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin, Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martina Živná
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kendrah Kidd
- Section on Nephrology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, North Carolina, USA
| | - Anthony J. Bleyer
- Section on Nephrology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, North Carolina, USA
| | - Astrid Weins
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Seth L. Alper
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Division of Nephrology, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jillian L. Shaw
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Maria Kost-Alimova
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Juan Lorenzo B. Pablo
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
| | - Anna Greka
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Nanamatsu A, de Araújo L, LaFavers KA, El-Achkar TM. Advances in uromodulin biology and potential clinical applications. Nat Rev Nephrol 2024; 20:806-821. [PMID: 39160319 PMCID: PMC11568936 DOI: 10.1038/s41581-024-00881-7] [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] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Uromodulin (also known as Tamm-Horsfall protein) is a kidney-specific glycoprotein secreted bidirectionally into urine and into the circulation, and it is the most abundant protein in normal urine. Although the discovery of uromodulin predates modern medicine, its significance in health and disease has been rather enigmatic. Research studies have gradually revealed that uromodulin exists in multiple forms and has important roles in urinary and systemic homeostasis. Most uromodulin in urine is polymerized into highly organized filaments, whereas non-polymeric uromodulin is detected both in urine and in the circulation, and can have distinct roles. The interactions of uromodulin with the immune system, which were initially reported to be a key role of this protein, are now better understood. Moreover, the discovery that uromodulin is associated with a spectrum of kidney diseases, including acute kidney injury, chronic kidney disease and autosomal-dominant tubulointerstitial kidney disease, has further accelerated investigations into the role of this protein. These discoveries have prompted new questions and ushered in a new era in uromodulin research. Here, we delineate the latest discoveries in uromodulin biology and its emerging roles in modulating kidney and systemic diseases, and consider future directions, including its potential clinical applications.
Collapse
Affiliation(s)
- Azuma Nanamatsu
- Department of Medicine, Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Larissa de Araújo
- Department of Medicine, Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kaice A LaFavers
- Department of Medicine, Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tarek M El-Achkar
- Department of Medicine, Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Roudebush VA Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
50
|
Gao Y, Li Q, Yang L, Zhao H, Wang D, Pesola AJ. Causal Association Between Sedentary Behaviors and Health Outcomes: A Systematic Review and Meta-Analysis of Mendelian Randomization Studies. Sports Med 2024; 54:3051-3067. [PMID: 39218828 DOI: 10.1007/s40279-024-02090-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Different types of sedentary behavior are associated with several health outcomes, but the causality of these associations remains unclear. OBJECTIVES To conduct a systematic review and meta-analysis of Mendelian randomization (MR) studies investigating the associations between sedentary behaviors and health outcomes. METHODS A systematic search on PubMed, Embase, Web of Science, Scopus, and PsycINFO up to August 2023 was conducted to identify eligible MR studies. We selected studies that assessed associations of genetically determined sedentary behaviors and health outcomes. A meta-analysis was conducted to examine the causal associations when two or more MR studies were available. We graded the evidence level of each MR association based on the results of the main method and sensitivity analyses in MR studies. RESULTS A total of 31 studies with 168 MR associations between six types of sedentary behavior and 47 health outcomes were included. Results from meta-analyses suggested a total of 47 significant causal associations between sedentary behaviors and health outcomes. Notably, more leisure TV watching is robustly correlated with increased risks of myocardial infarction, coronary artery disease, all-cause ischemic stroke, and type 2 diabetes. Conversely, robust inverse associations were observed between leisure computer use and risks of rheumatoid arthritis, Alzheimer's disease, and gastroesophageal reflux disease. CONCLUSION These findings suggest that different types of sedentary behavior have distinct causal effects on health outcomes. Therefore, interventions should focus not only on reducing sedentary time but also on promoting healthier types of sedentary behavior. PROSPERO REGISTRATION CRD42023453828.
Collapse
Affiliation(s)
- Ying Gao
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, China
| | - Qingyang Li
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, China
| | - Luyao Yang
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, China
| | - Hanhua Zhao
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, China
| | - Di Wang
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou, China.
| | - Arto J Pesola
- Active Life Lab, South-Eastern Finland University of Applied Sciences, Mikkeli, Finland
| |
Collapse
|