1
|
Lian Z, Liang Z, Chen Q, Xie C, Kong Y. Association between lipid-lowering drug targets and the risk of cystic kidney disease: a drug-target Mendelian randomization analysis. Ren Fail 2025; 47:2491657. [PMID: 40289090 PMCID: PMC12035922 DOI: 10.1080/0886022x.2025.2491657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Evidence regarding the causal relationship between lipid-lowering drugs and cystic kidney disease, including polycystic kidney disease (PKD), was limited. This study aimed to evaluate the causal relationship between lipid phenotypes mediated by lipid-lowering drug targets-3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGCR), proprotein convertase subtilisin/kexin type-9 (PCSK9), and Niemann-Pick C1-like 1 (NPC1L1)-and the risk of cystic kidney disease and PKD. METHODS Genetic variants encoding lipid-lowering drug targets-HMGCR, PCSK9, and NPC1L1-from published genome-wide association study (GWAS) statistics were collected to perform drug target Mendelian randomization (MR) analysis. Summary statistics for the GWAS of cystic kidney disease and PKD were obtained from the FinnGen consortium and the European Bioinformatics Institute. Inverse variance weighting (IVW) was used as the primary MR analysis method, with sensitivity analyses conducted to ensure the robustness of the results. RESULTS Increased gene expression of HMGCR was associated with an elevated risk of cystic kidney disease (IVW-MR: odds ratio [OR] = 3.05, 95% confidence interval [CI] = 1.19-7.84, p = 0.02) and PKD (IVW-MR: OR = 2.13, 95% CI = 1.01-4.46; p = 0.045). There was no evidence of causal effects of PCSK9 and NPC1L1 targets on cystic kidney disease and PKD. Cochran's Q test, MR-PRESSO, and MR-Egger intercept tests showed no heterogeneity or horizontal pleiotropy among the instrumental variables. CONCLUSIONS This study supported that increased HMGCR expression was associated with an increased risk of cystic kidney disease and PKD, suggesting potential benefits of statin therapy for cystic kidney disease and PKD. Further research is necessary to elucidate specific mechanisms and potential therapeutic applications of HMGCR inhibitors.
Collapse
Affiliation(s)
- Zhiwen Lian
- Division of Nephrology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Zijie Liang
- Division of Nephrology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Qiyan Chen
- Division of Nephrology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Chao Xie
- Division of Nephrology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Yaozhong Kong
- Division of Nephrology, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| |
Collapse
|
2
|
Liu X, Han J, Li X, Zhang Z, Li J, Yao Y. GPR146 regulates CYP7A1 transcription in cells and in vivo of mice. Biochem Biophys Res Commun 2025; 772:152045. [PMID: 40414012 DOI: 10.1016/j.bbrc.2025.152045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025]
Abstract
G protein-coupled receptor 146 (GPR146) plays a significant role in cholesterol metabolism in both humans and mice. Previous studies have shown that Gpr146 in mouse liver regulates cholesterol metabolism during long-term starvation, short-term starvation, and feeding conditions. Specifically, Gpr146 suppresses endogenous cholesterol synthesis and very-low-density lipoprotein secretion following feeding. However, its role in cholesterol metabolism under other feeding conditions remains unclear. The conversion of cholesterol to bile acids represents the primary pathway of cholesterol metabolism, with cytochrome P450 family 7 subfamily A member 1 (CYP7A1) serving as the critical rate-limiting enzyme. Studies have indicated that overexpression of Cyp7a1 can lower blood cholesterol levels. In this study, we systematically identified CYP7A1 as a target gene of GPR146 both in vivo and in cultured hepatocytes. Our findings revealed that silencing the expression of gpr146 in the mouse liver significantly reduced total blood cholesterol levels while markedly upregulating liver cyp7a1 expression during a 2-h fasting period. Importantly, this regulation occurs independently of farnesoid X-activated receptor (FXR)-dependent and FXR-independent cytokine pathways. These results strongly suggest that CYP7A1 is a crucial endogenous mediator of GPR146 in cholesterol metabolism both in vitro and in vivo.
Collapse
Affiliation(s)
- Xiao Liu
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Jinxin Han
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xuedan Li
- Wuhan Institute of Biological Products Co. Ltd., 430207, Wuhan, China
| | - Zhegang Zhang
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, 430207, Wuhan, China
| | - Jiawen Li
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yuxuan Yao
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
3
|
Skoumas I, Andrikou I, Simantiris S, Grigoriou K, Dima I, Terentes-Printzios D, Papanikolaou A, Akinosoglou K, Tsioufis K, Vlachopoulos C. Lipoprotein(a) in familial dyslipidemias: The effect on cardiovascular prognosis in patients with familial hypercholesterolemia or familial combined hyperlipidemia. Nutr Metab Cardiovasc Dis 2025; 35:103867. [PMID: 39986939 DOI: 10.1016/j.numecd.2025.103867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND AND AIMS Familial dyslipidemias are associated with increased cardiovascular risk. Increased lipoprotein(a) [Lp(a)] is considered as the most prevalent monogenic lipid disorder. The objective of the study was to identify the cardiovascular prognosis of patients with familial dyslipidemias (heterozygous familial hypercholesterolemia (FH) or familial combined hyperlipidemia (FCH)), without cardiovascular disease at baseline, investigating in parallel the effect of Lp(a). METHODS AND RESULTS 909 patients with FH (n = 433, mean age 44.2 ± 12.8 years) or FCH (n = 476, mean age 49.0 ± 11.1 years) were evaluated during a mean period of 10 years. The main endpoint was the composite of major cardiovascular events. The incidence of major cardiovascular events in the total population was 6.6 %, while greater in patients with FH compared to patients with FCH (8.1 % vs 5.5 %, p = 0.03). Multiple Cox regression analysis revealed that FH patients had greater cardiovascular risk compared to FCH patients (HR 2.17, 95 % CI 1.10-4.26, p = 0.02). In FH patients, increased baseline Lp(a) (≥30 mg/dl) was an independent predictor of adverse cardiovascular events (HR 2.37 95 % CI 1.41-4.90, p = 0.02), whereas in FCH patients was not. In FCH patients the presence of diabetes at baseline was a strong independent prognosticator of adverse cardiovascular events (HR 3.56 95 % CI 1.19-11.33, p = 0.03), after adjustment for confounders. CONCLUSIONS FH patients demonstrate double cardiovascular risk compared to FCH patients. In FH patients increased Lp(a) doubles the cardiovascular risk, beyond low density lipoprotein cholesterol. In FCH patients the presence of diabetes triples the cardiovascular risk, beyond Lp(a) which does not seem to convey an independent prognostic value.
Collapse
Affiliation(s)
- Ioannis Skoumas
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Andrikou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Spyridon Simantiris
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Grigoriou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Dima
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelos Papanikolaou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Karolina Akinosoglou
- Department of Internal Medicine and Infectious Diseases, University General Hospital of Patras, University of Patras, Patras, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Vlachopoulos
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
4
|
Shriner D, Bentley AR, Doumatey AP, Zhou J, Chen G, Rotimi CN, Adeyemo AA. Three Loci Affecting Variance of Body Mass Index in African Americans and Sub-Saharan Africans. Genet Epidemiol 2025; 49:e70009. [PMID: 40323147 PMCID: PMC12051743 DOI: 10.1002/gepi.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/08/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
Conventional genome-wide association studies (GWAS) are designed to assess the effect of a genetic locus on phenotypic mean by genotype. Such loci explain a proportion of phenotypic variance known as narrow-sense heritability. In contrast, variance quantitative trait loci (vQTL) are associated with the phenotypic variance by genotype. These loci explain an additional proportion of phenotypic variance and contribute to broad-sense heritability but not to narrow-sense heritability. Here, a genome-wide vQTL analysis in 22,805 African Americans yielded eight loci for body mass index (BMI). Of these loci, three were replicated in 6002 sub-Saharan Africans. No locus reached genome-wide significance using the standard additive model. Furthermore, no locus showed evidence for natural selection, haplotype effects, or gene × sex or gene × study interactions. Two loci showed evidence for an effect of locus-specific ancestry resulting from admixture and for a gene × gene interaction. One locus showed evidence for interaction with diastolic blood pressure, consistent with this vQTL capturing an unmodeled gene × covariate interaction. These analyses demonstrate that relevant BMI loci can be detected by evaluating vQTL and that these loci contribute to the underexplored broad-sense heritability for this trait.
Collapse
Affiliation(s)
- Daniel Shriner
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Amy R. Bentley
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Ayo P. Doumatey
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Jie Zhou
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Guanjie Chen
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Charles N. Rotimi
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Adebowale A. Adeyemo
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| |
Collapse
|
5
|
Stensrud VH, Rogne T, Flatby HM, Mohus RM, Gustad LT, Nilsen TIL. Examining socioeconomic differences in sepsis risk and mediation by modifiable factors: a Mendelian randomization study. BMC Infect Dis 2025; 25:739. [PMID: 40410669 PMCID: PMC12103053 DOI: 10.1186/s12879-025-11130-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Educational attainment is inversely related to sepsis risk, but the causal nature is still unclear. We therefore conducted the first Mendelian randomization (MR) study of genetically predicted educational attainment on sepsis that also uses a within-family genetic instrument for education. To further explore possible mechanistic pathways that can inform strategies to reduce sepsis risk, we examined the mediating effects of factors that are modifiable or can be prevented. METHODS The association between genetically predicted educational attainment and sepsis was estimated using summary-level data from recent genome-wide association studies. Possible bias due to population stratification, dynastic effects, and assortative mating in the genetic instrument for education was evaluated using summary-level data from a within-sibship genome-wide association study. We used inverse variance weighted MR analysis to estimate the effect of one standard deviation increase in years of education on sepsis risk. The robustness of the findings was assessed in sensitivity analyses, applying weighted median, weighted mode, and MR Egger regression. Finally, we applied multivariable MR analyses to estimate the mediating effects of smoking initiation, alcohol consumption, body mass index, high-density lipoprotein (HDL)-cholesterol, systolic blood pressure and type 2 diabetes. RESULTS For each standard deviation increase in genetically predicted educational attainment (3.4 years), the odds ratio (OR) for sepsis was 0.72 (95% confidence interval (CI) 0.66 to 0.78). The results of the analysis using the within-sibship genetic instrument and other sensitivity analyses were in line with this finding: within-sibship OR 0.88 (95% CI 0.64 to 1.18), weighted median OR 0.70 (95% CI 0.62 to 0.80), weighted mode OR 0.70 (95% CI 0.43 to 1.13), and MR Egger OR 0.65 (95% CI 0.50 to 0.85). The mediation analysis showed that 56% of the effect of educational attainment on sepsis risk can be explained by modifiable or preventable factors. CONCLUSIONS Higher educational attainment is strongly associated with a reduced risk of sepsis, pointing to important socioeconomic differences in this disease. The results also suggest that interventions targeting modifiable or preventable factors could contribute to reducing the socioeconomic differences in sepsis risk.
Collapse
Affiliation(s)
- Vilde Hatlevoll Stensrud
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Tormod Rogne
- Department of Community Medicine and Global Health, University of Oslo, Oslo, Norway
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Helene Marie Flatby
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Randi Marie Mohus
- Mid-Norway Centre for Sepsis Research, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Anaesthesia and Intensive Care, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Lise Tuset Gustad
- Faculty of Nursing and Health Sciences, Nord University, Levanger, Norway
- Department of Internal Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Tom Ivar Lund Nilsen
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Emergency Medicine and Prehospital Care, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
6
|
Yan W, Kong L, He T, Guo G, Zhu Q, Xi X, Fang M. Association between metabolic syndrome and low back pain: a two-sample Mendelian randomization study. Sci Rep 2025; 15:17686. [PMID: 40399540 PMCID: PMC12095482 DOI: 10.1038/s41598-025-02630-7] [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: 11/05/2024] [Accepted: 05/14/2025] [Indexed: 05/23/2025] Open
Abstract
This study uses two-sample MR analysis with GWAS summary statistics to evaluate the causal relationship between metabolic syndrome and low back pain. A two-sample Mendelian randomization analysis used GWAS summary statistics for low back pain from the FinnGen database and metabolic syndrome data, including waist circumference, hypertension, fasting blood glucose, HDL cholesterol, and triglyceride levels. Various methods like inverse variance weighted, MR-Egger, weighted median, and mode assessed the causal relationship, with sensitivity analyses addressing heterogeneity and pleiotropy. Our analysis found a statistically significant causal association between essential hypertension (OR 2.38, 95% CI 1.42-3.96; Padj = 0.002), metabolic syndrome (OR 1.05, 95% CI 1.01-1.10; Padj = 0.023) and waist circumference (OR 49, 95% CI 1.32-1.68; Padj < 0.001) and low back pain (OR 1.41, 95% CI 1.30-1.53, Padj < 0.001). In contrast, fasting blood glucose (FBG), HDL cholesterol, and triglycerides showed no significant associations with low back pain across all MR methods. The results of sensitivity analyses indicated that the heterogeneity and pleiotropy were unlikely to disturb the causal estimate. Our study indicates that increased essential hypertension, metabolic syndrome and waist circumference is causally associated with a higher risk of low back pain. Interventions targeting metabolic syndrome components, particularly blood pressure control and weight management, could help reduce the risk of low back pain. Further research is needed to explore the underlying biological pathways linking these metabolic factors to low back pain.
Collapse
Affiliation(s)
- Wei Yan
- Department of Clinical Medicine School, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingjun Kong
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianxiang He
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangxin Guo
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qingguang Zhu
- Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Institute of Tuina, Shanghai Institute of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaobing Xi
- Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Fang
- Department of Tuina, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Institute of Tuina, Shanghai Institute of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
7
|
Chen Y, Liu P, Sabo A, Guan D. Human genetic variation determines 24-hour rhythmic gene expression and disease risk. Nat Commun 2025; 16:4270. [PMID: 40341583 PMCID: PMC12062405 DOI: 10.1038/s41467-025-59524-5] [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: 07/23/2024] [Accepted: 04/24/2025] [Indexed: 05/10/2025] Open
Abstract
24-hour biological rhythms are essential to maintain physiological homeostasis. Disruption of these rhythms increases the risks of multiple diseases. Biological rhythms are known to have a genetic basis formed by core clock genes, but how individual genetic variation shapes the oscillating transcriptome and contributes to human chronophysiology and disease risk is largely unknown. Here, we mapped interactions between temporal gene expression and genotype to identify quantitative trait loci (QTLs) contributing to rhythmic gene expression. These newly identified QTLs were termed as rhythmic QTLs (rhyQTLs), which determine previously unappreciated rhythmic genes in human subpopulations with specific genotypes. Functionally, rhyQTLs and their associated rhythmic genes contribute extensively to essential chronophysiological processes, including bile acid and lipid metabolism. The identification of rhyQTLs sheds light on the genetic mechanisms of gene rhythmicity, offers mechanistic insights into variations in human disease risk, and enables precision chronotherapeutic approaches for patients.
Collapse
Affiliation(s)
- Ying Chen
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Panpan Liu
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Aniko Sabo
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Dongyin Guan
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
8
|
Ma F, Longo M, Meroni M, Bhattacharya D, Paolini E, Mughal S, Hussain S, Anand SK, Gupta N, Zhu Y, Navarro-Corcuera A, Li K, Prakash S, Cogliati B, Wang S, Huang X, Wang X, Yurdagul A, Rom O, Wang L, Fried SK, Dongiovanni P, Friedman SL, Cai B. EHBP1 suppresses liver fibrosis in metabolic dysfunction-associated steatohepatitis. Cell Metab 2025; 37:1152-1170.e7. [PMID: 40015280 PMCID: PMC12058419 DOI: 10.1016/j.cmet.2025.01.020] [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: 06/22/2023] [Revised: 11/27/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
Excess cholesterol accumulation contributes to fibrogenesis in metabolic dysfunction-associated steatohepatitis (MASH), but how hepatic cholesterol metabolism becomes dysregulated in MASH is not completely understood. We show that human fibrotic MASH livers have decreased EH-domain-binding protein 1 (EHBP1), a genome-wide association study (GWAS) locus associated with low-density lipoprotein (LDL) cholesterol, and that EHBP1 loss- and gain-of-function increase and decrease MASH fibrosis in mice, respectively. Mechanistic studies reveal that EHBP1 promotes sortilin-mediated PCSK9 secretion, leading to LDL receptor (LDLR) degradation, decreased LDL uptake, and reduced TAZ, a fibrogenic effector. At a cellular level, EHBP1 deficiency affects the intracellular localization of retromer, a protein complex required for sortilin stabilization. Our therapeutic approach to stabilizing retromer is effective in mitigating MASH fibrosis. Moreover, we show that the tumor necrosis factor alpha (TNF-α)/peroxisome proliferator-activated receptor alpha (PPARα) pathway suppresses EHBP1 in MASH. These data not only provide mechanistic insights into the role of EHBP1 in cholesterol metabolism and MASH fibrosis but also elucidate an interplay between inflammation and EHBP1-mediated cholesterol metabolism.
Collapse
Affiliation(s)
- Fanglin Ma
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Marica Meroni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Shama Mughal
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Syed Hussain
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Neha Gupta
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiwei Zhu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amaia Navarro-Corcuera
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth Li
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Satya Prakash
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruno Cogliati
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xin Huang
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Liheng Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
9
|
Xing S, Zhang Y, Chen Y, Feng S, Zhang Y, Moreira P. Comparing the impacts of different exercise interventions on patients with type 2 diabetes mellitus: a literature review and meta-analysis. Front Endocrinol (Lausanne) 2025; 16:1495131. [PMID: 40391012 PMCID: PMC12086073 DOI: 10.3389/fendo.2025.1495131] [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: 09/14/2024] [Accepted: 03/20/2025] [Indexed: 05/29/2025] Open
Abstract
Objective Exercise interventions are a recommended method of diabetes management through which patients can achieve blood glucose control, increase muscle volume, and improve insulin sensitivity, while also improving blood lipids, blood pressure, and cardiovascular health. A few studies on the effects of physical exercise on diabetic patients have been published in recent years. This article focuses on exploring evidence on which exercise interventions generate which effects in diabetic patients, namely, high-intensity interval training (HIIT), method training (MT), aerobic exercise training (AET), resistance training (RT), and combined training (CBT). Methods Randomized controlled trials (RCTs) that focused on the effects of exercise interventions on blood glucose and blood lipids of patients with type 2 diabetes mellitus were reviewed. A network meta-analysis was performed to compare the effects of the five exercise interventions in diabetic patients, namely the impacts on glycosylated hemoglobin (HbA1c), fasting blood glucose (FBG), total cholesterol (TC), triglycerides (TG), high-density lipoprotein (HDL), and low-density lipoprotein (LDL). The study was strictly conducted following the PRISMA Protocol, and the Cochrane Risk of Bias Assessment Tool 2.0 was used to objectively evaluate the risk of bias in the implementation of the study. Results This review included 25 RCTs in total, with 1,711 subjects. Meta-analysis suggests that, compared with conventional therapeutic treatment, exercise interventions can reduce blood glucose indexes, namely HbA1c, FBG, TC, TG, HDL, and LDL. RT and AET have been shown to reduce TC; HIIT, MT, AET, and CBT have been shown to improve HDL; and HIIT, MT, AET, and CBT have been shown to improve HDL. The MT and RT exercise types can reduce LDL. Evidence also suggests that MT can lower HbA1c, TG, and LDL levels, and RT lowers cholesterol levels. HIIT exercise appears to improve FBG and HDL levels. Conclusion The five types of exercise generate different effects on the key clinical dimensions of diabetes. MT seems to be the optimal choice to improve HbA1c, TG levels, and LDL, while HIIT improves FBG and HDL levels, whereas RT exercise appears to be the optimal exercise to lower cholesterol levels.
Collapse
Affiliation(s)
- Shuangtao Xing
- Physical Education Institute of Henan Normal University, Henan, Xinxiang, China
| | - Yifan Zhang
- Department of Public Basic Education of Henan Vocational University of Science and Technology, Henan, Zhoukou, China
| | - Yanjiao Chen
- Research Center for Social Work and Governance, College of Social Affairs, Henan Normal University, Henan, Xinxiang, China
| | - Shijie Feng
- Physical Education Institute of Henan Normal University, Henan, Xinxiang, China
| | - Yiqing Zhang
- Research Center for Social Work and Governance, College of Social Affairs, Henan Normal University, Henan, Xinxiang, China
| | - Paulo Moreira
- International Healthcare Management Research & Development Center (IHM-RDC), First Affiliated Hospital of the Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Atlantica Instituto Universitario, Gestao em Saude, Oeiras, Portugal
| |
Collapse
|
10
|
Cuttone A, Cannavò V, Abdullah RMS, Fugazzotto P, Arena G, Brancati S, Muscarà A, Morace C, Quartarone C, Ruggeri D, Squadrito G, Russo GT. Expanding the Use of SGLT2 Inhibitors in T2D Patients Across Clinical Settings. Cells 2025; 14:668. [PMID: 40358192 PMCID: PMC12071329 DOI: 10.3390/cells14090668] [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: 03/26/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2i) are currently recommended in patients with type 2 diabetes (T2D) to reduce serum glucose levels. Moreover, robust evidence has clearly demonstrated their beneficial cardiovascular and renal effects, making this class of drugs pivotal for the treatment of T2D, especially when complicated by diabetic kidney disease or heart failure. However, several other comorbidities are frequently encountered in T2D patients beyond these long-term diabetes complications, especially in the internal medicine setting. For some of these comorbidities, such as MAFLD and cognitive impairment, the association with diabetes is increasingly recognized, with the hypothesis of a common pathophysiologic background, whereas, for others, a coincident epidemiology linked to the ageing of populations, including that of T2D subjects, may be advocated. In the effort of personalizing T2D treatment, evidence on the potential effects of SGLT2i in these different clinical conditions is accumulating. The purpose of this narrative review is to update current literature on the effects of SGLT2i for the treatment of T2D in different clinical settings beyond glycaemic control, and to elucidate potential molecular mechanisms by which they exert these effects.
Collapse
Affiliation(s)
- Alessandro Cuttone
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Vittorio Cannavò
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Raouf Mastan Sheik Abdullah
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Pierluigi Fugazzotto
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giada Arena
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Simona Brancati
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Andrea Muscarà
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Carmela Morace
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Cristina Quartarone
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Domenica Ruggeri
- Internal Medicine and Diabetology Unit, University Hospital of Messina, 98124 Messina, Italy; (C.Q.); (D.R.)
| | - Giovanni Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| | - Giuseppina Tiziana Russo
- Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy; (V.C.); (R.M.S.A.); (P.F.); (G.A.); (S.B.); (A.M.); (C.M.); (G.S.); (G.T.R.)
| |
Collapse
|
11
|
Szatrowski A, Maggio Z, Khomtchouk B. HDL Cholesterol Is Remarkably Cardioprotective Against Coronary Artery Disease in Native Hawaiians and Pacific Islanders. JACC. ADVANCES 2025; 4:101741. [PMID: 40319838 PMCID: PMC12124629 DOI: 10.1016/j.jacadv.2025.101741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND High-density lipoprotein cholesterol (HDL-C) is inversely associated with cardiometabolic risk and exhibits nonlinear effects at extreme levels. Cardiometabolic diseases are a leading cause of death and are particularly prevalent among Native Hawaiian and Pacific Islanders (NHPIs). OBJECTIVES This study characterizes HDL-C's association with coronary artery disease (CAD), major adverse cardiovascular events (MACE), and type 2 diabetes (T2D) in NHPIs compared to the general population. METHODS Using electronic health record data from the National Institutes of Health All of Us Research Program, we applied Cox proportional hazards models to compare HDL-C's protective effects on CAD, MACE, and T2D between 261 NHPIs and the remaining cohort (n = 188,802). Models were adjusted for key confounders, and restricted cubic splines were used to assess nonlinear risk dynamics. RESULTS Tracking individuals across 10,534,661 person-years (mean age 55.7 ± 15.8 years, 38% male), HDL-C was more strongly associated with reduced CAD risk in NHPIs (HR: 0.32; 95% CI: 0.19-0.54) than in the general cohort (HR: 0.57; 95% CI: 0.56-0.58). A marginally stronger association was observed for MACE (NHPI HR: 0.40; 95% CI: 0.23-0.71 vs general HR: = 0.54; 95% CI: 0.53-0.56), while T2D associations were similar. Spline analysis indicated that low HDL-C increases risk for both CAD and T2D in NHPIs. CONCLUSIONS HDL-C's protective role against cardiometabolic diseases is more pronounced in NHPIs, particularly for CAD. These findings support further investigation into tailored clinical assessments for this population.
Collapse
Affiliation(s)
| | - Zane Maggio
- The College of the University of Chicago, Chicago, Illinois, USA; Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Indiana University, Indianapolis, Indiana, USA
| | - Bohdan Khomtchouk
- Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Indiana University, Indianapolis, Indiana, USA.
| |
Collapse
|
12
|
Fan Z, Su H, Qiao T, Shi S, Shi P, Zhang A. TEX10: A Novel Drug Target and Potential Therapeutic Direction for Sleep Apnea Syndrome. Nat Sci Sleep 2025; 17:731-746. [PMID: 40330585 PMCID: PMC12053781 DOI: 10.2147/nss.s499895] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Background Sleep apnea syndrome (SAS) is a prevalent sleep disorder strongly associated with obesity, metabolic dysregulation, and cardiovascular diseases. While its underlying pathophysiological mechanisms remain incompletely understood, genetic factors likely play a pivotal role in SAS pathogenesis. This study investigates the causal relationships between potential drug target genes and SAS using multiple statistical approaches, aiming to provide novel insights for targeted therapeutic development. Methods We conducted a comprehensive genetic analysis integrating multiple methodologies to investigate gene-SAS relationships. Using publicly available GWAS and eQTL databases, we performed Mendelian Randomization (MR) analysis with the inverse variance weighted (IVW) method, validated by weighted median and MR-Egger approaches. Summary-data-based MR (SMR) analysis, coupled with HEIDI testing, assessed direct gene expression-SAS associations while controlling for linkage disequilibrium (LD). Colocalization analysis evaluated the probability of shared causal variants between SNPs, gene expression, and SAS. Statistical significance was determined using Benjamini-Hochberg multiple testing correction (FDR < 0.05). Additionally, mediation analysis explored TEX10's influence on SAS through metabolic intermediates including BMI, waist circumference, and HDL cholesterol. Results We identified 18 candidate drug target genes significantly associated with SAS, with MAPKAPK3, TNXB, MPHOSPH8, and TEX10 showing consistent associations across multiple analyses. TEX10, in particular, exhibited significant associations with SAS risk in blood, cerebral cortex, hippocampus, and basal ganglia (PP.H4 > 0.9). Mediation analysis suggested that TEX10 might influence SAS risk indirectly through BMI, waist circumference, and HDL cholesterol levels. Conclusion Our study identified multiple potential therapeutic targets causally linked to SAS, with TEX10 emerging as a key candidate gene. These findings advance our understanding of SAS pathogenesis and offer promising directions for personalized diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Zhitao Fan
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Hui Su
- Department of Neurosurgery, Xingtai People’s Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Tong Qiao
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Sunan Shi
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Pengfei Shi
- Department of Ophthalmology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| | - Anqi Zhang
- Department of Otorhinolaryngology, Hebei Eye Hospital, Xingtai, Hebei Province, People’s Republic of China
| |
Collapse
|
13
|
Ma K, Wang H, Du Y, Chen T, Yang D, Li Y, Li D. Mendelian Randomization Assessment of the Genetic Effects of Lipid-Lowering Drugs on Digestive System Cancers. Food Sci Nutr 2025; 13:e70293. [PMID: 40443776 PMCID: PMC12121511 DOI: 10.1002/fsn3.70293] [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: 08/06/2024] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 06/02/2025] Open
Abstract
The relationship between lipid-lowering drugs and the risk of digestive system cancers remains unclear. This study aims to assess the risk association between lipid-lowering drugs and digestive system cancers through mendelian randomization (MR) analysis. We utilized genetic instruments to substitute for the exposure to lipid-lowering drugs, including expression quantitative trait loci (eQTL) for HMGCR, PCSK9, and NPC1L1, as well as genetic variants associated with low-density lipoprotein (LDL) from the Global Lipids Genetics Consortium's genome-wide association study (GWAS) data for target genes. We used MR and SMR methods to assess the risk estimates of lipid-lowering drug target genes on digestive system tumors. The MR analysis indicated a negative association between HMGCR-mediated LDL and hepatocellular carcinoma (OR = 0.06, 95% CI: 0.00-0.81, p = 0.03), and a positive association between NPC1L1-mediated LDL and gastric cancer risk (OR = 15.45, 95% CI: 5.96-40.56, p < 0.01). In the SMR analysis, it was observed that HMGCR expression decreased the risk of hepatocellular carcinoma (OR = 0.11, 95% CI: 0.02-0.68, p = 0.02), while NPC1L1 expression increased the risk of gastric cancer (OR = 1.33, 95% CI: 1.08-1.64, p < 0.01). Our study results suggested a potential risk association between HMGCR inhibitors and NPC1L1 with hepatocellular carcinoma and gastric cancer.
Collapse
Affiliation(s)
- Keru Ma
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbinChina
| | - Hao Wang
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbinChina
| | - Yubo Du
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbinChina
| | - Tianyu Chen
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbinChina
| | - Dongxu Yang
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbinChina
| | - Yue Li
- Department of Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
| | - Dalin Li
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbinChina
| |
Collapse
|
14
|
Li Q, Qu Y, Xue J, Kang H, Lyu C. Exploring lipid-modifying therapies for sepsis through the modulation of circulating inflammatory cytokines: a Mendelian randomization study. World J Emerg Med 2025; 16:256-261. [PMID: 40406288 PMCID: PMC12093423 DOI: 10.5847/wjem.j.1920-8642.2025.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/02/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Whether lipid-modifying drugs directly impact the outcome of sepsis remains uncertain. Therefore, systematic investigations are needed to explore the potential impact of lipid-related therapies on sepsis outcomes and to elucidate the underlying mechanisms involving circulating inflammatory cytokines, which may play critical roles in the pathogenesis of sepsis. This study aimed to utilize drug-target Mendelian randomization to assess the direct causal effects of genetically proxied lipid-modifying therapies on sepsis outcomes. METHODS First, a two-sample Mendelian randomization study was conducted to validate the causal associations among high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and sepsis. A subsequent drug-target Mendelian randomization study assessed the direct causal effects of genetically proxied lipid-modifying therapies on the risk of sepsis, sepsis-related critical care admission, and sepsis-related death. The identified lipid-modifying drug targets were subsequently explored for direct causal relationships with 36 circulating inflammatory cytokines. Finally, enrichment analyses of the identified cytokines were conducted to explore the potential relationships of lipid-modifying drugs with the inflammatory response. RESULTS Genetically proxied cholesteryl ester transfer protein (CETP) inhibitors were significantly associated with sepsis-related critical care admission (OR=0.84, 95% CI [0.74, 0.95], P=0.008,) and sepsis-related death (OR=0.68, 95% CI [0.52, 0.88], P=0.004). The genetically proxied CETP inhibitors were strongly associated with the levels of 15 circulating inflammatory cytokines. Enrichment analyses indicated that CETP inhibitors may modulate inflammatory cytokines and influence the inflammatory response pathway. CONCLUSION This study supports a causal effect of genetically proxied CETP inhibitors in reducing the risk of sepsis-related critical care admission and death. These findings suggest that the underlying mechanism may involve the modulation of some circulating inflammatory cytokines, influencing the inflammatory response pathway.
Collapse
Affiliation(s)
- Quan Li
- Department of Emergency Medicie, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Yun Qu
- Department of Emergency Medicie, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Jinfang Xue
- Emergency Department, the State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Hai Kang
- Department of Emergency Medicie, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Chuanzhu Lyu
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology ofChina, Chengdu 610072, China
| |
Collapse
|
15
|
Amente LD, Mills NT, Le TD, Hyppönen E, Lee SH. A latent outcome variable approach for Mendelian randomization using the stochastic expectation maximization algorithm. Hum Genet 2025; 144:559-574. [PMID: 40214754 PMCID: PMC12033120 DOI: 10.1007/s00439-025-02739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/18/2025] [Indexed: 04/27/2025]
Abstract
Mendelian randomization (MR) is a widely used tool to uncover causal relationships between exposures and outcomes. However, existing MR methods can suffer from inflated type I error rates and biased causal effects in the presence of invalid instruments. Our proposed method enhances MR analysis by augmenting latent phenotypes of the outcome, explicitly disentangling horizontal and vertical pleiotropy effects. This allows for explicit assessment of the exclusion restriction assumption and iteratively refines causal estimates through the expectation-maximization algorithm. This approach offers a unique and potentially more precise framework compared to existing MR methods. We rigorously evaluate our method against established MR approaches across diverse simulation scenarios, including balanced and directional pleiotropy, as well as violations of the Instrument Strength Independent of Direct Effect (InSIDE) assumption. Our findings consistently demonstrate superior performance of our method in terms of controlling type I error rates, bias, and robustness to genetic confounding, regardless of whether individual-level or summary data is used. Additionally, our method facilitates testing for directional horizontal pleiotropy and outperforms MR-Egger in this regard, while also effectively testing for violations of the InSIDE assumption. We apply our method to real data, demonstrating its effectiveness compared to traditional MR methods. This analysis reveals the causal effects of body mass index (BMI) on metabolic syndrome (MetS) and a composite MetS score calculated by the weighted sum of its component factors. While the causal relationship is consistent across most methods, our proposed method shows fewer violations of the exclusion restriction assumption, especially for MetS scores where horizontal pleiotropy persists and other methods suffer from inflation.
Collapse
Affiliation(s)
- Lamessa Dube Amente
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia.
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5000, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
- Epidemiology Department, Jimma University, 378, Jimma, Ethiopia.
| | - Natalie T Mills
- Discipline of Psychiatry, University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Thuc Duy Le
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - S Hong Lee
- Australian Centre for Precision Health, University of South Australia, Adelaide, SA, 5000, Australia.
- UniSA Allied Health and Human Performance, University of South Australia, Adelaide, SA, 5000, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia.
| |
Collapse
|
16
|
Packard CJ, Taskinen MR, Björnson E, Matikainen N, Söderlund S, Andersson L, Adiels M, Borén J. Genetically determined increase in apolipoprotein C-III (APOC3 gain-of-function) delays very low-density lipoprotein clearance in humans. Atherosclerosis 2025; 404:119166. [PMID: 40203662 DOI: 10.1016/j.atherosclerosis.2025.119166] [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: 01/15/2025] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
AIM Apolipoprotein C-III (apoC-III) is an important regulator of triglyceride (TG) metabolism and a target for intervention. The present study examined the effects of gain-of-function (GOF) variants in APOC3 on apolipoprotein B kinetics to understand further how changes in the synthesis of this apolipoprotein impact triglyceride-rich lipoprotein (TRL) metabolism. METHODS Two groups of subjects were recruited by population screening, 9 carriers of known APOC3 GOF variants and 9 age-, sex- and BMI-matched non-carriers. The kinetics of TRL were determined using stable isotope tracers of apoprotein and triglyceride metabolism in a non-steady-state protocol involving administration of a fat-rich meal. RESULTS APOC3 GOF carriers had 47 % higher plasma apoC-III levels compared to non-carriers (P = 0.022) and higher production rates for the apolipoprotein. Post-prandial response (total area-under-curve) for plasma TG was 108 % greater in GOF carriers compared to non-carriers (P = 0.002) due specifically to higher levels of VLDL1. In contrast, no difference was seen in the chylomicron apoB48 response. Comparison of TRL kinetics between groups showed that APOC3 GOF carriers had lower fractional clearance rates for VLDL1-apoB100 and VLDL1-apoB48-containing particles (P < 0.02), but no difference in VLDL1-apoB100 or chylomicron apoB48 production rates. Both the rate of VLDL lipolysis and the rate of clearance of VLDL particles from the circulation were lower in APOC3 GOF carriers than in non-carriers. In contrast, chylomicron apoB clearance rates did not differ between APOC3 GOF carriers and non-carriers. CONCLUSION APOC3 GOF carriers showed specific alterations in TRL metabolism (compared to matched non-carriers), namely slower lipolysis and delayed clearance of VLDL1-sized particles, but no difference in chylomicron metabolism. Our findings suggest that intervention to reduce apoC-III production can be modelled as a reduction in TRL, particularly VLDL particle levels, without deleterious effects on fat absorption or hepatic VLDL production.
Collapse
Affiliation(s)
- C J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - M R Taskinen
- Research Programs Unit, Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - E Björnson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden
| | - N Matikainen
- Research Programs Unit, Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland; Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - S Söderlund
- Research Programs Unit, Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland; Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - L Andersson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden
| | - M Adiels
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden
| | - J Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden; Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
17
|
Guo X, Wu L, Lai J, Wu Y, Chen D. Causal Associations Between Lipids, NPC1L1, and Liver Cancer Risk: Insights From Mendelian Randomization and Bioinformatics. J Gastroenterol Hepatol 2025. [PMID: 40312834 DOI: 10.1111/jgh.16897] [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: 12/06/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND AND AIM The study aims to investigate the potential causal effects of lipids on liver cancer risk and to analyze the possible impact of lipid-lowering drug targets on liver cancer. METHODS Genetic variants linked to lipid traits and drug targets were obtained from the Global Lipids Genetics Consortium and DrugBank. Liver cancer data were sourced from FinnGen. Mendelian randomization (MR) was used to assess causal relationships between lipid traits and liver cancer. Functional analyses included protein-protein interaction (PPI), KEGG pathway enrichment, transcription factor (TF) network analysis, and survival analysis. NPC1L1 expression, DNA methylation, and immune infiltration were analyzed using UALCAN, TCGA-LIHC, and TIMER, respectively. RESULTS MR analysis showed higher genetically predicted LDL-C levels reduced liver cancer risk (OR = 0.5981, p = 0.034). Drug target MR indicated that NPC1L1 inhibition (OR = 1.0638, p = 0.0311) and elevated PPARɑ levels (OR = 1.1339, p < 0.01) increased liver cancer risk. Functional analysis revealed NPC1L1 was highly expressed in liver cancer tissues due to hypomethylation and linked to immune cell infiltration, indicating its role in immune evasion and tumor progression. CONCLUSION The study demonstrates that elevated LDL-C levels are associated with a reduced risk of liver cancer and NPC1L1 plays a key role in regulating lipid metabolism and influencing immune evasion.
Collapse
Affiliation(s)
- Xiaoyan Guo
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lili Wu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Lai
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuankai Wu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dianke Chen
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Wu X, Yang Q, Xie Y, Xia L, Li J, An W, Lu X. Drug-targeted Mendelian randomization analysis combined with transcriptome sequencing to explore the molecular mechanisms associated with cognitive impairment. J Alzheimers Dis 2025:13872877251335891. [PMID: 40267292 DOI: 10.1177/13872877251335891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundCurrent therapies for cognitive impairment, including Alzheimer's disease (AD) and mild cognitive impairment, are limited by a lack of universal treatment and adverse effects associated with polypharmacy. Investigating genetic and molecular mechanisms underlying cognitive decline is critical for the development of targeted therapeutics.ObjectiveTo identify causal genes and potential therapeutic targets for cognitive impairment through integrative genomic analyses.MethodsGenome-wide association study data on cognitive impairment were combined with the expression quantitative trait loci (eQTL) data from the eQTLGen consortium. Mendelian randomization (MR) and colocalization analyses were employed to infer causal relationships. Gene Set Enrichment Analysis and Gene Set Variation Analysis evaluated the pathway and functional differences. Immune cell infiltration patterns and the immunometabolic pathways were assessed, followed by drug target prediction.ResultsMR analysis identified seven gene-eQTL pairs significantly associated with cognitive impairment. SMR colocalization prioritized three key genes: HNMT (histamine metabolism), TNFSF8 (inflammatory signaling), and S1PR5 (sphingolipid signaling). HNMT, TNFSF8, and S1PR5 had 39, 24, and 30 predicted targeted drugs, respectively, including arsenic trioxide, aspirin, and immunomodulators.ConclusionsThis study implicates HNMT, TNFSF8, and S1PR5 as potential therapeutic targets for cognitive impairment. Further validation is required to confirm their clinical relevance.
Collapse
Affiliation(s)
- Xixi Wu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Qingyan Yang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Yudi Xie
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Lingfeng Xia
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Jiatao Li
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Wenting An
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| | - Xiao Lu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Cao J, Su T, Chen S, Du Z, Lai C, Chi K, Li Q, Wang S, Wu Q, Hu Y, Fang Y, Hu Y, Zhu Z, Huang Y, Zhang X, Yu H. Evaluating lipid-lowering drug targets for full-course diabetic retinopathy. Br J Ophthalmol 2025:bjo-2024-325771. [PMID: 39900481 DOI: 10.1136/bjo-2024-325771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 01/09/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Implementing lipid control in patients with diabetes is regarded as a potential strategy for halting the advancement of diabetic retinopathy (DR). This study seeks to use Mendelian randomisation (MR) to assess the causal relationship between lipid traits and lipid-lowering drug targets and full-course DR (background DR, severe non-proliferative DR (NPDR) and proliferative DR (PDR)). METHODS A two-sample MR and drug target MR to decipher the causal effects of lipid traits and lipid-lowering drug targets on full-course DR, including background DR, severe NPDR and PDR, was conducted in the study. Genetic variants associated with lipid traits and genes encoding the protein targets of lipid-lowering drugs were extracted from the Global Lipids Genetics Consortium and UK Biobank. Summary-level data of full-course DR are obtained from FinnGen. RESULTS No significant causal relationship was found between lipid traits and full-course DR. However, in drug target MR analysis, peroxisome proliferator-activated receptor gamma (PPARG) enhancement was associated with lower risks of background DR (OR=0.12, p=0.005) and PDR (OR=0.25, p=0.006). Additionally, mediation MR analysis showed that lowering fasting insulin (p=0.015) and HbA1c (p=0.005) levels mediated most of the association between PPARG and full-course DR. CONCLUSIONS This study reveals PPARG may be a promising drug target for full-course DR. The activation of PPARG could reduce the risk of full-course DR, especially background DR and PDR. The mechanism of the PPARG agonists' protection of full-course DR may be dependent on the glucose-lowering effect.
Collapse
Affiliation(s)
- Jiahui Cao
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Su
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Shuilian Chen
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Zijing Du
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Chunran Lai
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Kaiyi Chi
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Qinyi Li
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shan Wang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Qiaowei Wu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yunyan Hu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Fang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yijun Hu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Zhuoting Zhu
- Royal Victorian Eye and Ear Hospital, Centre for Eye Research Australia Ltd, East Melbourne, Victoria, Australia
| | - Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Zhou G, Qie X, Zhao H. A Bayesian approach to correcting the attenuation bias of regression using polygenic risk score. Genetics 2025; 229:iyaf018. [PMID: 39891671 DOI: 10.1093/genetics/iyaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Polygenic risk score has become increasingly popular for predicting the value of complex traits. In many settings, polygenic risk score is used as a covariate in regression analysis to study the association between different phenotypes. However, measurement error in polygenic risk score causes attenuation bias in the estimation of regression coefficients. In this paper, we employ a Bayesian approach to accounting for the measurement error of polygenic risk score and correcting the attenuation bias in linear and logistic regression. Through simulation, we show that our approach is able to obtain approximately unbiased estimation of coefficients and credible intervals with correct coverage probability. We also empirically compare our Bayesian measurement error model with the conventional regression model by analyzing real traits in the UK Biobank. The results demonstrate the effectiveness of our approach as it significantly reduces the error in coefficient estimates.
Collapse
Affiliation(s)
- Geyu Zhou
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| | - Xinyue Qie
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
21
|
Schmidt HM, Jarrett KE, de Aguiar Vallim TQ, Tarling EJ. Pathways and Molecular Mechanisms Governing LDL Receptor Regulation. Circ Res 2025; 136:902-919. [PMID: 40208925 PMCID: PMC11989972 DOI: 10.1161/circresaha.124.323578] [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: 04/12/2025]
Abstract
Clearance of circulating plasma LDL (low-density lipoprotein) cholesterol by the liver requires hepatic LDLR (low-density lipoprotein receptor). Complete absence of functional LDLR manifests in severe hypercholesterolemia and premature atherosclerotic cardiovascular disease. Since the discovery of the LDLR 50 years ago by Brown and Goldstein, all approved lipid-lowering medications have been aimed at increasing the abundance and availability of LDLR on the surface of hepatocytes to promote the removal of LDL particles from the circulation. As such a critical regulator of circulating and cellular cholesterol, it is not surprising that LDLR activity is tightly regulated. Despite over half a century's worth of study, there are still many facets of LDLR biology that remain unexplored. This review will focus on pathways that regulate the LDLR and emerging concepts of LDLR biology.
Collapse
Affiliation(s)
- Heidi M. Schmidt
- Department of Medicine, Division of Cardiology, University of California Los Angeles, CA, USA
| | - Kelsey E. Jarrett
- Department of Medicine, Division of Cardiology, University of California Los Angeles, CA, USA
| | - Thomas Q. de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California Los Angeles, CA, USA
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
- Molecular Biology Institute, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
| | - Elizabeth J. Tarling
- Department of Medicine, Division of Cardiology, University of California Los Angeles, CA, USA
- Molecular Biology Institute, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
- Lead contact
| |
Collapse
|
22
|
Zhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier KR, Chittoor G, Josyula NS, Meyer M, Gupta S, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SLR, Kelly TN, Lange EM, LeNoir M, Li C, Le Marchand L, McDonald MLN, McHugh CP, Morrison AC, Naseri T, O'Connell J, O'Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao DC, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PWF, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Ida Chen YD, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, et alZhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier KR, Chittoor G, Josyula NS, Meyer M, Gupta S, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SLR, Kelly TN, Lange EM, LeNoir M, Li C, Le Marchand L, McDonald MLN, McHugh CP, Morrison AC, Naseri T, O'Connell J, O'Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao DC, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PWF, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Ida Chen YD, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, Kooperberg C, Minster RL, Mitchell BD, Nouraie M, Psaty BM, Raffield LM, Reiner AP, Rich SS, Rotter JI, Benjamin Shoemaker M, Smith NL, Taylor KD, Telen MJ, Weiss ST, Zhang Y, Heard-Costa N, Sun YV, Lin X, Cupples LA, Lange LA, Liu CT, Loos RJF, North KE, Justice AE. Whole genome sequencing analysis of body mass index identifies novel African ancestry-specific risk allele. Nat Commun 2025; 16:3470. [PMID: 40216759 PMCID: PMC11992084 DOI: 10.1038/s41467-025-58420-2] [Show More Authors] [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: 08/21/2023] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Obesity is a major public health crisis associated with high mortality rates. Previous genome-wide association studies (GWAS) investigating body mass index (BMI) have largely relied on imputed data from European individuals. This study leveraged whole-genome sequencing (WGS) data from 88,873 participants from the Trans-Omics for Precision Medicine (TOPMed) Program, of which 51% were of non-European population groups. We discovered 18 BMI-associated signals (P < 5 × 10-9), including two secondary signals. Notably, we identified and replicated a novel low-frequency single nucleotide polymorphism (SNP) in MTMR3 that was common in individuals of African descent. Using a diverse study population, we further identified two novel secondary signals in known BMI loci and pinpointed two likely causal variants in the POC5 and DMD loci. Our work demonstrates the benefits of combining WGS and diverse cohorts in expanding current catalog of variants and genes confer risk for obesity, bringing us one step closer to personalized medicine.
Collapse
Affiliation(s)
- Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanfei Xu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kendra R Ferrier
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Mariah Meyer
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Shreyash Gupta
- Population Health Sciences, Geisinger, Danville, PA, USA
| | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zilin Li
- Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
- School of Mathematics and Statistics and KLAS, Northeast Normal University, Changchun, Jilin, China
| | - Matthew A Allison
- Department of Family Medicine, Division of Preventive Medicine, The University of California San Diego, La Jolla, CA, USA
| | - Diane M Becker
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jai G Broome
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Erin J Buth
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Kyong-Mi Chang
- The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sameer Chavan
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yen-Feng Chiu
- Institute of Population Health Sciences, National Health Research Institutes, Taipei, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, Division of Metabolism/Endocrinology, National Taiwan University Hospital, Taipei, Taiwan
| | - Matthew P Conomos
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Dawn L DeMeo
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ravindranath Duggirala
- Life Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
- Department of Health and Behavioral Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Celeste Eng
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Alison E Fohner
- Epidemiology, Institute of Public Health Genetics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Barry I Freedman
- Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiuqing Guo
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Chris Haiman
- Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin D Heavner
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - James E Hixson
- Department of Epidemiology, School of Public Health, UTHealth Houston, Houston, TX, USA
| | - Yuk-Lam Ho
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Brian D Hobbs
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Donglei Hu
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Chii-Min Hwu
- Department of Medicine, Division of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei, Taiwan, Taiwan
| | | | - Deepti Jain
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Rita R Kalyani
- Department of Medicine, Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Ethan M Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael LeNoir
- Department of Pediatrics, Bay Area Pediatrics, Oakland, CA, USA
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Merry-Lynn N McDonald
- Department of Medicine, Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caitlin P McHugh
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Take Naseri
- Naseri & Associates Public Health Consultancy Firm and Family Health Clinic, Apia, Samoa
- International Health Institute, Brown University, Providence, RI, USA
| | - Jeffrey O'Connell
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, MD, USA
| | - Christopher J O'Donnell
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James A Perry
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D C Rao
- Center for Biostatistics and Data Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Elizabeth A Regan
- Department of Medicine, Rheumatology, National Jewish Health, Denver, CO, USA
| | | | - Dan M Roden
- Medicine, Pharmacology, and Biomedical Informatics, Clinical Pharmacology and Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | | | - Zeyuan Wang
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Daniel E Weeks
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer Wessel
- Department of Epidemiology, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
- Diabaetes Translational Research Center, Indiana University, Indianapolis, IN, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Peter W F Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lisa R Yanek
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachary T Yoneda
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics, Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Donna K Arnett
- Department of Epidemiology, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Esteban G Burchard
- Bioengineering and Therapeutic Sciences and Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yii-Der Ida Chen
- Department of Medical Genetics, Genomic Outcomes, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Myriam Fornage
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Victor R Gordeuk
- Department of Medicine, School of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lifang Hou
- Northwestern University, Chicago, IL, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ryan L Minster
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland, Baltimore, MD, USA
| | - Mehdi Nouraie
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - M Benjamin Shoemaker
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas L Smith
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veterans Affairs, Seattle, WA, USA
| | - Kent D Taylor
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marilyn J Telen
- Department of Medicine, Division of Hematology, Duke University School of Medical, Durham, NC, USA
| | - Scott T Weiss
- Department of Medicine, Channing Division of Network Medicine, Harvard Medical School, Boston, MA, USA
| | - Yingze Zhang
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nancy Heard-Costa
- Framingham Heart Study, School of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
| | - L Adrienne Cupples
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne E Justice
- Population Health Sciences, Geisinger, Danville, PA, USA.
| |
Collapse
|
23
|
Petty LE, Chen HH, Frankel EG, Zhu W, Downie CG, Graff M, Lin P, Sharma P, Zhang X, Scartozzi AC, Roshani R, Landman JM, Boehnke M, Bowden DW, Chambers JC, Mahajan A, McCarthy MI, Ng MCY, Sim X, Spracklen CN, Zhang W, Preuss M, Bottinger EP, Nadkarni GN, Loos RJF, Chen YDI, Tan J, Ipp E, Genter P, Emery LS, Louie T, Sofer T, Stilp AM, Taylor KD, Xiang AH, Buchanan TA, Roll K, Gao C, Palmer ND, Norris JM, Wagenknecht LE, Nousome D, Varma R, McKean-Cowdin R, Guo X, Hai Y, Hsueh W, Sandow K, Parra EJ, Cruz M, Valladares-Salgado A, Wacher-Rodarte N, Rotter JI, Goodarzi MO, Rich SS, Bertoni A, Raffel LJ, Nadler JL, Kandeel FR, Duggirala R, Blangero J, Lehman DM, DeFronzo RA, Thameem F, Wang Y, Gahagan S, Blanco E, Burrows R, Huerta-Chagoya A, Florez JC, Tusie-Luna T, González-Villalpando C, Orozco L, Haiman CA, Hanis CL, Rohde R, Whitsel EA, Reiner AP, Kooperberg C, Li Y, Duan Q, Lee M, Correa-Burrows P, Fried SK, North KE, McCormick JB, Fisher-Hoch SP, Gamazon ER, Morris AP, Mercader JM, Highland HM, Below JE. Large-scale multi-omics analyses in Hispanic/Latino populations identify genes for cardiometabolic traits. Nat Commun 2025; 16:3438. [PMID: 40210677 PMCID: PMC11985957 DOI: 10.1038/s41467-025-58574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 03/21/2025] [Indexed: 04/12/2025] Open
Abstract
Here, we present a multi-omics study of type 2 diabetes and quantitative blood lipid and lipoprotein traits conducted to date in Hispanic/Latino populations (nmax = 63,184). We conduct a meta-analysis of 16 type 2 diabetes and 19 lipid trait GWAS, identifying 20 genome-wide significant loci for type 2 diabetes, including one novel locus and novel signals at two known loci, based on fine-mapping. We also identify sixty-one genome-wide significant loci across the lipid/lipoprotein traits, including nine novel loci, and novel signals at 19 known loci through fine-mapping. Next, we analyze genetically regulated expression, perform Mendelian randomization, and analyze association with transcriptomic and proteomic measure using multi-omics data from a Hispanic/Latino population. Using this approach, we identify genes linked to type 2 diabetes and lipid/lipoprotein traits, including TMEM205 and NEDD9 for HDL cholesterol, TREH for triglycerides, and ANXA4 for type 2 diabetes.
Collapse
Affiliation(s)
- Lauren E Petty
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Elizabeth G Frankel
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wanying Zhu
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carolina G Downie
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Phillip Lin
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Priya Sharma
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alyssa C Scartozzi
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rashedeh Roshani
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua M Landman
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Donald W Bowden
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Anubha Mahajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Maggie C Y Ng
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology and Biostatistics, University of Massachusetts-Amherst, Amherst, MA, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Hasso-Plattner-Institut, Potsdam, Germany
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jingyi Tan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Eli Ipp
- Division of Endocrinology & Metabolism, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pauline Genter
- Division of Endocrinology & Metabolism, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leslie S Emery
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tamar Sofer
- Departments of Medicine and Biostatistics, Harvard University, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Adrienne M Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Anny H Xiang
- Department of Research & Evaluation, Division of Biostatistics Research, Kaiser Permanente of Southern California, Pasadena, CA, USA
| | - Thomas A Buchanan
- Department of Medicine, Division of Endocrine, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Kathryn Roll
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-, Salem, NC, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jill M Norris
- Department of Epidemiology, University of Colorado Denver, Aurora, CO, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Darryl Nousome
- Department of Preventative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Rohit Varma
- Southern California Eye Institute, CHA Hollywood Presbyterian Medical Center, Los Angeles, CA, USA
| | - Roberta McKean-Cowdin
- Department of Preventative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yang Hai
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Willa Hsueh
- Department of Internal Medicine, Diabetes and Metabolism Research Center, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kevin Sandow
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Esteban J Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, ON, Canada
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Adan Valladares-Salgado
- Unidad de Investigación Médica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Niels Wacher-Rodarte
- Unidad de Investigación Médica en Epidemiologia Clinica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mark O Goodarzi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Alain Bertoni
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Leslie J Raffel
- Department of Pediatrics, Division of Medical Genetics, UCI Irvine School of Medicine, Irvine, CA, USA
| | - Jerry L Nadler
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Fouad R Kandeel
- Departments of Clinical Diabetes, Endocrinology & Metabolism and Translational Research and Cellular Therapeutics, City of Hope, Duarte, CA, USA
| | | | - John Blangero
- Human Genetics and STDOI, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Donna M Lehman
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ralph A DeFronzo
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Farook Thameem
- Department of Biochemistry, Faculty of Medicine, Health Science Center, Kuwait University, Safat, Kuwait
| | - Yujie Wang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sheila Gahagan
- Division of Academic General Pediatrics, Child Development and Community Health, University of California at San Diego, San Diego, CA, USA
| | - Estela Blanco
- College y Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Raquel Burrows
- Department of Public Health Nutrition, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Alicia Huerta-Chagoya
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Tusie-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Clicerio González-Villalpando
- Centro de Estudios en Diabetes, Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Centro de Investigacion en Salud Poblacional, Instituto Nacional de Salud Pública, Mexico City, Mexico
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Craig L Hanis
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rebecca Rohde
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Yun Li
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Qing Duan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miryoung Lee
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Paulina Correa-Burrows
- Department of Public Health Nutrition, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Susan K Fried
- Diabetes, Obesity Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joseph B McCormick
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Clare Hall & MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
| | - Josep M Mercader
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
24
|
Zhou S, Zhou P, Yang T, Si J, An W, Jiang Y. Glucosamine supplementation contributes to reducing the risk of type 2 diabetes: Evidence from Mendelian randomization combined with a meta-analysis. J Int Med Res 2025; 53:3000605251334460. [PMID: 40300556 PMCID: PMC12041707 DOI: 10.1177/03000605251334460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/21/2025] [Indexed: 05/01/2025] Open
Abstract
ObjectiveObservational studies on glucosamine supplementation and type 2 diabetes risk have shown inconsistent results, necessitating the use of Mendelian randomization to clarify the true causal relationship.MethodsThe glucosamine supplementation-related genome-wide association study dataset was obtained from the MRC Integrative Epidemiology Unit consortium, whereas type 2 diabetes-related genome-wide association study datasets were obtained from the FinnGen consortium (discovery) and Xue et al.'s meta-analysis (validation). Two-sample Mendelian randomization analyses were performed separately in the discovery and validation datasets, followed by meta-analysis and multivariable Mendelian randomization analyses to verify the robustness of the results of two-sample Mendelian randomization. The estimation of the causal relationship was conducted through the inverse variance weighted method.ResultsGlucosamine supplementation exhibited a significant protective effect against type 2 diabetes, as identified by two-sample Mendelian randomization analysis in the FinnGen consortium (odds ratio: 0.13, 95% confidence interval: 0.02-0.89) and validated in Xue et al.'s meta-analysis (odds ratio: 0.06, 95%; confidence interval: 0.01-0.29). A combined meta-analysis (odds ratio: 0.08, 95%; confidence interval: 0.02-0.27) of the results of two-sample Mendelian randomization confirmed the robustness of these findings. Additionally, multivariable Mendelian randomization analysis (odds ratio: 0.12, 95%; confidence interval: 0.02-0.94), after adjusting for confounding factors, supported the results of two-sample Mendelian randomization. No evidence of heterogeneity or pleiotropy was observed.ConclusionOverall, our results revealed that genetically predicted glucosamine supplementation was inversely associated with the risk of type 2 diabetes, highlighting the potential importance of glucosamine supplementation in preventing type 2 diabetes.
Collapse
Affiliation(s)
- Shuai Zhou
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Peiwen Zhou
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Tianshi Yang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Junzhuo Si
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Wenyan An
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, China
| |
Collapse
|
25
|
Song B, Sun L, Qin X, Fei J, Yu Q, Chang X, He Y, Liu Y, Shi M, Guo D, Shen O, Zhu Z. Associations of Lipid-Lowering Drugs With Blood Pressure and Fasting Glucose: A Mendelian Randomization Study. Hypertension 2025; 82:743-751. [PMID: 39902581 DOI: 10.1161/hypertensionaha.124.23829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Observational studies have linked LDL-C (low-density lipoprotein-cholesterol)-lowering drugs with lower blood pressure (BP) and higher fasting glucose, but the causality remains unclear. We conducted a drug target Mendelian randomization study to assess the causal associations of genetically proxied inhibition of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), PCSK9 (proprotein convertase subtilisin/kexin type 9), and NPC1L1 (Niemann-Pick C1-Like 1) with BP and fasting glucose. METHODS Single-nucleotide polymorphisms in HMGCR, NPC1L1, and PCSK9 associated with LDL-C in a genome-wide association study meta-analysis from the Global Lipid Genetics Consortium (173 082 European individuals) were used to proxy LDL-C-lowering drug targets. BP and fasting glucose data were obtained from genome-wide association studies conducted by the International Consortium of Blood Pressure (757 601 European participants) and the Glucose and Insulin-related Traits Consortium (58 074 European participants). We used the inverse-variance weighted method and a series of sensitivity analyses for assessment. RESULTS Genetically proxied inhibition of HMGCR was negatively associated with systolic BP (β, -0.81 [95% CI, -1.26 to -0.37 mm Hg]; P=3.72×10-4) and diastolic BP (β, -1.58 [95% CI, -2.24 to -0.91 mm Hg]; P=3.23×10-6). Conversely, we observed a positive association between genetically proxied inhibition of HMGCR and high fasting glucose (β, 0.13 [95% CI, 0.08-0.17 mmol/L]; P=4.25×10-8). However, there was no association of PCSK9 and NPC1L1 inhibition with BP or fasting glucose. CONCLUSIONS Genetically proxied inhibition of HMGCR was significantly associated with low BP and high fasting glucose, while there was no effect of PCSK9 and NPC1L1 inhibition on BP or fasting glucose.
Collapse
Affiliation(s)
- Beiping Song
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Lulu Sun
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Xiaoli Qin
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Jiawen Fei
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Quan Yu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Xinyue Chang
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Yu He
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Yi Liu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Mengyao Shi
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (M.S., Z.Z.)
| | - Daoxia Guo
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
| | - Ouxi Shen
- Department of Occupational Health, Suzhou Industrial Park Center for Disease Control and Prevention, China (O.S.)
| | - Zhengbao Zhu
- Department of Psychiatry, Affiliated Guangji Hospital of Soochow University, School of Public Health, Suzhou Medical College of Soochow University, Jiangsu Province, China (B.S., L.S., X.Q., J.F., Q.Y., X.C., Y.H., Y.L., M.S., D.G., Z.Z.)
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (M.S., Z.Z.)
| |
Collapse
|
26
|
Zhang B, Rimbert A, Lainé A, Huijkman N, Kloosterhuis N, Smit M, van de Sluis B, Kuivenhoven JA, Tharehalli U. A study into rare GPR146 gene variants in humans and mice. Atherosclerosis 2025; 403:119137. [PMID: 40120432 DOI: 10.1016/j.atherosclerosis.2025.119137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/30/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND AND AIMS G-protein coupled receptor 146 (GPR146)-deficient mice exhibit a moderate 21 % reduction in plasma cholesterol. This is associated with decreased phosphorylation of ERK1/2 and reduced SREBP2 activity in the liver, which leads to lower VLDL secretion. Insight into the role of GPR146 in humans is however limited. We therefore set out to study rare genetic variants in GPR146 to improve our understanding of this new player in lipid metabolism. METHODS We used whole genome sequencing data from UK Biobank participants to search for rare coding variants in GPR146. We first carried out gene-based burden tests (using SAIGE-GENE-framework) and examined the association of individual variants with plasma cholesterol levels. One of the variants (P62L) was also studied using the Global Lipids Genetics Consortium (GLGC) data set and in a knock-in mouse model. RESULTS We found that the combination of rare genetic variants identified in GPR146 is significantly associated with plasma cholesterol levels. Three rare variants, i.e. P62L, I129I, and A175T were individually associated with reduced plasma cholesterol. In the GLGC cohort, the P62L variant was associated with reductions in both HDL and LDL cholesterol. Follow-up experiments show lower plasma cholesterol levels in GPR146P61L male and female mice (-13 %, p < 0.05 and -15 %, p < 0.005, respectively) when compared to controls due to a reduction in HDL cholesterol. The GPR146P61L mice did not exhibit a change in VLDL secretion. In line, the ERK1/2 signalling pathway and Srebp2 mRNA expression in liver homogenates, and the secretion of apoB by primary hepatocytes of GPR146P61L and wild-type mice were unchanged. CONCLUSIONS This study shows that rare GPR146 gene variants are associated with lower plasma cholesterol levels in humans. One of these variants, P62L is associated with reductions of HDL cholesterol and LDL cholesterol in humans while the ortholog in mice confers a loss of GPR146 function leading to only reduced HDL cholesterol. How GPR146 affects HDL metabolism in humans and mice remains to be resolved.
Collapse
Affiliation(s)
- Boyan Zhang
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Antoine Rimbert
- Nantes Université, CNRS, Inserm, Institut du Thorax, F-44000, Nantes, France
| | - Antoine Lainé
- Nantes Université, CNRS, Inserm, Institut du Thorax, F-44000, Nantes, France
| | - Nicolette Huijkman
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Niels Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marieke Smit
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Umesh Tharehalli
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
27
|
Dron JS, Natarajan P, Peloso GM. The breadth and impact of the Global Lipids Genetics Consortium. Curr Opin Lipidol 2025; 36:61-70. [PMID: 39602359 PMCID: PMC11888832 DOI: 10.1097/mol.0000000000000966] [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: 11/29/2024]
Abstract
PURPOSE OF REVIEW This review highlights contributions of the Global Lipids Genetics Consortium (GLGC) in advancing the understanding of the genetic etiology of blood lipid traits, including total cholesterol, LDL cholesterol, HDL cholesterol, triglycerides, and non-HDL cholesterol. We emphasize the consortium's collaborative efforts, discoveries related to lipid and lipoprotein biology, methodological advancements, and utilization in areas extending beyond lipid research. RECENT FINDINGS The GLGC has identified over 923 genomic loci associated with lipid traits through genome-wide association studies (GWASs), involving more than 1.65 million individuals from globally diverse populations. Many loci have been functionally validated by individuals inside and outside the GLGC community. Recent GLGC studies show increased population diversity enhances variant discovery, fine-mapping of causal loci, and polygenic score prediction for blood lipid levels. Moreover, publicly available GWAS summary statistics have facilitated the exploration of lipid-related genetic influences on cardiovascular and noncardiovascular diseases, with implications for therapeutic development and drug repurposing. SUMMARY The GLGC has significantly advanced the understanding of the genetic basis of lipid levels and serves as the leading resource of GWAS summary statistics for these traits. Continued collaboration will be critical to further understand lipid and lipoprotein biology through large-scale genetic assessments in diverse populations.
Collapse
Affiliation(s)
- Jacqueline S. Dron
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge
| | - Pradeep Natarajan
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge
- Cardiovascular Research Center, Massachusetts General Hospital
- Department of Medicine, Harvard Medical School
| | - Gina M. Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Zhang P, Wang W, Xu Q, Cui J, Zhu M, Li Y, Liu Y, Liu Y. Genetic association of circulating lipids and lipid-lowering drug targets with vascular calcification. Atherosclerosis 2025; 403:119136. [PMID: 39985880 DOI: 10.1016/j.atherosclerosis.2025.119136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/27/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND AND AIMS Vascular calcification (VC) significantly increases the incidence and mortality of many diseases. The causal relationships of dyslipidaemia and lipid-lowering drug use with VC severity remain unclear. This study explores the genetic causal associations of different circulating lipids and lipid-lowering drug targets with coronary artery calcification (CAC) and abdominal aortic artery calcification (AAC). METHODS We obtained single-nucleotide polymorphisms (SNPs) and expression quantitative trait loci (eQTLs) associated with seven circulating lipids and 13 lipid-lowering drug targets from publicly available genome-wide association studies and eQTL databases. Causal associations were investigated by univariable, multivariable, drug-target, and summary data-based Mendelian randomization (MR) analyses. Potential mediation effects of metabolic risk factors were evaluated. RESULTS MR analysis revealed that genetic proxies for low-density lipoprotein cholesterol (LDL-C), triglycerides (TC) and Lipoprotein (a) (Lp(a)) were causally associated with CAC severity, and apolipoprotein B (apoB) level was causally associated with AAC severity. A significant association was detected between hepatic Lipoprotein(A) (LPA) gene expression and CAC severity. Colocalisation analysis supported the hypothesis that the association between LPA expression and CAC quantity is driven by different causal variant sites within the ±1 Mb flanking region of LPA. Serum calcium and phosphorus had causal associations with CAC severity. CONCLUSIONS Inhibitors targeting LPA might represent CAC drug candidates. Moreover, T2DM, hypercalcemia, and hyperphosphatemia are positively causally associated with CAC severity, while chronic kidney disease and estimated glomerular filtration rate are not.
Collapse
Affiliation(s)
- Pengfei Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wenting Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Qian Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jing Cui
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Mengmeng Zhu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yiwen Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanfei Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China; The Second Department of Gerontology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yue Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
29
|
Xie Y, Bai R, Ren L, Fan H, Tuo H, Duan L, Zhou X, Fang C, Li Z, Zheng Y. Potential Causal Relationship Between Extensive Lipid Profiles and Various Hair Loss Diseases: Evidence From Univariable and Multivariable Mendelian Randomization Analyses. J Cosmet Dermatol 2025; 24:e70176. [PMID: 40208087 PMCID: PMC11984456 DOI: 10.1111/jocd.70176] [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: 01/19/2025] [Revised: 03/05/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Hair loss disorders, including non-cicatricial forms such as alopecia areata (AA) and androgenetic alopecia (AGA), as well as cicatricial forms, represent significant dermatological concerns influenced by various factors, including lipid metabolism. While observational studies and clinical trials have suggested a link between lipid levels and hair loss, the causal relationship remains unclear. METHODS We conducted a comprehensive analysis of 983 lipid variables [including triglycerides (TG), fatty acids, cholesterol, cholesterol esters, phospholipids, and lipoproteins] and 4 hair loss disorders. Two-sample univariable Mendelian randomization (UVMR) and multivariable Mendelian randomization (MVMR) analyses were employed to investigate the causal effects of lipids on hair loss disorders. Sensitivity analyses were performed to ensure the robustness of our findings. RESULTS The UVMR analysis identified 56 significant causal associations between lipid levels and hair loss disorders, with cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), TG, apolipoprotein A1, apolipoprotein B, and lipoprotein(a) emerging as key contributors. The MVMR analysis evaluated the independent effects of HDL-C, LDL-C, and TG on alopecia disorders, identifying significant associations only between HDL-C, TG, and AA. Sensitivity analyses confirmed the consistency and robustness of these results. CONCLUSION This study provides strong evidence for potential causal associations between lipids and hair loss disorders, highlighting potential therapeutic targets and the importance of lipid management in affected patients.
Collapse
Affiliation(s)
- Yuhan Xie
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Ruimin Bai
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Landong Ren
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Hengtong Fan
- Department of UrologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Huihui Tuo
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Longmei Duan
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Xiaolin Zhou
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Chengyu Fang
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Ziyan Li
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Yan Zheng
- Department of DermatologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
30
|
Xu H, Li B, Lv P, Chen Y, Lin Y, Zhang A, Zhao J, Zhou G, Wu L. Inhibition of Putative Ibrutinib Targets Promotes Atrial Fibrillation, Conduction Blocks, and Proarrhythmic Electrocardiogram Indices: A Mendelian Randomization Analysis. CANCER INNOVATION 2025; 4:e70004. [PMID: 40078362 PMCID: PMC11897533 DOI: 10.1002/cai2.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/05/2024] [Accepted: 10/16/2024] [Indexed: 03/14/2025]
Abstract
Background The mechanism by which ibrutinib, a Bruton's tyrosine kinase inhibitor, can elevate the risk of arrhythmias is not fully elucidated. In this study, we explored how inhibition of off-target kinases can contribute to this phenomenon. Methods We performed a Mendelian randomization analysis to examine the causal associations between genetically proxied inhibition of six putative ibrutinib drug targets (ErbB2/HER2, CSK, JAK3, TEC, BLK, and PLCG2) and the atrial fibrillation (AF) risk, proarrhythmic ECG indices, and cardiometabolic traits and diseases. Inverse-variance weighted random-effects models and Wald ratio were used to examine the associations between genetically proxied inhibition of these drug targets and the risk of outcomes. Colocalization analyses were employed to examine the robustness of the causally significant findings. ELISAs were used to measure ErbB2 levels in intracardiac plasma samples. Results Genetically proxied ErbB2 inhibition was associated with an increased AF risk, higher P wave terminal force, and prolonged QTc interval. Patients with AF had significantly higher intracardiac ErbB2 levels compared with patients with paroxysmal supraventricular tachycardia. CSK inhibition prolonged the QRS duration, decreased the QTc interval, and was potentially linked to conduction blocks. PLCG2 inhibition led to decreased P wave terminal force, shorter QTc interval, and increased risk of left bundle branch block. BLK inhibition shortened the QTc interval and was also associated with atrioventricular block. Conclusion The off-target effects and downstream targets of ibrutinib, including CSK, PLCG2, ERBB2, TEC, and BLK, may lead to cardiac electrical homeostasis imbalances and lethal cardiovascular diseases. Using drugs that inhibit these targets should be given extra caution.
Collapse
Affiliation(s)
- Hongxuan Xu
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Bingxun Li
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Pinchao Lv
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Ying Chen
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Yanyun Lin
- Department of CardiologyPeking University First HospitalBeijingChina
| | - An Zhang
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Jing Zhao
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Guoxiong Zhou
- Department of CardiologyPeking University First HospitalBeijingChina
| | - Lin Wu
- Department of CardiologyPeking University First HospitalBeijingChina
- State Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijingChina
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular ResearchSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
31
|
El Rouby N, Owusu‐Obeng A, Preuss MH, Lee S, Shi M, Nadukuru R, Van Driest SL, Mosley JD, DelBello M. Genome Wide Association Study (GWAS) Identifies Novel Genetic Loci for Second-Generation Antipsychotics (SGA)-Induced Metabolic Syndrome (MetS). Clin Transl Sci 2025; 18:e70216. [PMID: 40259522 PMCID: PMC12011641 DOI: 10.1111/cts.70216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Second-generation antipsychotics (SGA) are widely used for treating psychiatric disorders; however, their use is associated with an increased risk of metabolic syndrome (MetS). To identify common genetic associations of SGA-induced metabolic syndrome (SGA-MetS), we conducted a genome-wide association study (GWAS) in a diverse patient population within the BioVU and BioMe electronic health records (EHRs)-linked biobanks. Additionally, we performed Mendelian Randomization (MR) analysis to investigate the association between the individual metabolic parameters comprising MetS (body mass index [BMI], fasting glucose, blood pressure, HDL, and triglycerides) and SGA-MetS. The meta-analysis of European ancestry GWAS from BioVU and BioMe (N = 9248) identified a genome-wide signal (rs61900075, β = -0.27, SE = 0.05, p = 1.6 × 10-8) on chromosome 11. Multiple associated variants met the suggestive level of association (p ≤ 10-5) in the PELO-ITGA1 locus on chromosome 5 and were associated among the Hispanic Ancestry within BioMe. The meta-analysis of the African Ancestry patients of BioVU and BioMe (N = 2018) identified multiple genome-wide signals that were functionally mapped to NPPC-DIS3L2 in chromosome 2. Finally, the inverse-variance weighted average MR (BMI: OR = 1.2, 95% CI: 1.1-1.4, p = 0.002) showed that genetically predicted, higher BMI was associated with an increased risk of SGA-MetS. Similar results were seen in the sensitivity analyses using the weighted median and Egger MR. This study identified novel variants for SGA-MetS and suggested a role of BMI in increasing the risk of SGA-MetS. The findings highlight the value of EHR biobanks for identifying the genetics underlying SGA-MetS. The associations in chromosome 2 and 5 will need further replication.
Collapse
Affiliation(s)
- Nihal El Rouby
- James L Winkle College of PharmacyUniversity of CincinnatiCincinnatiOHUSA
- Department of Psychiatry & Behavioral NeuroscienceCollege of Medicine, University of CincinnatiCincinnatiOhioUSA
- St. Elizabeth HealthcareEdgewoodKentuckyUSA
| | - Aniwaa Owusu‐Obeng
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Michael H. Preuss
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Simon Lee
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Mingjian Shi
- Department of Biomedical InformaticsVUMCNashvilleTennesseeUSA
| | - Rajiv Nadukuru
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Sara L. Van Driest
- Department of PediatricsVanderbilt University Medical Center (VUMC)NashvilleTennesseeUSA
- All of Us Research ProgramNational Institutes of HealthBethesdaMarylandUSA
| | - Jonathan D. Mosley
- Department of Biomedical InformaticsVUMCNashvilleTennesseeUSA
- Department of MedicineVUMCNashvilleTennesseeUSA
| | - Melissa DelBello
- Department of Psychiatry & Behavioral NeuroscienceCollege of Medicine, University of CincinnatiCincinnatiOhioUSA
- College of MedicineUniversity of CincinnatiCincinnatiOHUSA
| |
Collapse
|
32
|
Sun Z, Zhang J, Li Y, Tuo M, Yu L, Wang Y, Sun Y. Statins' protective effects on focal epilepsy are independent of LDL-C. Epilepsia Open 2025; 10:521-528. [PMID: 40053059 PMCID: PMC12014916 DOI: 10.1002/epi4.70008] [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: 07/25/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 04/24/2025] Open
Abstract
OBJECTIVE This study evaluates the potential protective effects of statins against epilepsy, focusing on their differential impacts on focal and generalized epilepsy. It investigates the role of statins through the HMGCR gene and associated low-density lipoprotein (LDL) cholesterol levels. METHODS A two-sample Mendelian randomization (MR) and summary-data-based MR (SMR) approach were employed using genetic instruments from genome-wide association studies (GWASs) and expression quantitative trait loci (eQTLs). Subgroup analyses examined focal and generalized epilepsy, with sensitivity tests, including MR-Egger regression and MR-PRESSO, to assess horizontal pleiotropy and robustness. RESULTS SMR analysis found no significant association between HMGCR expression and epilepsy risk across subtypes (p > 0.05). However, inverse-variance-weighted MR (IVW-MR) showed that elevated LDL cholesterol mediated by HMGCR was linked to an increased risk of focal epilepsy (OR = 1.251, 95% CI = 1.135-1.378). No such association was observed for generalized epilepsy. Statins showed promise in reducing post-stroke epilepsy risk, likely through anti-inflammatory and neuroprotective effects. SIGNIFICANCE The findings suggest that statins' protective effects may be subtype-specific, particularly in post-stroke focal epilepsy. Further research is needed to elucidate underlying mechanisms and optimize their therapeutic potential in epilepsy management. PLAIN LANGUAGE SUMMARY Statins, drugs typically used to manage cholesterol, may also lower the risk of developing certain types of epilepsy, especially post-stroke focal epilepsy, by reducing inflammation and protecting brain cells. The research found no clear effect of statins on generalized epilepsy or epilepsy caused by other factors. These results could aid in creating better treatments for epilepsy in the future.
Collapse
Affiliation(s)
- Zhen Sun
- Department of NeurologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Jing Zhang
- Department of Endocrinology and MetabolismThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yulong Li
- Department of NeurologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Miao Tuo
- Department of NursingThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Limin Yu
- Department of NeurologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yun Wang
- Department of NeurologyZhucheng Integrated Traditional Chinese and Western Medicine HospitalWeifangChina
| | - Yanping Sun
- Department of NeurologyThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
33
|
Ottensmann L, Tabassum R, Ruotsalainen SE, Gerl MJ, Klose C, McCartney DL, Widén E, Simons K, Ripatti S, Vitart V, Hayward C, Pirinen M. Examining the link between 179 lipid species and 7 diseases using genetic predictors. EBioMedicine 2025; 114:105671. [PMID: 40157129 PMCID: PMC11995710 DOI: 10.1016/j.ebiom.2025.105671] [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: 12/05/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Genome-wide association studies of lipid species have identified several loci shared with various diseases, however, the relationship between lipid species and disease risk remains poorly understood. Here we investigated whether the plasma levels of lipid species are causally linked to disease risk. METHODS We built genetic predictors of 179 lipid species, measured in 7174 Finnish individuals, by utilising either 11 high-impact genomic loci or genome-wide polygenic scores (PGS). We assessed the impact of the lipid species on seven diseases by performing disease association across FinnGen (n = 500,348), UK Biobank (n = 420,531), and Generation Scotland (n = 20,032). We performed univariable Mendelian randomisation (MR) and multivariable MR (MVMR) analyses to examine whether lipid species impact disease risk independently of standard lipids. FINDINGS PGS explained >4% of the variance for 34 lipid species but variants outside the high-impact loci had only a marginal contribution. Variants within the high-impact loci showed association with all seven diseases. MVMR supported a causal role of ApoB in ischaemic heart disease after accounting for lipid species. Phosphatidylethanolamine-increasing LIPC variants seemed to lower age-related macular degeneration risk independently of HDL-cholesterol. MVMR suggested a protective effect of four lipid species containing arachidonic acid on cholelithiasis risk independently of Total Cholesterol. INTERPRETATION Our study demonstrates how genetic predictors of lipid species can be utilised to gain insights into disease risk. We report potential links between lipid species and age-related macular degeneration and cholelithiasis risk, which can be explored for their utility in disease risk prediction and therapy. FUNDING The funders had no role in the study design, data analyses, interpretation, or writing of this article.
Collapse
Affiliation(s)
- Linda Ottensmann
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland; Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom.
| | - Rubina Tabassum
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanni E Ruotsalainen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Samuli Ripatti
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland; Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Matti Pirinen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland; Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
34
|
Wei L, Ding E, Lu D, Rui Z, Shen J, Fan G. Assessing the effect of modifiable risk factors on hepatocellular carcinoma: evidence from a bidirectional Mendelian randomization analysis. Discov Oncol 2025; 16:437. [PMID: 40164825 PMCID: PMC11958933 DOI: 10.1007/s12672-025-02177-z] [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: 12/12/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The pathogenesis of hepatocellular carcinoma (HCC) involves a variety of environmental risk factors, some of which have yet to be fully clarified. Using the Mendelian randomization (MR) approach, this study comprehensively investigates the causal effect of genetically predicted modifiable risk factors on HCC. METHODS Genetic variants related to the 50 risk factors that had been identified in previous research were derived from genome-wide association studies. Summary statistics for the discovery cohort and validation cohort of HCC were sourced from the FinnGen consortium and the UK Biobank, respectively. Bidirectional MR analysis and sensitivity analysis were performed to establish causative risk factors for HCC. RESULTS Through the inverse variance weighted method, the results of the discovery cohort indicated that waist circumference, nonalcoholic fatty liver disease (NAFLD), alanine aminotransferase (ALT) levels, and aspartate aminotransferase (AST) levels were significantly linked to HCC occurrence risk. Furthermore, body fat percentage, glycated hemoglobin (HbA1c), obesity class 1-3, waist-to-hip ratio, iron, ferritin, transferrin saturation, and urate had suggestive associations with HCC. The validation cohort further confirmed that NAFLD and ALT levels were strongly related to HCC. Reverse MR indicated that genetic susceptibility to HCC was connected to NAFLD and transferrin saturation. Sensitivity analyses showed that most of the findings were robust. CONCLUSION This MR study delivers evidence of the complex causal relationship between modifiable risk factors and HCC. These findings offer new insights into potential prevention and treatment strategies for HCC.
Collapse
Affiliation(s)
- Lijuan Wei
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Enci Ding
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Dongyan Lu
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Zhongying Rui
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Jie Shen
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Guoju Fan
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, No. 23, Pingjiang Road, Hexi District, Tianjin, 300211, China.
| |
Collapse
|
35
|
Grapentine S, Agarwal P, Dolinsky VW, Bakovic M. Epigenome-wide methylation analysis shows phosphonoethylamine alleviates aberrant DNA methylation in NASH caused by Pcyt2 deficiency. PLoS One 2025; 20:e0320510. [PMID: 40153413 PMCID: PMC11952270 DOI: 10.1371/journal.pone.0320510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/19/2025] [Indexed: 03/30/2025] Open
Abstract
BACKGROUND Aberrant DNA methylation can lead to the onset of pathological phenotypes and is increasingly being implicated in age-related metabolic diseases. In our preceding study we show that the heterozygous ablation of Pcyt2, the rate limiting enzyme in phosphatidylethanolamine (PE) synthesis, causes an age-dependent development of non-alcoholic steatohepatitis (NASH), and that treatment with the Pcyt2 substrate phosphonoethylamine (PEA) can attenuate phenotypic NASH pathologies. Here, we hypothesize that abnormal DNA methylation patterns underly the development of Pcyt2 + /- NASH. In this study, we conduct an epigenome-wide methylation analysis to characterize the differential methylation of Pcyt2 + /- livers and investigate whether the attenuation of NASH with PEA treatment is associated with changes in DNA methylation. RESULTS Pcyt2 + /- NASH liver experiences significant alterations in DNA methylation pattens relative to Pcyt2 + / + . Differentially methylated genes belong to pathways including PI3K-Akt signalling pathway, Foxo signalling pathway, oxidative phosphorylation and insulin signalling/secretion, indicating that epigenetic regulation underlies many of our previously established functional pathological mechanisms of Pcyt2 + /- NASH. Previously unidentified pathways during Pcyt2 deficiency are highlighted, such as cell cycle regulation and cellular senescence that may contribute to NASH development. Treatment with PEA dramatically attenuates aberrant total and protein-coding DNA methylation patterns by 96%. PEA treatment restored the methylation status of key genes involved in epigenetic modifications and induced differential methylation of genes associated with obesity and T2DM such as Adyc3, Celsr2, Fam63b. CONCLUSION The Pcyt2 + /- liver methylome and transcriptome is altered and likely underlies much of the pathology in Pcyt2 + /- NASH phenotype. The treatment with PEA significantly attenuates aberrant DNA methylation in Pcyt2 + /- liver and corrects the DNA methylation of genes involved in the pathogenesis of NASH, indicating its therapeutic potential. This analysis provides critical insight into the epigenetic basis of NASH pathophysiology and suggests diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Sophie Grapentine
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - Prasoon Agarwal
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada
| | - Vernon W. Dolinsky
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| |
Collapse
|
36
|
Ma X, Ding L, Li S, Fan Y, Wang X, Han Y, Yuan H, Sun L, He Q, Liu M. Druggable genome-wide Mendelian randomization identifies therapeutic targets for metabolic dysfunction-associated steatotic liver disease. Lipids Health Dis 2025; 24:113. [PMID: 40140823 PMCID: PMC11938603 DOI: 10.1186/s12944-025-02515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) affects > 25% of the global population, potentially leading to severe hepatic and extrahepatic complications, including metabolic dysfunction-associated steatohepatitis. Given that the pathophysiology of MASLD is incompletely understood, identifying therapeutic targets and optimizing treatment strategies are crucial for addressing this severe condition. METHODS Mendelian randomization (MR) analysis was conducted using two genome-wide association study datasets: a European meta-analysis (8,434 cases; 770,180 controls) and an additional study (3,954 cases; 355,942 controls), identifying therapeutic targets for MASLD. Of 4302 drug-target genes, 2,664 genetic instrument variables were derived from cis-expression quantitative trait loci (cis-eQTLs). Colocalization analyses assessed shared causal variants between MASLD-associated single nucleotide polymorphisms and eQTLs. Using the drug target gene cis-eQTL of liver tissue from the genotype-tissue expression project, we performed MR and summary MR to validate the significance of the gene results of the blood eQTL MR. RNA-sequencing data from liver biopsies were validated using immunohistochemistry and quantitative polymerase chain reaction (qPCR) tests to confirm gene expression findings. RESULT MR analysis across both datasets identified significant MR associations between MASLD and two drug targets-milk fat globule-EGF factor 8 (MFGE8) (odds ratio [OR] 0.89, 95% confidence interval [CI] 0.85-0.94; P = 2.15 × 10-6) and cluster of differentiation 33 (CD33) (OR 1.17, 95% CI 1.10-1.25; P = 1.39 × 10-6). Both targets exhibited strong colocalization with MASLD. Genetic manipulation indicating MFGE8 activation and CD33 inhibition did not increase the risk for other metabolic disorders. RNA-sequencing, qPCR, and immunohistochemistry validation demonstrated consistent differential expressions of MFGE8 and CD33 in MASLD. CONCLUSION CD33 inhibition can reduce MASLD risk, while MFGE8 activation may offer therapeutic benefits for MASLD treatment.
Collapse
Affiliation(s)
- Xiaohui Ma
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Endocrinology and Metabolism, Baotou Central Hospital, Baotou, China
| | - Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shuo Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yu Fan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xin Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yitong Han
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hengjie Yuan
- Department of Pharmacy, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
37
|
Tang B, Lin N, Liang J, Yi G, Zhang L, Peng W, Xue C, Jiang H, Li M. Leveraging pleiotropic clustering to address high proportion correlated horizontal pleiotropy in Mendelian randomization studies. Nat Commun 2025; 16:2817. [PMID: 40118820 PMCID: PMC11928562 DOI: 10.1038/s41467-025-57912-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 03/05/2025] [Indexed: 03/24/2025] Open
Abstract
Mendelian randomization harnesses genetic variants as instrumental variables to infer causal relationships between exposures and outcomes. However, certain genetic variants can affect both the exposure and the outcome through a shared factor. This phenomenon, called correlated horizontal pleiotropy, may result in false-positive causal findings. Here, we propose a Pleiotropic Clustering framework for Mendelian randomization, PCMR. PCMR detects correlated horizontal pleiotropy and extends the zero modal pleiotropy assumption to enhance causal inference in trait pairs with correlated horizontal pleiotropic variants. Simulations show that PCMR can effectively detect correlated horizontal pleiotropy and avoid false positives in the presence of correlated horizontal pleiotropic variants, even when they constitute a high proportion of the variants connecting both traits (e.g., 30-40%). In datasets consisting of 48 exposure-common disease pairs, PCMR detects horizontal correlated pleiotropy in 7 out of the exposure-common disease pairs, and avoids detecting false positive causal links. Additionally, PCMR can facilitate the integration of biological information to exclude correlated horizontal pleiotropic variants, enhancing causal inference. We apply PCMR to study causal relationships between three common psychiatric disorders as examples.
Collapse
Affiliation(s)
- Bin Tang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Nan Lin
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Junhao Liang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Guorong Yi
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Liubin Zhang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Wenjie Peng
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Chao Xue
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Hui Jiang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Miaoxin Li
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| |
Collapse
|
38
|
Spagnuolo CM, Wang J, McIntyre AD, Kennedy BA, Hegele RA. Comparison of Patients With Familial Chylomicronemia Syndrome and Multifactorial Chylomicronemia Syndrome. J Clin Endocrinol Metab 2025; 110:1158-1165. [PMID: 39238074 PMCID: PMC11913094 DOI: 10.1210/clinem/dgae613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/07/2024]
Abstract
CONTEXT Patients with rare familial chylomicronemia syndrome (FCS) and relatively common multifactorial chylomicronemia syndrome (MCS) both express severe hypertriglyceridemia, defined as plasma triglyceride concentration ≥10 mmol/L (≥885 mg/dL). Clinically there can be confusion between the 2 conditions. OBJECTIVE To compare clinical and biochemical phenotypes in patients with genotypically characterized FCS and MCS. METHODS We performed targeted sequencing of DNA from 193 patients with severe hypertriglyceridemia, classified them as having either FCS or MCS, and compared clinical and biochemical characteristics. RESULTS Patients with FCS were significantly younger than patients with MCS (31.4 ± 16.7 vs 51.0 ± 11.3 years; P = .003), with earlier age at symptom onset (15.0 ± 15.8 vs 37.8 ± 8.8 years; P = .00066), lower body mass index (23.3 ± 3.1 vs 30.7 ± 5.0 kg/m2; P = .000016), and higher prevalence of pancreatitis events (81.8% vs 35.2%; P = .003). Furthermore, patients with FCS had a higher ratio of triglyceride to total cholesterol (ie, 4.18 ± 0.92 vs 1.08 ± 0.51; P < .0001) and lower plasma apolipoprotein B (ie, 0.56 ± 0.15 vs 1.02 ± 0.43 g/L; P < .0001) than patients with MCS. Patients with MCS with heterozygous pathogenic variants had a relatively more severe clinical presentation than other MCS genetic subgroups. CONCLUSION Patients with FCS have notable phenotypic differences from patients with MCS, although there is overlap. While genetic analysis of patients with persistent severe hypertriglyceridemia can definitively diagnose FCS, 8.8% of patients with MCS with sustained refractory hypertriglyceridemia behave functionally as if they have FCS, which should influence their eligibility for novel therapies for severe hypertriglyceridemia.
Collapse
Affiliation(s)
- Catherine M Spagnuolo
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
| | - Jian Wang
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
| | - Adam D McIntyre
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
| | - Brooke A Kennedy
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
| | - Robert A Hegele
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
| |
Collapse
|
39
|
Wang T, Yao Y, Gao X, Luan H, Wang X, Liu L, Sun C. Genetic association of lipids and lipid-lowering drug target genes with breast cancer. Discov Oncol 2025; 16:331. [PMID: 40095250 PMCID: PMC11914663 DOI: 10.1007/s12672-025-02041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Although several preclinical and epidemiological studies have shown that blood lipids and lipid-lowering drugs can reduce the risk of breast cancer, this finding remains controversial. This study aimed to explore the causal relationship between dyslipidemia,lipid-lowering drugs, and breast cancer. We also aimed to evaluate the potential impact of lipid-lowering drug targets on breast cancer. METHOD Data of 431 lipid- and lipid-related phenotypes were obtained from genome-wide association study (GWAS), and mendelian randomization (MR) analyses were performed using two independent breast cancer datasets as endpoints. Genetic variants associated with genes encoding lipid-lowering drug targets were extracted from the Global Lipid Genetics Consortium. Expression quantitative trait loci data in relevant tissues were used to further validate lipid-lowering drug targets that reached significance and combined with bioinformatics approaches for molecular expression and prognostic exploration. Further mediation analyses were performed to explore potential mediators. RESULT In two independent datasets, phosphatidylcholine (18:1_0:0 levels) was associated with breast cancer risk (discovery: odds ratio (OR) = 1.255 [95% confidence interval (CI) 1.120-1.406]; p = 8.936 × 10-5, replication: OR = 1.016 [95% CI, 1.003-1.030]; p = 0.017), HMG- CoA reductase (HMGCR) inhibition was genetically modeled and associated with a reduced risk of breast cancer (discovery: OR = 0.833 [95% CI 0.752-0.923], p = 5.12 × 10-4; replication: OR = 0.975 [95% CI 0.960-0.990], p = 1.65 × 10-3). There was a significant MR correlation between HMGCR expression in whole blood and breast cancer (OR = 1.11 [95% 1.01-1.22] p = 0.04). Bioinformatics analysis revealed that HMGCR expression higher in breast cancer tissues than in normal tissues, along with poor overall survival and relapse-free survival, and was associated with multiple immune cell infiltration. Finally, the mediation analysis showed that HMGCR inhibitors affected breast cancer through different immune cell phenotypes and C-reactive protein levels. CONCLUSION In this study, we found for the first time that phosphatidylcholine (18:1_0:0) levels are associated with breast cancer risk. We found that HMGCR inhibitors are associated with a reduced risk of breast cancer, and part of their action may be through pathways other than lipid-lowering, including modulation of immune function and reduction of inflammation represented by C-reactive protein levels.
Collapse
Affiliation(s)
- Tianhua Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Xinhai Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Luan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue Wang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China
| | - Lijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
40
|
Shan H, Fan S, Li Q, Liang R, Chen Z, Wang S, Wang X, Li Y, Chen S, Yu K, Fei T. Systematic interrogation of functional genes underlying cholesterol and lipid homeostasis. Genome Biol 2025; 26:59. [PMID: 40098013 PMCID: PMC11912599 DOI: 10.1186/s13059-025-03531-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Dyslipidemia or hypercholesterolemia are among the main risk factors for cardiovascular diseases. Unraveling the molecular basis of lipid or cholesterol homeostasis would help to identify novel drug targets and develop effective therapeutics. RESULTS Here, we adopt a systematic approach to catalog the genes underlying lipid and cholesterol homeostasis by combinatorial use of high-throughput CRISPR screening, RNA sequencing, human genetic variant association analysis, and proteomic and metabolomic profiling. Such integrative multi-omics efforts identify gamma-glutamyltransferase GGT7 as an intriguing potential cholesterol and lipid regulator. As a SREBP2-dependent target, GGT7 positively regulates cellular cholesterol levels and affects the expression of several cholesterol metabolism genes. Furthermore, GGT7 interacts with actin-dependent motor protein MYH10 to control low-density lipoprotein cholesterol (LDL-C) uptake into the cells. Genetic ablation of Ggt7 in mice leads to reduced serum cholesterol levels, supporting an in vivo role of Ggt7 during cholesterol homeostasis. CONCLUSIONS Our study not only provides a repertoire of lipid or cholesterol regulatory genes from multiple angles but also reveals a causal link between a gamma-glutamyltransferase and cholesterol metabolism.
Collapse
Affiliation(s)
- Haihuan Shan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shuangshuang Fan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Quanrun Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Ruipu Liang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Zhisong Chen
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shengnan Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Xiaofeng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Yurong Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shuai Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Kun Yu
- College of Medicine and Bioinformation Engineering, Northeastern University, Shenyang, 110819, China
| | - Teng Fei
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China.
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China.
| |
Collapse
|
41
|
He Y, Wei Y, Liang H, Wan Y, Zhang Y, Zhang J. Causal association between metabolic syndrome and ovarian dysfunction: a bidirectional two-sample mendelian randomization. J Ovarian Res 2025; 18:50. [PMID: 40069881 PMCID: PMC11895234 DOI: 10.1186/s13048-025-01614-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/31/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The relationship between Metabolic Syndrome (MetS) and ovarian dysfunction has been widely reported in observational studies, yet it remains not fully understood. This study employs genetic prediction methods and utilizes summary data from genome-wide association studies (GWAS) to investigate this causal link. METHODS We employed a bidirectional two-sample Mendelian Randomization (MR) analysis utilizing MetS and ovarian dysfunction summary data from GWAS. Inverse variance weighted (IVW) was employed as the primary MR method, supplemented by Weighted Median, Weighted Mode, and MR-Egger methods. The robustness of the results was further assessed through sensitivity analyses including MR-Egger regression, MR-PRESSO, Cochran's Q, and leave-one-out test. RESULTS Our MR analysis identified a causal relationship between genetically determined insulin resistance (OR = 0.26, 95% CI: 0.08-0.89, P = 0.03), waist circumference (OR = 2.14, 95% CI: 1.45-3.15, P < 0.001), BMI (OR = 2.1, 95% CI: 1.56-2.83, P < 0.001) and ovarian dysfunction. Conversely, reverse MR analysis confirmed causal effects of ovarian dysfunction on metabolic syndrome (OR = 0.98, 95% CI: 0.97-0.99, P < 0.001) and waist circumference (OR = 0.99, 95% CI: 0.98-0.99, P = 0.02). The results of MR-Egger regression test indicated that the whole analysis was not affected by horizontal pleiotropy. Additionally, the MR-PRESSO test identified outliers in SNPs, but after removal of outliers, results remained unchanged. CONCLUSION This study reveals a bidirectional causal connection between metabolic syndrome and ovarian dysfunction via genetic prediction methods. These findings are crucial for advancing our understanding of the interactions between these conditions and developing strategies for prevention and treatment.
Collapse
Affiliation(s)
- Ying He
- Department of Obstetrics and Gynecology, Xijing 986 Hospital Department, Air force Medical University, No. 6 Jianshe West Road, Xi'an, 710054, Shaanxi, China
| | - Yanling Wei
- Department of Obstetrics and Gynecology, Xijing Hospital, Air force Medical University, No. 15 Changle West Road, Xi'an, 710033, Shaanxi, China
| | - Haixia Liang
- Department of Obstetrics and Gynecology, Xijing 986 Hospital Department, Air force Medical University, No. 6 Jianshe West Road, Xi'an, 710054, Shaanxi, China
| | - Yi Wan
- Department of Health Service, Air force Medical University, Xi'an, 710032, Shaanxi, China
| | - Ying Zhang
- Department of Obstetrics and Gynecology, Xijing 986 Hospital Department, Air force Medical University, No. 6 Jianshe West Road, Xi'an, 710054, Shaanxi, China.
| | - Jianfang Zhang
- Department of Obstetrics and Gynecology, Xijing Hospital, Air force Medical University, No. 15 Changle West Road, Xi'an, 710033, Shaanxi, China.
| |
Collapse
|
42
|
Xin J, Xu Z, Zhang F, Sun Y, Wang X, Wu C, Zhao L. Mendelian randomization-based observational cohort study on drug targets: Impact of antihypertensive and lipid-lowering therapies on inflammatory cytokines. Medicine (Baltimore) 2025; 104:e41771. [PMID: 40068040 PMCID: PMC11903001 DOI: 10.1097/md.0000000000041771] [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: 01/17/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
This study assesses the causal effects of antihypertensive and lipid-lowering drugs on inflammatory cytokines using a Mendelian randomization (MR) approach. We conducted a drug-targeted MR analysis using data from large-scale genome-wide association studies and eQTL datasets. SNPs near drug target genes served as instrumental variables to investigate the impact of antihypertensive (angiotensin-converting enzyme inhibitors [ACEIs], ARBs) and lipid-lowering drugs (HMGCR inhibitors, proprotein convertase subtilisin/Kexin type 9 [PCSK9] inhibitors, Niemann-Pick C1-like 1 inhibitors) on inflammatory cytokines. Sensitivity analyses, including leave-one-out and MR-Egger tests, were performed to confirm the robustness of the findings. ACEIs were associated with decreased levels of IL-1β, TNF-α, and CRP. ARBs did not show significant effects on inflammatory cytokines. HMGCR inhibitors significantly reduced MCP-1, MIP-1β, TNF-α, and IFN-γ, while PCSK9 inhibitors were linked to reductions in IL-1β and IL-6. Sensitivity analyses supported the reliability of these findings. The study demonstrated distinct anti-inflammatory effects of ACEIs, HMGCR inhibitors, and PCSK9 inhibitors. These findings support the potential use of these drugs to mitigate inflammation-related complications in patients with chronic conditions.
Collapse
Affiliation(s)
- Jiechen Xin
- Department of Critical Care Medicine, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Zhibin Xu
- Department of Organ Transplantation, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feng Zhang
- Department of Critical Care Medicine, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Yue Sun
- Department of Critical Care Medicine, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Xiaoyan Wang
- Department of Critical Care Medicine, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Chaojun Wu
- Department of Critical Care Medicine, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Longshu Zhao
- Department of Critical Care Medicine, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| |
Collapse
|
43
|
Vargas-Alarcón G, Posadas-Sánchez R, Pérez-Méndez O, Manuel Fragoso J. Cholesterol 7 alpha-hydroxylase ( CYP7A1) gene polymorphisms are associated with increased LDL-cholesterol levels and the incidence of subclinical atherosclerosis. BIOMOLECULES & BIOMEDICINE 2025; 25:822-832. [PMID: 39207177 PMCID: PMC11959385 DOI: 10.17305/bb.2024.10764] [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: 05/21/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
The cholesterol 7 alpha-hydroxylase (CYP7A1) enzyme plays an important role in the conversion of cholesterol to bile acid, contributing to the reduction of cholesterol plasma levels in normal conditions. Nonetheless, recent studies have shown that some genetic variants in the enhancer and promoter regions of the CYP7A1 gene reduce the expression of the CYP7A1 enzyme, increasing plasma lipid levels, as well as the risk of developing coronary heart disease. The aim of this work was to explore whether the genetic variants (rs2081687, rs9297994, rs10107182, rs10504255, rs1457043, rs8192870, and rs3808607) of the CYP7A1 gene are involved in subclinical atherosclerosis and plasma lipid levels. We included 416 patients with subclinical atherosclerosis (SA) with coronary artery calcium (CAC) greater than zero, and 1046 controls with CAC = 0. According to the inheritance models (co-dominant, dominant, recessive, over-dominant and additive), the homozygosity of the minor allele frequencies of 7 analyzed polymorphisms showed a high incidence of SA (P < 0.05). In a sub-analysis performed including only the patients with SA, the same SNPs were associated with increased low-density lipoprotein cholesterol (LDL-C) levels. On the other hand, our findings showed that the haplotype (TGCGCTG) increases the risk of developing SA (P < 0.05). In conclusion, the rs2081687, rs9297994, rs10107182, rs10504255, rs1457043, rs8192870, and rs3808607 polymorphisms of CYP7A1 confer a risk of developing SA and elevated LDL-C levels. Our results suggest that the CYP7A1 is involved in the incidence of SA through the increase in the plasma lipid profile.
Collapse
Affiliation(s)
- Gilberto Vargas-Alarcón
- Direccion de Investigación, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, México
| | - Rosalinda Posadas-Sánchez
- Departmento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, México
| | - Oscar Pérez-Méndez
- Departmento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, México
| | - José Manuel Fragoso
- Departmento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, México
| |
Collapse
|
44
|
Fu S, Li Q, Cheng L, Wan S, Wang Q, Min Y, Xie Y, Liu H, Hu T, Liu H, Chen W, Zhang Y, Xiong F. Causal Relationship Between Intelligence, Noncognitive Education, Cognition and Urinary Tract or Kidney Infection: A Mendelian Randomization Study. Int J Nephrol Renovasc Dis 2025; 18:71-85. [PMID: 40070673 PMCID: PMC11895678 DOI: 10.2147/ijnrd.s511736] [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: 12/12/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Background The occurrence of urinary tract or kidney infection is correlated with intelligence, noncognitive education and cognition, but the causal relationship between them remains uncertain, and which risk factors mediate this causal relationship remains unknown. Methods The intelligence (n=269,867), noncognitive education (n=510,795) and cognition data (n=257,700) were obtained from genome-wide association studies (GWAS) conducted in individuals of European ethnicities. The genetic associations between these factors and urinary tract or kidney infection (UK Biobank, n=397,867) were analyzed using linkage disequilibrium score regression. We employed a two-sample univariate and multivariate Mendelian randomization to evaluate the causal relationship, and utilized a two-step Mendelian randomization to examine the involvement of 28 potential mediators and their respective mediating proportions. Results The genetic correlation coefficients of intelligence, noncognitive education, cognition, and urinary tract or kidney infection were -0.338, -0.218, and -0.330. The Mendelian randomization using the inverse variance weighted method revealed each 1-SD increase in intelligence, the risk of infection decreased by 15.9%, while after adjusting for noncognitive education, the risk decreased by 20%. For each 1-SD increase in noncognitive education, the risk of infection decreased by 8%, which further reduced to 7.1% after adjusting for intelligence and to 6.7% after adjusting for cognition. For each 1-SD increase in cognition, the risk of infection decreased by 10.8%, increasing to 11.9% after adjusting for noncognitive education. The effects of intelligence and cognition are interdependent. 2 out of 28 potential mediating factors exhibited significant mediation effects in the causal relationship between noncognitive education and urinary tract or kidney infection, with body mass index accounting for 12.1% of the mediation effect and smoking initiation accounting for 14.7%. Conclusion Enhancing intelligence, noncognitive education, and cognition can mitigate the susceptibility to urinary tract or kidney infection. Noncognitive education exhibited independent effect, while body mass index and smoking initiation assuming a mediating role.
Collapse
Affiliation(s)
- Shuai Fu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Qiang Li
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Li Cheng
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Sheng Wan
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Quan Wang
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yonglong Min
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yanghao Xie
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Huizhen Liu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Taotao Hu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Hong Liu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Weidong Chen
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yanmin Zhang
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Fei Xiong
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
45
|
Kim S, Yoo HY. Sex differences in predicting dyslipidemia using polygenic risk score with fatty liver index and fibrotic nonalcoholic steatohepatitis index. Sci Rep 2025; 15:7849. [PMID: 40050666 PMCID: PMC11885555 DOI: 10.1038/s41598-025-92766-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 03/09/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are recognized risk factors for dyslipidemia. Current prediction models that rely solely on dyslipidemia polygenic risk score (PRS) have certain limitations. We aimed to validate simple indexes for NAFLD and NASH as predictors of dyslipidemia using the PRS. This study utilized cohort data from an urban population-based dataset comprising 48,263 South Koreans. The incidence of dyslipidemia was higher in men than in women (32.4% and 27.8%; p < 0.001). The PRS model predicted dyslipidemia more accurately in men (AUROC [95% confidence intervals]: 0.645 [0.636-0.754]). Notably, integrating the fatty liver index (FLI) and fibrotic NASH index (FNI) with the PRS model resulted in the highest accuracy in diagnosing dyslipidemia, particularly in men (AUROC [95% confidence intervals]: 0.704 [0.698-0.711]). In conclusion, a predictive model combining the PRS with FLI and FNI was validated. This model offers more accurate predictive value for diagnosing dyslipidemia, particularly in East Asian men. Thus, our study has the clinical potential for identifying high-risk individuals and determining preventive measures for dyslipidemia in a sex-specific manner.
Collapse
Affiliation(s)
- Sei Kim
- Department of Nursing, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hae Young Yoo
- Department of Nursing, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
46
|
Ponikowska M, Di Domenico P, Bolli A, Busby GB, Perez E, Bottà G. Precision Medicine in Cardiovascular Disease Prevention: Clinical Validation of Multi-Ancestry Polygenic Risk Scores in a U.S. Cohort. Nutrients 2025; 17:926. [PMID: 40077796 PMCID: PMC11901995 DOI: 10.3390/nu17050926] [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: 01/29/2025] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Polygenic risk score (PRS) quantifies the cumulative effects of common genetic variants across the genome, including both coding and non-coding regions, to predict the risk of developing common diseases. In cardiovascular medicine, PRS enhances risk stratification beyond traditional clinical risk factors, offering a precision medicine approach to coronary artery disease (CAD) prevention. This study evaluates the predictive performance of a multi-ancestry PRS framework for cardiovascular risk assessment using the All of Us (AoU) short-read whole-genome sequencing dataset comprising over 225,000 participants. METHODS We developed PRSs for lipid traits (LDL-C, HDL-C, triglycerides) and cardiometabolic conditions (type 2 diabetes, hypertension, atrial fibrillation) and constructed two metaPRSs: one integrating lipid and cardiometabolic PRSs (risk factor metaPRS) and another incorporating CAD PRSs in addition to these risk factors (risk factor + CAD metaPRS). Predictive performance was evaluated separately for each trait-specific PRS and for both metaPRSs to assess their effectiveness in CAD risk prediction across diverse ancestries. Model predictive performance, including calibration, was assessed separately for each ancestry group, ensuring that all metrics were ancestry-specific and that PRSs remain generalizable across diverse populations Results: PRSs for lipids and cardiometabolic conditions demonstrated strong predictive performance across ancestries. The risk factors metaPRS predicted CAD risk across multiple ancestries. The addition of a CAD-specific PRS to the risk factors metaPRS improved predictive performance, highlighting a genetic component in CAD etiopathology that is not fully captured by traditional risk factors, whether clinically measured or genetically inferred. Model calibration and validation across ancestries confirmed the broad applicability of PRS-based approaches in multi-ethnic populations. CONCLUSION PRS-based risk stratification provides a reliable, ancestry-inclusive framework for personalized cardiovascular disease prevention, enabling better targeted interventions such as pharmacological therapy and lifestyle modifications. By incorporating genetic information from both coding and non-coding regions, PRSs refine risk prediction across diverse populations, advancing the integration of genomics into precision medicine for common diseases.
Collapse
Affiliation(s)
- Małgorzata Ponikowska
- Allelica Inc., San Francisco, CA 94105, USA; (M.P.); (A.B.)
- Department of Biology and Medical Genetics, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| | | | | | | | - Emma Perez
- Allelica Inc., San Francisco, CA 94105, USA; (M.P.); (A.B.)
- Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Giordano Bottà
- Allelica Inc., San Francisco, CA 94105, USA; (M.P.); (A.B.)
| |
Collapse
|
47
|
Yang Q, Li M, Chen P, Dou N, Liu M, Lu P, Yu C. Systematic Evaluation of the Impact of a Wide Range of Dietary Habits on Myocardial Infarction: A Two-Sample Mendelian Randomization Analysis. J Am Heart Assoc 2025; 14:e035936. [PMID: 40008582 DOI: 10.1161/jaha.124.035936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/19/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Myocardial infarction is a cardiovascular disease that significantly contributes to global morbidity and disability. Given the significant role of diet in the pathogenesis and prevention of cardiovascular diseases, this study rigorously investigates the causal relationship between dietary habits and myocardial infarction. METHODS AND RESULTS This study used large-scale genome-wide association studies with pooled UK Biobank data to explore associations between 9 dietary categories (83 types) and myocardial infarction. A 2-sample Mendelian randomization approach was applied to assess these associations, while multivariate Mendelian randomization and mediation analyses investigated the role of lipids in mediating the effects of diet on myocardial infarction. Univariate Mendelian analyses revealed genetic associations among 9 categories of dietary habits (83 types) and myocardial infarction. Notably, robust evidence indicates the "tablespoons of cooked vegetables per day" as the most significant risk factor for myocardial infarction development. "Coffee consumption(cups per day)" and "frequency of adding salt to food" were also identified as supplementary risk factors. In contrast, "overall alcohol intake" showed a protective effect, potentially by increasing high-density lipoprotein cholesterol (4.48% mediation) and reducing triglycerides (6.24% mediation). Cereal category, particularly "cereal consumption (bowls per week)" was associated with reduced myocardial infarction risk, contributing by raising high-density lipoprotein cholesterol (3.69% mediation) and lowering total cholesterol (8.33% mediation). Additionally, "overall cheese consumption" was also protective against myocardial infarction. CONCLUSIONS Our findings elucidate the influence of dietary habits on myocardial infarction, showing underlying genetic mechanisms and emphasizing the regulatory role of lipids as an intermediate.
Collapse
Affiliation(s)
- Qian Yang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Man Li
- Department of Geratology Qilu Hospital of Shandong University Jinan Shandong China
| | - Pengcheng Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Naixin Dou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Mei Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Peng Lu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Chunxiao Yu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| |
Collapse
|
48
|
Wang Z, Luo J, Jiang L, Tang C, Chen Y, Yang K, Wang Z, Dong J, Chen X, Yin Z, Li J, Shen W. Sirolimus as a repurposed drug for tendinopathy: A systems biology approach combining computational and experimental methods. Comput Biol Med 2025; 186:109665. [PMID: 39809087 DOI: 10.1016/j.compbiomed.2025.109665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND Effective drugs for tendinopathy are lacking, resulting in significant morbidity and re-tearing rate after operation. Applying systems biology to identify new applications for current pharmaceuticals can decrease the duration, expenses, and likelihood of failure associated with the development of new drugs. METHODS We identify tendinopathy signature genes employing a transcriptomics database encompassing 154 clinical tendon samples. We then proposed a systems biology based drug prediction strategy that encompassed multiplex transcriptional drug prediction, systematic review assessment, deep learning based efficacy prediction and Mendelian randomization (MR). Finally, we evaluated the effects of drug target using gene knockout mice. RESULTS We demonstrate that sirolimus is a repurposable drug for tendinopathy, supported by: 1) Sirolimus achieves top ranking in drug-gene signature-based multiplex transcriptional drug efficacy prediction, 2) Consistent evidence from systematic review substantiates the efficacy of sirolimus in the management of tendinopathy, 3) Genetic prediction indicates that plasma proteins inhibited by mTOR (the target of sirolimus) are associated with increased tendinopathy risk. The effectiveness of sirolimus is further corroborated through in vivo testing utilizing tendon tissue-specific mTOR gene knockout mice. Integrative pathway enrichment analysis suggests that mTOR inhibition can regulate heterotopic ossification-related pathways to ameliorate clinical tendinopathy. CONCLUSIONS Our study assimilates knowledge of system-level responses to identify potential drugs for tendinopathy, and suggests sirolimus as a viable candidate. A systems biology approach could expedite the repurposing of drugs for human diseases that do not have well-defined targets.
Collapse
Affiliation(s)
- Zetao Wang
- Department of Orthopedics, Affiliated Huzhou Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Huzhou, China; Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Junchao Luo
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Luyong Jiang
- Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Chenqi Tang
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China; Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Yangwu Chen
- Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Kun Yang
- Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Zicheng Wang
- Department of Orthopedics, Affiliated Huzhou Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Huzhou, China
| | - Jiabao Dong
- Department of Orthopedics, Affiliated Huzhou Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Huzhou, China
| | - Xiao Chen
- Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Zi Yin
- Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China
| | - Jianyou Li
- Department of Orthopedics, Affiliated Huzhou Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Huzhou, China
| | - Weiliang Shen
- Department of Orthopedics, Affiliated Huzhou Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Huzhou, China; Department of Sports Medicine & Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of sports medicine, Zhejiang University, Hangzhou, China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
49
|
Zheng SL, Jurgens SJ, McGurk KA, Xu X, Grace C, Theotokis PI, Buchan RJ, Francis C, de Marvao A, Curran L, Bai W, Pua CJ, Tang HC, Jorda P, van Slegtenhorst MA, Verhagen JMA, Harper AR, Ormondroyd E, Chin CWL, Pantazis A, Baksi J, Halliday BP, Matthews P, Pinto YM, Walsh R, Amin AS, Wilde AAM, Cook SA, Prasad SK, Barton PJR, O'Regan DP, Lumbers RT, Goel A, Tadros R, Michels M, Watkins H, Bezzina CR, Ware JS. Evaluation of polygenic scores for hypertrophic cardiomyopathy in the general population and across clinical settings. Nat Genet 2025; 57:563-571. [PMID: 39966645 PMCID: PMC11906360 DOI: 10.1038/s41588-025-02094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/21/2025] [Indexed: 02/20/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is an important cause of morbidity and mortality, with pathogenic variants found in about a third of cases. Large-scale genome-wide association studies (GWAS) demonstrate that common genetic variation contributes to HCM risk. Here we derive polygenic scores (PGS) from HCM GWAS and genetically correlated traits and test their performance in the UK Biobank, 100,000 Genomes Project, and clinical cohorts. We show that higher PGS significantly increases the risk of HCM in the general population, particularly among pathogenic variant carriers, where HCM penetrance differs 10-fold between those in the highest and lowest PGS quintiles. Among relatives of HCM probands, PGS stratifies risks of developing HCM and adverse outcomes. Finally, among HCM cases, PGS strongly predicts the risk of adverse outcomes and death. These findings support the broad utility of PGS across clinical settings, enabling tailored screening and surveillance and stratification of risk of adverse outcomes.
Collapse
Affiliation(s)
- Sean L Zheng
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kathryn A McGurk
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Xiao Xu
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Chris Grace
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Pantazis I Theotokis
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Rachel J Buchan
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Catherine Francis
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Antonio de Marvao
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Department of Women and Children's Health, King's College London, London, UK
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Lara Curran
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Wenjia Bai
- Biomedical Image Analysis Group, Department of Computing, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Chee Jian Pua
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore
| | - Hak Chiaw Tang
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Paloma Jorda
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Marjon A van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Judith M A Verhagen
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrew R Harper
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elizabeth Ormondroyd
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Calvin W L Chin
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Antonis Pantazis
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - John Baksi
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Brian P Halliday
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul Matthews
- Department of Brain Sciences, Imperial College London, London, UK
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Roddy Walsh
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Arthur A M Wilde
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - Stuart A Cook
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Department of Cardiology, National Heart Centre, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Sanjay K Prasad
- National Heart Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul J R Barton
- National Heart Lung Institute, Imperial College London, London, UK
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Declan P O'Regan
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - R T Lumbers
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK London, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
| | - Anuj Goel
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Rafik Tadros
- Cardiovascular Genetics Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Michelle Michels
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Paris, France
| | - James S Ware
- National Heart Lung Institute, Imperial College London, London, UK.
- Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK.
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Imperial College Healthcare NHS Trust, London, UK.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
50
|
Tschiderer L, Bakker MK, Gill D, Burgess S, Willeit P, Ruigrok YM, Peters SAE. Sex differences in risk factor relationships with subarachnoid haemorrhage and intracranial aneurysms: A Mendelian randomization study. Eur Stroke J 2025; 10:216-224. [PMID: 39081091 PMCID: PMC7616166 DOI: 10.1177/23969873241265224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/13/2024] [Indexed: 08/10/2024] Open
Abstract
Background The prevalence of intracranial aneurysms (IAs) and incidence of aneurysmal subarachnoid haemorrhage (aSAH) is higher in women than in men. Although several cardiometabolic and lifestyle factors have been related to the risk of IAs or aSAH, it is unclear whether there are sex differences in causal relationships of these risk factors. Aims The aim of this study was to determine sex differences in causal relationships between cardiometabolic and lifestyle factors and risk of aSAH and IA. Methods We conducted a sex-specific two-sample Mendelian randomization study using summary-level data from genome-wide association studies. We analysed low-density lipoprotein cholesterol, high-density lipoprotein cholesterol [HDL-C], triglycerides, non-HDL-C, total cholesterol, fasting glucose, systolic and diastolic blood pressure, smoking initiation, and alcohol use as exposures, and aSAH and IA (i.e. aSAH and unruptured IA combined) as outcomes. Results We found statistically significant sex differences in the relationship between genetically proxied non-HDL-C and aSAH risk, with odds ratios (ORs) of 0.72 (95% confidence interval 0.58, 0.88) in women and 1.01 (0.77, 1.31) in men (p-value for sex difference 0.044). Moreover, genetic liability to smoking initiation was related to a statistically significantly higher risk of aSAH in men compared to women (p-value for sex difference 0.007) with ORs of 3.81 (1.93, 7.52) and 1.12 (0.63, 1.99), respectively, and to a statistically significantly higher IA risk in men compared to women (p-value for sex difference 0.036) with ORs of 3.58 (2.04, 6.27) and 1.61 (0.98, 2.64), respectively. In addition, higher genetically proxied systolic and diastolic blood pressure were related to a higher risk of aSAH and IA in both women and men. Conclusions Higher genetically proxied non-HDL-C was related to a lower risk of aSAH in women compared to men. Moreover, genetic liability to smoking initiation was associated with a higher risk for aSAH and IA in men compared to women. These findings may help improve understanding of sex differences in the development of aSAH and IA.
Collapse
Affiliation(s)
- Lena Tschiderer
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Mark K Bakker
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, Utrecht University, The Netherlands
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Peter Willeit
- Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ynte M Ruigrok
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, Utrecht University, The Netherlands
| | - Sanne AE Peters
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- The George Institute for Global Health, School of Public Health, Imperial College London, London, UK
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|